Biosensors and Biodetection - Methods and Protocols Volume 1 - Optical-Based Detectors PDF
Biosensors and Biodetection - Methods and Protocols Volume 1 - Optical-Based Detectors PDF
Avraham Rasooly
Ben Prickril Editors
Biosensors
and Biodetection
Methods and Protocols
Volume 1: Optical-Based Detectors
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Avraham Rasooly
National Cancer Institute
National Institutes of Health
Rockville, MD, USA
Ben Prickril
National Cancer Institute
National Institutes of Health
Rockville, MD, USA
Editors
Avraham Rasooly Ben Prickril
National Cancer Institute National Cancer Institute
National Institutes of Health National Institutes of Health
Rockville, MD, USA Rockville, MD, USA
Biosensor Technologies
A biosensor is defined by the International Union of Pure and Applied Chemistry (IUPAC)
as “a device that uses specific biochemical reactions mediated by isolated enzymes, immu-
nosystems, tissues, organelles or whole cells to detect chemical compounds usually by
electrical, thermal or optical signals” [1]; all biosensors are based on a two-component
system:
1. Biological recognition element (ligand) that facilitates specific binding or biochemical
reaction with the target analyte
2. Signal conversion unit (transducer)
Since the publication of the first edition of this book in 2009, “classical” biosensor
modalities such as electrochemical or surface plasmon resonance (SPR) continue to be
developed. New biosensing technologies and modalities have also been developed, includ-
ing the use of nanomaterials for biosensors, fiber-optic-based biosensors, genetic code based
sensors, field effect transistors, and the use of mobile communication device-based biosen-
sors. Although it is impossible to describe the fast-moving field of biosensing in a single
publication, this book presents descriptions of methods and uses for some of the basic types
of biosensors while also providing the reader a sense of the enormous importance and
potential for these devices. In order to present a more comprehensive overview, the book
also describes other biodetection technologies.
Dr. Leland C. Clark, who worked on biosensors in the early 1960s, provided an early
reference to the concept of a biosensor by developing an “enzyme electrode” for glucose
concentration measurement using the enzyme glucose oxidase (GOD) [2]. Glucose moni-
toring is essential for diabetes patients, and even today the most common clinical biosensor
technology for glucose analysis is the electrochemical detection method envisioned by Clark
more than 50 years ago. Today, glucose monitoring is performed using rapid point of care
biosensors made possible through advances in electronics that have enabled sensor minia-
turization. The newest generation of biosensors includes phone-based optical detectors with
high-throughput capabilities.
vii
viii Preface
Biosensor Classification
In general biosensors can be divided into two groups: direct recognition sensors in which
the biological interaction is directly measured and indirect detection sensors which rely on
secondary elements (often catalytic) such as enzymes or fluorescent tags for measurements.
Figure 1 illustrates the two types of biosensors. In each group there are several types of
A B
Transducer
Output Interface
Fig. 1 General schematic of biosensors. (A) Direct detection biosensors where the recognition element is
label-free; (B) indirect detection biosensors using “sandwich” assay where the analyte is detected by labeled
molecule. Direct detection biosensors are simpler and faster but typically yield a higher limit of detection
compared to indirect detection systems
Preface ix
Organization
The publication is divided into two volumes. Volume I (Springer Vol. 1571) focuses on
optical-based detectors, while Vol. II (Springer Vol. 1572) focuses on electrochemical,
bioelectronic, piezoelectric, cellular, and molecular biosensors.
xii Preface
References
1. International Union of Pure and Applied Chemistry (1992) IUPAC compendium of chemical
terminology, 2nd edn (1997). International Union of Pure and Applied Chemistry, Research Triangle
Park, NC
2. Clark LC Jr, Lyons C (1962) Electrode systems for continuous monitoring in cardiovascular surgery.
Ann N Y Acad Sci 102:29–45
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xix
1 Localized Surface Plasmon Resonance (LSPR)-Coupled Fiber-Optic
Nanoprobe for the Detection of Protein Biomarkers . . . . . . . . . . . . . . . . . . . . . . . . 1
Jianjun Wei, Zheng Zeng, and Yongbin Lin
2 Ultra-Sensitive Surface Plasmon Resonance Detection by Colocalized
3D Plasmonic Nanogap Arrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Wonju Lee, Taehwang Son, Changhun Lee, Yongjin Oh,
and Donghyun Kim
3 Two-Dimensional Surface Plasmon Resonance Imaging System
for Cellular Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Tanveer Ahmad Mir and Hiroaki Shinohara
4 Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings. . . . . . . . . 47
Christophe Caucheteur, Clotilde Ribaut, Viera Malachovska,
and Ruddy Wattiez
5 Biosensing Based on Magneto-Optical Surface Plasmon Resonance . . . . . . . . . . . 73
Sorin David, Cristina Polonschii, Mihaela Gheorghiu,
Dumitru Bratu, and Eugen Gheorghiu
6 Nanoplasmonic Biosensor Using Localized Surface Plasmon
Resonance Spectroscopy for Biochemical Detection . . . . . . . . . . . . . . . . . . . . . . . . . 89
Diming Zhang, Qian Zhang, Yanli Lu, Yao Yao,
Shuang Li, and Qingjun Liu
7 Plasmonics-Based Detection of Virus Using Sialic Acid Functionalized
Gold Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
Changwon Lee, Peng Wang, Marsha A. Gaston, Alison A. Weiss,
and Peng Zhang
8 MicroRNA Biosensing with Two-Dimensional Surface
Plasmon Resonance Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Ho Pui Ho, Fong Chuen Loo, Shu Yuen Wu, Dayong Gu,
Ken-Tye Yong, and Siu Kai Kong
9 Gold Nanorod Array Biochip for Label-Free, Multiplexed
Biological Detection. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Zhong Mei, Yanyan Wang, and Liang Tang
10 Resonant Waveguide Grating Imager for Single Cell Monitoring
of the Invasion of 3D Speheroid Cancer Cells Through Matrigel . . . . . . . . . . . . . 143
Nicole K. Febles, Siddarth Chandrasekaran, and Ye Fang
xv
xvi Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
Contributors
xix
xx Contributors
YA-XIAN YAN Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology,
Shanghai Jiao Tong University, Shanghai, China
LI YANG Natural Products Research Center, Chengdu Institution of Biology, Chinese
Academy of Science, Chengdu, China
YAO YAO Biosensor National Special Laboratory, Key Laboratory for Biomedical
Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang
University, Hangzhou, China
AFSHAN YASMEEN Natural Products Research Center, Chengdu Institution of Biology,
Chinese Academy of Science, Chengdu, China
KEN-TYE YONG Department of Electronic Engineering, Center for Advanced Research in
Photonics, The Chinese University of Hong Kong, N.T. Hong Kong SAR, China
ZHENG ZENG Department of Nanoscience, Joint School of Nanoscience and
Nanoengineering (JSNN), University of North Carolina at Greensboro, Greensboro,
NC, USA
ROLAND ZENGERLE Hahn-Schickard, Freiburg, Germany; Laboratory for MEMS
Applications, Department of Microsystems Engineering—IMTEK, University of Freiburg,
Freiburg, Germany
QIAN ZHANG Biosensor National Special Laboratory, Key Laboratory for Biomedical
Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang
University, Hangzhou, China
PENG ZHANG Department of Chemistry, University of Cincinnati, Cincinnati, OH, USA
DIMING ZHANG Biosensor National Special Laboratory, Key Laboratory for Biomedical
Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang
University, Hangzhou, China
Chapter 1
Abstract
Here is presented a miniaturized, fiber-optic (FO) nanoprobe biosensor based on the localized surface
plasmon resonance (LSPR) at the reusable dielectric-metallic hybrid interface with a robust, gold nano-disk
array at the fiber end facet. The nanodisk array is directly fabricated using electron beam lithography (EBL)
and metal lift-off process. The free prostate-specific antigen (f-PSA) has been detected with a mouse anti-
human prostate-specific antigen (PSA) monoclonal antibody (mAb) as a specific receptor linked with a self-
assembled monolayer (SAM) at the LSPR-FO facet surfaces. Experimental investigation and data analysis
found near field refractive index (RI) sensitivity at ~226 nm/RIU with the LSPR-FO nanoprobe, and
demonstrated the lowest limit of detection (LOD) at 100 fg/mL (~3 fM) of f-PSA in PBS solutions. The
SAM shows insignificant nonspecific binding to the target biomarkers in the solution. The control
experimentation using 5 mg/mL bovine serum albumin in PBS and nonspecific surface test shows the
excellent specificity and selectivity in the detection of f-PSA in PBS. These results indicate important
progress toward a miniaturized, multifunctional fiber-optic technology that integrates informational com-
munication and sensing function for developing a high-performance, label-free, point-of-care (POC)
device.
Key words Fiber optics, Protein biomarker biosensors, Nanofabrication, Au nanodisk array, Localized
surface plasmon resonance (LSPR), Signal transduction
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_1,
© Springer Science+Business Media LLC 2017
1
2 Jianjun Wei et al.
Fig. 2 Schematic diagrams illustrating (a) a localized surface plasmon [18], (b) a
configuration of representative experimental setup and procedure for LSPR
sensing [19]
Au Nanodisk
140
Reflectance Intensity
120
100 After
White Binding
Cladding
80
Light Antigen 60
40
Optical Fiber Tip 20
Core 0
500 600 700 800 900
LSPR Reflectance (nm)
LSPR Reflectance
Fig. 3 A diagram illustrating the principle of LSPR coupling on fiber optic probe for biosensing. The nanodisk
arrays are fabricated on optical fiber tip end. The LSPR reflectance is recorded with a white incident light
guided in the fiber
Localized Surface Plasmon Resonance (LSPR)-Coupled Fiber-Optic. . . 5
7. The fiber clamp for hoisting fiber for vibration was obtained
from Newport Corporation, CA, USA.
8. Ultra clean convection oven (Ultra-clean 100) was obtained
from Thermo Fisher Scientific, OH, USA.
9. Nano pattern generation system (NPGS) was obtained from JC
Nabity Lithography Systems, MT, USA.
10. Field emission scanning electron microscope (FESEM) was a
LEO 1550 model.
11. Three cathode vacuum sputter system (Denton Discovery-18
Sputter System) was obtained from Denton Vacuum LLC, NJ,
USA.
12. Vacuum thermal evaporate system was donated to University
of Alabama in Huntsville by US Army.
13. Reactive Ion Etching (RIE) system (Plasma-Therm 790) was
obtained from Plasma-Therm, FL, USA.
14. Optical fiber coupled Tungsten Halogen light source (LS-1)
was obtained from Ocean Optics, FL, USA.
15. Mini-spectrometer (USB2000-VIS-NIR) was obtained from
Ocean Optics, FL, USA.
16. 2 2 Single-mode fiber optic couplers for 633 nm wavelength
were obtained from Newport Corporation, CA, USA.
3 Methods
Fig. 4 A schematic flow-chart displays the fabrication procedure for nanodisk arrays at the optical fiber end
facet. (a) optical fiber tip, (b) 2 nm Cr deposition, (c) resist deposition, (d) EBL process, (e) gold film deposition,
(f) lift-off process, (g) Cr etch to get the nanodisk arrays
Localized Surface Plasmon Resonance (LSPR)-Coupled Fiber-Optic. . . 7
Fig. 5 Illustrations of the procedure called “dip and vibration” technique, (a) dip in the diluted e-beam resist,
(b) after dip, (c) vibration, and (d) after bake
8 Jianjun Wei et al.
The fiber tip is then vibrated manually by pulling the fiber tip to
one side and then releasing to get rid of excessive resist by
means of cantilever beam free vibration. The vibration fre-
quency and strength is controlled by the length of fiber tip
outside the fiber clamp and the initial displacement of the fiber
tip. The thickness of resist on the optical fiber tip is dependent
on the ZEP dilution ratio and vibration strength. The iterative
method is used to optimize the vibration process. The initial
fiber tip displacement for the vibration is 2–5 mm, and the fiber
tip length is 15–25 mm. The ZEP dilution ratio used is from
20% to 40%, diluted in ZEP thinner. The resulted e-beam resist
layer thickness on the fiber tip is 100–200 nm, measured by
SEM observation. The fiber tip is held vertically upward and
baked in a 120 C oven for 60 min (Fig. 5d).
6. The EBL process based on the Nano Pattern Generation Sys-
tem (NPGS) and a field emission scanning electron microscope
(FESEM) is used to create nanodot array pattern on the e-beam
resist on the fiber end face (Fig. 4d). A Newport fiber clamp is
used to hold the fiber vertically on the translation stage in the
SEM chamber. The EBL voltage is 30 kV and the exposure
dose is 70 μC/cm2. The fiber tip is developed by dipping in
resist developer (ZEP N50) for 1 min. The fiber tip is then
rinsed in DI water and baked in the 120 C oven for 10 min for
dehydration.
7. An oxygen plasma descum procedure in a reactive ion etcher
(RIE, 25 watts power for 3 min) is used to remove the thin
residual layers of photoresist following the photoresist devel-
opment step (see Note 1).
8. The deposition of 40 nm Au overlay over the patterned area is
carried out by using the standard thermal evaporation coating
technique (Fig. 4e). The 2 nm Cr layer previously deposited as
a conductive layer for the EBL process is now served as an
adhesive layer for Au overlay. To lift off the e-beam resist, the
fiber tip is dipped in the ZEP remover for 10 min, followed by a
1-min ultrasonic bath to assist the lift-off process (Fig. 4f). The
fiber tip is rinsed in DI water and checked under an optical
microscope to make sure that the resist layer on the fiber end
face has been removed (see Note 2).
9. The fiber tip is dipped into the Cr remover solution for 30 s to
remove the Cr layer that is not covered by the Au nanoparticles
(Fig. 4g). The fiber tip is rinsed again in DI water and baked in
the 120 C oven for 10 min for dehydration. Figure 6 shows
the scanning electron micrographs of a gold nanodisks array on
an optical fiber tip end facet.
10. Finally, the Au nanodisk array on fiber end face is annealed at
530 C for 5 min and ready for next usage (see Note 3).
Localized Surface Plasmon Resonance (LSPR)-Coupled Fiber-Optic. . . 9
Fig. 6 Images of (a) overview of the optical fiber tip end, (b) SEM images of gold nanodisk arrays on the optical
fiber facet after the lift-off process
Fig. 7 A schematic diagram illustrating optical setup for the fiber tip LSPR sensor based on reflection spectrum
measurement
3.2 Optical 1. In this optical setup (Fig. 7), a 2 1 single-mode optical fiber
Measurement coupler for 633 nm wavelength is used, which has three light
connection ports. In Fig. 7, port (a) and port (b) are two
connections on one side of optical fiber coupler, and port (c)
is connection on the opposite side. Port (a) is connected to the
tungsten halogen white light source; port (b) is connected to a
mini-spectrometer; and port (c) is connected to the LSPR-FO
probe using a fusion splicer. The white light (450–950 nm) is
launched from port (a), propagated to port (c), and reflected
from the end facet with the Au nanodisks arrays. The reflection
light propagated to port (b), where the spectrum of the
reflection light is measured by the mini-spectrometer, which
is connected to a computer for data acquisition and processing.
Figure 8 shows the photograph of the Optical setup for
the fiber tip LSPR sensor based on reflection spectrum
measurement.
2. During fiber optic LSPR detection, the light is launched and
guided to the LSPR fiber tip facet. The light wave propagates
along the core in the center of the optical fiber tip where the
10 Jianjun Wei et al.
Fig. 8 Photograph of the optical components (optical fiber, fiber coupler, mini-
spectrometer, light source) and setup for the fiber tip LSPR sensor. Computer
connection to the mini spectrometer via USB cable is not shown
3.3 LSPR-FO 1. The nanoprobe sensitivity is used to determine if the sensor tip
Nanoprobe Sensitivity in experimentation would be sensitive enough to determine
Characterization small wavelength shifts accurately.
2. The solvents used in determining the bulk RI sensitivity of the
LSPR tip are acetone, methanol, ethanol, isopropyl alcohol,
and water of different RIs.
3. The LSPR-FO nanoprobe is dipped in the various solvents, and
the spectra of the reflected light were recorded, respectively, as
shown in Fig. 9a (see Note 4).
4. To determine the peak wavelength in the LSPR reflection
spectrum, a Matlab program was created to fit the data to an
eighth-order polynomial function over a range of 80 nm [17],
centered at the wavelength of maximum reflection in the raw
data.
5. The calculated LSPR wavelength red shifted as the RI of the
solvent increased. A linear relationship between the LSPR peak
wavelength and the bulk RI of the medium is obtained (Fig. 9b
black dots), with the sensitivity of 226 nm/RIU (RIU, refrac-
tive index unit) used in this study.
6. The light intensity-based RI sensitivity is investigated as well
(Fig. 9b red diamonds). The return light intensities at the
LSPR peak wavelength are plotted against the bulk RI of the
media. The LSPR peak intensity is seen to be linearly propor-
tional to the RI, and the gradient of the line is 123 per RIU.
The return light intensity of a single-wavelength laser can be
used as a probing light for the refractive index change due to
Fig. 9 Sensitivity measurements of the LSPR-FO nanoprobe. (a) Measured reflectance spectra for the LSPR-FO
probe in various solvents of different refractive index. (b) Correlation of the LSPR peak wavelengths (left axis)
and LSPR peak intensity (right axis) with the refractive index (RI) of the solvents, the linear fit of the changes
vs. the RI gives the sensing sensitivity. In the inset equation, x and y represent the values of x and y axis. R2 is
the coefficient of determination in the linear fitting
12 Jianjun Wei et al.
Fig. 10 Detection of protein biomarker. (a) The representative reflectance spectra of the anti-PSA mAb
modified LSPR-FO nanoprobe in various concentrations of f-PSA (from 0 up to 0.5 ng/mL). Insert plot shows
spectra normalized to peak intensity to show the LSPR reflectance peak shifts. (b) Correlation of f-PSA
concentration to the reflectance light of resonance peak wavelength shift
4 Notes
1. The optional step after the oxygen descum is to dip the fiber tip
in the chrome remover solution for 60 s to remove the thin Cr
layer on the nanodisk area. This step will eliminate the Cr
adhesion layer under the Au nanodisk, avoiding the undesired
side effects of Cr layer under Au nanodisk in the LSPR sensing
process, such as spectrum broadening and dephasing. How-
ever, this step will weaken the adhesion strength between the
Au nanodisks and the fiber end glass substrate, and will reduce
the reusability of the fiber LSPR probe.
2. If the lift-off process is not complete, ZEP remover and ultra-
sonic bath may be used to further lift off the e-beam resist.
3. The fiber probe is inserted horizontally into the oven through
an access hole on the sidewall of a high-temperature oven. In
order to protect the polymer buffer on the fiber, only
10–15 mm of fiber probe needs to be inside the oven.
4. Note that there is a noisy region in all spectra from ~760 to
800 nm, which is caused by the transmission minimum for the
optical fiber coupler (single mode at 633 nm) in the same
wavelength range.
14 Jianjun Wei et al.
Reference
1. Mukundan H et al (2009) Optimizing a wave- 9. Leskova TA, Maradudin AA, Zierau W (2005)
guide-based sandwich immunoassay for tumor Surface plasmon polariton propagation near an
biomarkers: evaluating fluorescent labels and index step. Optics Commun 249(1–3):23–35
functional surfaces. Bioconjug Chem 20 10. Jung LS et al (1998) Quantitative interpreta-
(2):222–230 tion of the response of surface plasmon reso-
2. Liu X et al (2008) A one-step homogeneous nance sensors to adsorbed films. Langmuir 14
immunoassay for cancer biomarker detection (19):5636–5648
using gold nanoparticle probes coupled with 11. Mitsui K, Handa Y, Kajikawa K (2004) Optical
dynamic light scattering. J Am Chem Soc 130 fiber affinity biosensor based on localized sur-
(9):2780–2782 face plasmon resonance. Appl Phys Lett 85
3. Sekhar PK, Ramgir NS, Bhansali S (2008) (18):4231–4233
Metal-decorated silica nanowires: an active sur- 12. Chang T-C et al (2012) Using a fiber optic
face-enhanced raman substrate for cancer bio- particle plasmon resonance biosensor to deter-
marker detection. J Phys Chem C 112 mine kinetic constants of antigen–antibody
(6):1729–1734 binding reaction. Anal Chem 85(1):245–250
4. Mani V et al (2009) Ultrasensitive immunosen- 13. 1985. http://apps.who.int/iris/bitstream/
sor for cancer biomarker proteins using gold 10665/38405/1/WHO_TRS_725.pdf.
nanoparticle film electrodes and multienzyme- 14. Nishi Y, Doering R (2000) Handbook of semi-
particle amplification. ACS Nano 3 conductor manufacturing technology. CRC
(3):585–594 Press, Boca Raton, FL
5. Henne WA et al (2006) Detection of folate 15. May GS, Spanos CJ (2006) Fundamentals of
binding protein with enhanced sensitivity semiconductor manufacturing and process
using a functionalized quartz crystal microbal- control. John Wiley & Sons, New York, NY
ance sensor. Anal Chem 78(14):4880–4884
16. Obando LA, Booksh KS (1999) Tuning
6. Baker GA, Desikan R, Thundat T (2008) dynamic range and sensitivity of white-light,
Label-free sugar detection using phenylboro- multimode, fiber-optic surface plasmon reso-
nic acid-functionalized piezoresistive micro- nance sensors. Anal Chem 71(22):5116–5122
cantilevers. Anal Chem 80(13):4860–4865
17. Wu H-J et al (2012) Membrane-protein binding
7. Lee HJ, Nedelkov D, Corn RM (2006) Surface measured with solution-phase plasmonic nano-
plasmon resonance imaging measurements of cube sensors. Nat Methods 9(12):1189–1191
antibody arrays for the multiplexed detection
of low molecular weight protein biomarkers. 18. Willets KA, Van Duyne RP (2007) Localized
Anal Chem 78(18):6504–6510 surface plasmon resonance spectroscopy and
sensing. Annu Rev Phys Chem 58(1):267–297
8. Raether H (1988) Surface plasmons on
smooth and rough surfaces and on gratings. 19. Zhao J et al (2006) Localized surface plasmon
Springer tracts in modern physics. Springer, resonance biosensors. Nanomedicine 1
Berlin (2):219–228
Chapter 2
Abstract
Ultra-sensitive detection based on surface plasmon resonance (SPR) was investigated using 3D nanogap
arrays for colocalization of target molecular distribution and localized plasmon wave in the near-field.
Colocalization was performed by oblique deposition of a dielectric mask layer to create nanogap at the side
of circular and triangular nanoaperture, where fields localized by surface plasmon localization coincide with
the spatial distribution of target molecular interactions. The feasibility of ultra-sensitivity was experimen-
tally verified by measuring DNA hybridization. Triangular nanopattern provided an optimum to achieve
highly amplified angular shifts and led to enhanced detection sensitivity on the order of 1 fg/mm2 in terms
of molecular binding capacity. We confirmed improvement of SPR sensitivity by three orders of magnitude,
compared with conventional SPR sensors, using 3D plasmonic nanogap arrays.
Key words Surface plasmon resonance, Localized surface plasmon resonance, Surface plasmon reso-
nance detection, Nanogap arrays, Nanoapertures, Colocalization, DNA hybridization
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_2,
© Springer Science+Business Media LLC 2017
15
16 Wonju Lee et al.
2 Materials
Fig. 2 (a) Schematics of optical setup for a SPR sensor based on colocalization
using plasmonic nanogap arrays and (b) photograph of the experimental setup
2.2 Optical Setup A schematic illustration of the optical setup for colocalized SPR
detection is presented in Fig. 2 (see Note 1).
1. He-Ne laser (36 mW, λ ¼ 632.8 nm, Nominal beam diameter:
650 μm; Melles-Griot, Carlsbad, CA).
2. Glan-Thompson Linear Polarizer (Thorlabs, Inc., Newton, NJ).
3. Chopper and controller (SR540; Stanford Research Systems,
Sunnyvale, CA).
4. Two concentric motorized rotation stages (URS75PP and
ESP330; Newport, Irvine, CA) (see Note 2).
5. Low-noise lock-in amplifier (Model SR830; Stanford Research
Systems, Sunnyvale, CA).
6. A p-i-n photodiode (818-UV; Newport, Irvine, CA).
7. Lab ViewLabVIEW (National Instruments, Austin, TX).
8. Index-matching gel (n ¼ 1.725; Cargille Laboratories, Cedar
Grove, NJ).
Ultra-Sensitive Surface Plasmon Resonance Detection. . . 19
2.3 DNA Preparation 1. HPLC purified 24-mer sequence length capture probe and
target oligonucleotides (IDT, Coralville, IA).
2. The sequence of single-stranded probe DNA (p-DNA) was 50 -
TTT TTT CGG TAT GCA TGC CAT GGC-3 modified with
thiol at 50 .
3. The sequence of single-stranded target DNA (t-DNA) was 50 -
GCC ATG GCA TGC ATA CCG AAA AAA-30 .
4. Plasma cleaner (Harrick Scientific Products, Pleasantville, NY).
5. Micropump (KD Scientific, Holliston, MA).
6. Acetone (1009-4404; DAEJUNG CHEMICAL & METALS
CO., Siheung, Korea).
7. Phosphate Buffered Saline (PBS) buffer (pH ¼ 7.4, BP3991;
Fisher Scientific, Pittsburgh, PA).
3 Methods
Fig. 3 (a) SEM image of fabricated linear nanograting arrays and (b) magnified image of 2D nanogap arrays
20 Wonju Lee et al.
3.2 Optical Setup A He-Ne laser beam (36 mW, λ ¼ 632.8 nm, nominal beam
diameter: 650 μm, Melles-Griot, Carlsbad, CA) is TM-polarized
by linear polarizer and temporally modulated by optical chopper
(SR540; Stanford Research Systems, Sunnyvale, CA), which alters
the state of light on and off at a specific frequency. The chopping
frequency is used as the reference input of a lock-in amplifier
composed with usually set to 0.6 kHz lock-in amplifier and feed-
back system. Among the signals captured at a photodiode, noise
signal at frequency other than the chopping can be removed. Two
motorized rotation stages (URS75PP; Newport, Irvine, CA) are
employed; one is for rotating a prism on which a plasmonic nanos-
tructured sample is located, and the other is for rotating the pho-
todiode to keep laser alignment to photodiode. The angle of the
stage on which the nanostructure sample located can be corrected
by using the calibration constant and the other stage with the
Ultra-Sensitive Surface Plasmon Resonance Detection. . . 21
3.3 Estimation of A 2D nanogap area GΛ,2D produced by linear nanograting per unit
Colocalization Areas grating length with the thickness dg can be calculated as
3.3.1 2D Nanogap G Λ, 2D ¼ d g ð1 þ tan θeva Þ ð1Þ
Production
where θeva is the oblique evaporation angle (see Fig. 1b) in
Eq. 1, the first term dg and the second term dgtan θeva come from
vertical nanograting edge and horizontal surface which are not
evaporated with dielectric, respectively. For example, when the
nanograting with dg ¼ 20 nm and θeva ¼ 30 produces a nanogap
with an area of 31.56 nm times the grating length.
3.3.2 Nanogap Reduction 3D nanoaperture arrays can reduce the colocalization area, in which
of the Colocalization Area target molecular binding overlaps with localized fields to amplify
optical signatures of the target interaction. A 3D nanogap area
defined by an equi-triangular aperture with a side length (L) is
calculated as
d g tan θeva
G Λ, 3D ¼ Ld g ð1 þ tan θeva Þ þ pffiffiffi ð2 tan θeva Þ ð2Þ
3L
where the direction of oblique evaporation is parallel to a side
of a triangular pattern. The first term Ldg represents the area open
to the side, while the second term Ldg tan θeva is the area formed at
nanogap surface. The last term in Eq. 2 is the correction due to the
triangular shape. If L is much longer than dg, the above equation
can be simplified as
πϕ 1
G Λ, 3D d g 1 þ tan θeva : ð4Þ
2 2
Using 3D nanogap arrays, the number of molecular interac-
tions can be adjusted by changing the size to period ratio L/Λ.
Fig. 5 Numerically calculated near-field intensity distribution |E |2 of (a) circular and (b) triangular nanopatterns
with ϕ, L ¼ 600 nm at Λ ¼ 2 μm. A 5-nm-thick ITO layer was assumed to be deposited on the nanopattern
with θeva ¼ 30
Ultra-Sensitive Surface Plasmon Resonance Detection. . . 23
Fig. 6 SEM images: (a) circular and (b) triangular nanopattern arrays. (c) A magnified image of a triangular
nanogap pattern. A very narrow nanogap is clearly visible at the side ridge of a triangular nanoaperture.
Each inset of (a) and (b) shows the nanogap created on a circular and triangular aperture (scale bars: 2 μm for
(a) and (b), 200 nm for (c), insets of (a) and (b))
3.5 Experimental (c) 1 ml Phosphate buffered saline (PBS) buffer (pH ¼ 7.4)
Measurements of DNA including 4 μM thiol hexane labeled probe ssDNA
Hybridization (HSssDNA) was injected to flow channel by a sylinge pump.
The sample chip was immersed in the buffer at 4 C for 4 h to
3.5.1 DNA Preparation immobilize the p-DNAs at surface (see Note 5). In order to
obtain SPR angle shift, the reference SPR angle was measured
and smoothed by 50th order Fourier filter.
(d) For DNA hybridization, PBS buffer including 4 μM t-DNAs
was injected on the p-DNA-immobilized sample chip surface
for 120 min by a fluidic channel of a micropump (KD Scien-
tific, Holliston, MA) at a volume flow rate of 150 μL/hr. The
sample chip was incubated to improve the efficiency of hybri-
dization through mild heating. For 2 h, the solution of t-
DNAs is warmed to 65 C and slowly cooled down to room
temperature. Finally, the SPR angle shift was obtained by
comparing SPR angles before and after the DNA
hybridization.
SPR shifts were measured using linear nanogratings and 3D
nanopattern arrays to analyze the sensor performance, as shown in
Figs. 7 and 8 for DNA hybridization. Two metrics were set to
evaluate the sensor performance: (1) sensitivity enhancement fac-
tor, i.e., resonance shift with respect to the shift obtained from
conventional SPR detection, and (2) molecular binding capacity
measured by colocalization in the nanogap.
3.5.2 Sensitivity First, resonance angle shifts were measured based on colocalization
Enhancement using plasmonic nanogap arrays, such as nanogratings, circular and
triangular nanopatterns, compared to non-colocalized SPR detec-
tion without nanogap structures. In the non-colocalized detection,
DNA hybridization occurs on the overall metal surface of nanos-
tructures. This leads to inefficient detection of target molecules,
majority of which may be distributed in the region with weak near-
fields.
Figure 7a shows raw data of measured SPR angle shifts θSPR in
various platforms. In case of non-colocalized detection, resonance
angle shifts made on nanostructures were detected to be slightly
larger than that of the thin film control. The larger resonance shift
is associated with the increase of the total area available for DNA
hybridization due to the presence of nanopatterns. For colocalized
detection, SPR angle shift tends to be larger with an increase of the
ITO-evaporation angle (θeva). A larger θeva enlarges the nanogap
surface area, which allows more probe-DNAs (p-DNAs) to immo-
bilize at the nanogap surface. Increased resonance shifts were
observed for colocalization by nanogratings because the nanogap
area by grating patterns is much larger than that of 3D nanogap
patterns and therefore induces more p-DNA targets to participate
in hybridization.
Ultra-Sensitive Surface Plasmon Resonance Detection. . . 25
Fig. 7 (a) Measured SPR angle shifts as the evaporation angle (θeva) varies. (b)
Relative angle shifts (RAS) after normalization by the nanogap area. Reproduced
from Y. Oh et al. 2014 with permission from Elsevier
Fig. 8 (a) Angular SPR shifts (ΔθSPR) measured as the array period (Λ) is varied.
3D nanogap arrays were created by triangular nanopatterns at θeva ¼ 60 and
the concentration of t-DNA was fixed at 4 μM. Inset shows a linear relation
between ΔθSPR and log(Nnp). (b) Angular shifts measured as the concentration of
t-DNAs (Ct-DNA) varied with the fixed p-DNA concentration at 4 μM. Inset also
shows a linear relation between ΔθSPR and log(Ct-DNA). Reproduced from Y. Oh et
al. 2014 with permission from Elsevier
3.5.3 Molecular Binding To evaluate molecular binding capacity as the sensor performance,
Capacity Fig. 8 shows the isotherm characteristics measured in two ways
using the optimum triangular nanogap arrays in 3D at θeva ¼ 60 ,
which was determined to produce the maximum RAS as shown in
Fig. 7b. First, the array period was increased when the concentra-
tion of target DNA was fixed as 4 μM, which in effect decreases the
concentration of probe DNA. Secondly, the isotherm characteritic
was also measured by varying the concentration when the array
period and the concentration of probe DNA were fixed at 2 μm and
4 μM respectively.
To extract the detection sensitivity of colocalized SPR sensing
based on 3D nanogaps, resonance angle shifts (ΔθSPR) were
measured by varying the array period as shown in Fig. 8a, where
the amount of probe DNA is gradually reduced with an increase of
array period while the amount of target DNA remains constant.
The relation between ΔθSPR and the number of nanopatterns (Nnp)
was presented as
ΔθSPR ¼ 0:015 þ 0:030logN np : ð5Þ
In Eq. 5, ΔθSPR is proportional to log(Nnp), which means SPR
shifts are linearly increased when the amount of p-DNA is expo-
nentially increased. As Nnp is reduced, the detection and the nature
of resonance angle shifts change from being target-limited to
probe-limited with transition taking place at Nnp ¼ 3300~13,000
(Λ ¼ 5–10 μm), i.e., the limit of detection arises in the probe-
limited detection of DNA hybridization. If we assume surface
probe density to be 4 1012 molecules/cm2 and hybridization
efficiency 40%, a total effective nanogap area of 264,000 nm2 in a
650-μm-diameter beam spot measures approximately 4200 mole-
cules. This corresponds to the molecular binding capacity of
1.6 fg/mm2 based on the molecular weight of 7.4 kDa for a 24-
mer t-DNA. The results suggest that the detection sensitivity of 3D
nanogap-based SPR sensing, compared to that of conventional SPR
sensors, should be improved by more than three orders of
magnitude.
Figure 8b shows the measured angular shifts when the concen-
tration of target DNAs (Ct-DNA) was varied. In this case, p-DNA
concentration was fixed at 4 μM and other conditions remained the
same as in Fig. 8a. Surprisingly, the relation between ΔθSPR and Ct-
DNA is similar to what was measured from ΔθSPR and Nnp, which
implies that the amount of DNA hybridization is limited by
28 Wonju Lee et al.
4 Notes
References
1. Kim D (2005) Effect of the azimuthal orienta- 3. Dasary SSR, Singh AK, Senapati D, Yu H, Ray
tion on the performance of grating-coupled PC (2009) Gold nanoparticle based label-free
surface-plasmon resonance biosensors. Appl SERS probe for ultrasensitive and selective
Optics 44:3218–3223 detection of trinitrotoluene. J Am Chem Soc
2. Campbell CT, Kim G (2007) SPR microscopy 131:13806–13812
and its application to high-throughput analyses 4. Vollmer F, Arnold S (2008) Whispering-gal-
of biomolecular binding events and their kinet- lery-mode biosensing: label-free detection
ics. Biomaterials 28:2380–2392 down to single molecules. Nat Methods
5:591–596
Ultra-Sensitive Surface Plasmon Resonance Detection. . . 29
5. Oh Y, Lee W, Kim Y, Kim D (2014) Self- localized surface plasmon resonance biosen-
aligned colocalization of 3D plasmonic nano- sing. Opt Express 22:28412–28426
gap arrays for ultra-sensitive surface plasmon 14. Ryu Y, Moon S, Oh Y, Kim Y, Lee T, Kim DH,
resonance detection. Biosens Bioelectron Kim D (2014) Effect of coupled graphene
51:401–407 oxide on the sensitivity of surface plasmon res-
6. Byun KM, Kim S, Kim D (2005) Design study onance detection. Appl Optics 53:1419–1426
of highly sensitive nanowire-enhanced surface 15. Chung K, Rani A, Lee J-E, Kim JE, Kim Y,
plasmon resonance biosensors using rigorous Yang H, Kim SO, Kim D, Kim DH (2015) A
coupled wave analysis. Opt Express systematic study on the sensitivity enhance-
13:3737–3742 ment in graphene plasmonics sensors based
7. Kim K, Yoon SJ, Kim D (2006) Nanowire- on layer-by-layer self-assembled graphene
based enhancement of localized surface plas- oxide multilayers and their reduced anologues.
mon resonance for highly sensitive detection: ACS Appl Mater Interfaces 7:144–151
a theoretical study. Opt Express 16. Kubatkin S, Danilov A, Hjort M, Cornil J,
14:12419–12431 Brédas J-L, Stuhr-Hansen N, Hedegård P,
8. Moon S, Kim Y, Oh Y, Lee H, Kim HC, Lee K, Bjørnholm T (2003) Single-electron transistor
Kim D (2012) Grating-based surface plasmon of a single organic molecule with access to
resonance detection of core-shell nanoparticle several redox states. Nature 425:698–701
mediated DNA hybridization. Biosens Bioelec- 17. Elhadj S, Singh G, Saraf RF (2004) Optical
tron 32:141–147 properties of an immobilized DNA monolayer
9. Yu H, Kim K, Ma K, Lee W, Choi JW, Yun CO, from 255 to 700 nm. Langmuir
Kim D (2013) Enhanced detection of virus 20:5539–5543
particles by nanoisland-based localized surface 18. Fromm DP, Sundaramurthy A, Schuck PJ,
plasmon resonance. Biosens Bioelectron Kino G, Moerner WE (2004) Gap-dependent
41:249–255 optical coupling of single “bowtie” nanoanten-
10. Hoa XD, Kirk AG, Tabrizian M (2009) nas resonant in the visible. Nano Lett
Enhanced SPR response from patterned immo- 4:957–961
bilization of surface bioreceptors on nano-grat- 19. Haes AJ, Zou S, Schatz GC, Van Duyne RP
ings. Biosens Bioelectron 24:3043–3048 (2004) Nanoscale optical biosensor: short
11. Ma K, Kim DJ, Kim K, Moon S, Kim D (2010) range distance dependence of the localized sur-
Target-localized nanograting-based surface face plasmon resonance of noble metal nano-
plasmon resonance detection toward label-free particles. J Phys Chem B 108:6961–6968
molecular biosensing. IEEE J Sel Topics Quan- 20. Szunerits S, Castel X, Boukherroub R (2008)
tum Electron 16:1004–1014 Surface plasmon resonance investigation of sil-
12. Kim Y, Chung K, Lee W, Kim DH, Kim D ver and gold rilms coated with thin indium tin
(2012) Nanogap-based dielectric-specific colo- oxide layers: Influence on stability and sensitiv-
calization for highly sensitive surface plasmon ity. J Phys Chem C 112:15813–15817
resonance detection of biotin-streptavidin 21. Steel AB, Herne TM, Tarlov MJ (1998) Elec-
interaction. Appl Phys Lett 101:233701 trochemical quantitation of DNA immobilized
13. Yang H, Lee W, Hwang T, Kim D (2014) on gold. Anal Chem 70:4670–4677
Probabilistic evaluation of surface-enhanced
Chapter 3
Abstract
Optical biosensors based on surface plasmon resonance (SPR) phenomenon have received a great deal of
attention in cellular analysis applications. Sensitive and high-resolution SPR imaging (SPRi) platforms are
very useful for real-time monitoring and measurement of individual cell responses to various exogenous
substances. In cellular analysis, mainstream SPR-based sensors have potential for investigations of cell
responses under ambient conditions. Evaluations that account only for the average response of cell
monolayers mask the understanding of precise cell-molecular interactions or intracellular reactions at the
level of individual cells. SPR/SPRi technology has attracted a great deal of attention for detecting the
response of cell monolayers to various substances cultivated on the gold sensor chip. To unleash the full
strength of SPRi technology in complex cell bio-systems, the applied SPR imaging system needs to be
sufficiently effective to allow evaluation of a compound’s potency, specificity, selectivity, toxicity, and
effectiveness at the level of the individual cell. In our studies, we explore the utility of high-resolution
2D-SPR imaging for real-time monitoring of intracellular translocation of protein kinase C (PKC), and
detection of neuronal differentiation in live cells at the level of individual cells. The PC12 cell line, which is
one of the most commonly used neuronal precursor cell lines for research on neuronal differentiation, was
chosen as a nerve cell model. Two dimensional SPR (2D-SPR) signals/images are successfully generated.
We have found that cells treated with the differentiation factor nerve growth factor (NGF) showed a
remarkable enhancement of SPR response to stimulation by muscarine, a nonselective agonist of the
muscarinic acetylcholine receptor.
Key words Nerve growth factor, Surface plasmon resonance, Surface plasmon resonance imaging,
Muscarine, Protein kinase C
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_3,
© Springer Science+Business Media LLC 2017
31
32 Tanveer Ahmad Mir and Hiroaki Shinohara
Reflection
P-polarizer Prism Prism light
Incident
light q
LED
Incident
angle
Collimator Magnification CCD
lens unit camera
(1,2,4,7X)
Fig. 1 (a) Shows a 2D-SPRi apparatus of Kretchmann configuration. (b) Schematic illustration of the used 2D-
SPR system equipped with collimator and a CCD camera with four kinds of lens (1, 2, 4, and 7) for
image magnification. A light emitting diode (LED) produces the incident light beam (670 or 770 nm) after
passing through a collimator lens that illuminates the back side of the sensor chip through the coupling prism.
The light beam from a collimated source passes through a polarizer filter and impinges on the sensor chip
containing cells at a specific angle near the SPR angle. The reflected light is passed through a narrow band-
pass filter, a 7 microscope objective collects the reflected light and forms reflectivity images that are
detected recorded by the digital CCD camera at a fixed angle
34 Tanveer Ahmad Mir and Hiroaki Shinohara
2 Materials
1. PC12 cell line (cell no. RCB0009) derived from rat adrenal
gland is obtained from RIKEN BioResource Center.
2. Dulbecco’s Modified Eagle Medium (DMEM) is obtained
from GIBCO (Cat. No. 31600-034).
3. Horse serum is obtained from GIBCO (Cat. No. 16050-122).
4. Fetal bovine serum (FBS) is obtained from (ICN Biomedicals,
Cat. No. F4135).
5. Penicillin/streptomycin is obtained from GIBCO (Cat. No.
15070-063).
6. 0.05% Trypsin-EDTA is obtained from GIBCO (Cat. No.
25200).
7. NaCl is obtained from Wako Pure Chemicals Industries (Cat.
No. 191-01665).
8. NaOH is obtained from Kanto Chemical (Cat. No. 37184-00).
9. KCl is obtained from Wako Pure Chemicals Industries (Cat.
No. 163-03545).
10. CaCl2 2H2O is obtained from Wako Pure Chemicals Industries
(Cat. No. 031-00435).
11. NaHCO3 is obtained from Wako Pure Chemicals Industries
(Cat. No. 191-01305).
12. Glucose is obtained from Wako Pure Chemicals Industries
(Cat. No. 041-00595).
36 Tanveer Ahmad Mir and Hiroaki Shinohara
3 Methods
3.1 Cell Culture and 1. A 50-nm gold thin film-coated high-refractive index glass
Preparation of SPR (SF6) chip adhered with a square well of flexiPERM (11 7
Sensor Surface 10 mm) is used (see Note 1).
2. Prior to cell culture, SPR sensor slides are put into small cell
culture dishes and are exposed to ultraviolet (UV) sterilization
for 10 min.
3. Cryopreserved nerve model cells (PC12 cells) are harvested
and then transferred into a 15 ml centrifuge tube containing
5 ml DMEM medium supplemented with 10% horse serum
(HS) and 5% fetal bovine serum (FBS) (see Note 2).
4. T-25 flasks are filled with 5 ml of cell suspension and incubated
in a cell culture incubator for 3–4 days (see Note 3).
5. After a given incubation period, T-25 flask is taken into a sterile
clean bench. Old medium is removed and cells are thoroughly
washed with sterile Hanks’ balanced salts (HBBS) solution.
6. To detach the cells from T-25 flask surface, 1 ml of 0.05%
trypsin-EDTA is added and agitated gently for 1 min. Finally,
4 ml of medium (DMEM, 10% HS, 5% FBS) is added, the total
content is transferred into a 15 ml tube to stop the function of
trypsin-EDTA and centrifuged.
7. After centrifugation, supernatant is discarded and the pellet
aggregate is mechanically separated into single cells by gentle
trituration of the suspension using pipette.
8. To prepare a SPR sensor surface of cellular analysis, a homoge-
neous solution is diluted in culture medium so that the cell
suspension becomes 4.0 105 cells/ml. A total volume of
300 μl of medium containing cell suspension is poured onto
the sensor chip to prepare one sensor sample (see Note 4).
9. For cellular analysis using NGF-treated neuronal differentiated
PC12 cells, a homogeneous solution is diluted in culture
medium so that the cell suspension becomes 3.0 105. A total
volume of 300 μl of medium containing cell suspension is
poured onto the sensor chip to prepare one sample (see Note 5).
38 Tanveer Ahmad Mir and Hiroaki Shinohara
3.3 2D-SPR Imaging 2D-SPR experiments are performed using a high-resolution 2D-
Experimental Setup SPR apparatus, as shown schematically in Fig. 1. A 50-nm gold-
coated high-refractive index glass chip, assembled with a square
wall-type reusable cell culture chamber system (flexiPERM) made
of silicone that can easily stick on the sensor chip due to its naturally
adhesive underside to all smooth surfaces without glue creating
growth chambers. PC12 cells are maintained on the sensor chips
for over 24 h in a humidified atmosphere containing 5% CO2 at
37 C. After taking out sensor chip from the incubator, the growth
medium is discarded, and cells are rinsed twice with Hanks’ solu-
tion (pH 7.4, 37 C). After washing, Hanks’ solution (240 μl) is
added to the sensor chip. To eliminate the unwanted reflection and
recording of reflection image of P-polarized parallel incident light,
the sensor chip is adjusted on the top of the prism using index
matching fluid. Thereafter, a volume of 30 μl of stimulant solution
(KCl and PMA) or reference solution for negative control (Hanks’
solution) is carefully injected onto the sensor slides using a manual
pipette. During the experiment, SPR curves (reflection intensity
SPR Imaging for Cellular Analysis 39
changes) are monitored in real time and SPR images are recorded.
Offline data is analyzed on a computer connected with the mea-
surement system using 2D-SPR analysis software (2D-SPR analysis
software provided by NTT-AT Company with the 2D-SPR appara-
tus). For inhibitory response, PC12 cells are pre-incubated with
100 nM staurosporine (a PKC inhibitor) solution for 30 min prior
to PMA treatment.
In order to evaluate reflection intensity change response of
various cell locations with 2D-SPRI at the individual cell level,
from the total cell population on the chip observation region a
random sorting of cell regions of interest is done and the reflection
intensity percentage of the selected cell areas is averaged. The time-
dependent reflection intensity change responses are determined as
the percentage of reflected light intensity of P-polarized light to
that of s-polarized light at a single incident angle as a function of
time by simply subtracting cell region SPR responses recorded
before (negative control) and after exposing to stimuli. To deter-
mine a resonance angle, SPR angles of the sensor surface with cells
and the bare surface are measured by changing the incident angle
from 49 to 57 . For overall data analysis, a threshold of reflection
intensity is used to decide whether the cell has responded to stimuli
or not. For example, responded cells are determined by ΔRI ≧ 15%
over reflection intensity increase at 1 min from K+ stimulation,
ΔRI ≧ 5% over reflection intensity increase at 24 min from phor-
bol-12-myristate-13-acetate (PMA) injection, ΔRI ≧ 3% over
reflection intensity increase 20 min from muscarine injection,
respectively.
Fig. 2 (a) 2D-SPR image of the sensor chip surface is recorded before K+ injection and after the injection of
Hanks’ solution (negative control). (b) 2D-SPR image of the sensor chip surface recorded after 100 mM KCl
injection, showing a robust increase in reflection intensity in PC12 cell regions. (c) Time course of the
reflection intensity changes at individual PC12 cell regions on KCl stimulation with the 2D-SPR imager. (d)
Representative columns indicating the percentages of SPR-responding cells according to concentrations of
KCl. Reprinted from ref. 6, with permission from Elsevier
3.5 High KCl-Induced 1. Prior to experiment, the 2D-SPR imager is assembled. The
Response in PC12 sensor chip on which cells are adhered is fixed on the prism of
Cells Observed with the 2D-SPR imager.
2D-SPR Imager 2. The cell and blank areas on the sensor chip surface are observed
and then the measurement is started. SPR curve that is depen-
dence of reflectivity of the area of interest on incident angle is
first evaluated to get exact resonance angle and to obtain
suitable measurement angle.
3. SPR curve in terms of time course measurement after exposing
to stimulant is observed at the suitable measurement resonance
angle.
4. After stabilization of baseline, cells are first exposed to Hanks’
solution as reference. As expected, no change in reflection
intensity is observed after exposing to negative control solution
(see Note 10).
5. To monitor SPRi sensorgram for intracellular signal transduc-
tion pathway (mainly PKC translocation) via depolarization,
the cells are exposed to different concentrations of K+ solution
and sequential alterations of average reflected light intensity in
terms time course observation for each pixel of the selected
areas of interest on the monitor are obtained and analyzed
using the 2D-SPR analyzer software and images are captured
by the CCD camera.
6. After exposing cell to K+ solution, 2D-SPR images of PC12 cell
areas are gone from dim to bright and variation in cell-to-cell
reflection intensity change response is also observed. It is
speculated that this variation may be due to differences in the
physique of the respective cells (Fig. 2) (see Note 11).
3.6 PKC Agonist and 1. To confirm the results whether K+-induced reflection intensity
Antagonist-Induced change in 2D-SPR sensing is genuinely implicated to intracel-
Response in PC12 lular signal transduction (translocation of PKC) in PC12 cells,
Cells Observed with cells are exposed to Phorbol-12-myristate-13-acetate (PMA), a
2D-SPR Imager specific activator of Protein Kinase C (PKC).
2. SPR images of the PC12 cells recorded with the CCD camera
before and after stimulation of varying the concentrations of
Phorbol-12-myristate-13-acetate (PMA), the obtained SPR
curves showed a slow increase with fluctuation after gaining
phorbol-12-myristate-13-acetate (PMA) access to the cell inte-
rior (Fig. 3) (see Note 12).
3. To further validate the results and examine the inhibitory
effects, PC12 cells are pretreated with a potent inhibitor of
PKC (staurosporine) for 30 min, on the Phorbol-12-myris-
tate-13-acetate-induced response (see Note 13).
42 Tanveer Ahmad Mir and Hiroaki Shinohara
Fig. 3 (a) 2D-SPR image of the sensor chip surface obtained after the injection of Hanks’ solution for
background observation. (b) 2D-SPR image of PC12 cell regions obtained after phorbol-12-myristate-13-
acetate (PMA)stimulation (500 nM), which showed enhancement of the reflection intensity in cell regions. (c)
Reflection intensity change corresponding to the intracellular refractive index change, measured with the 2D-
SPR imager for PC12 cells adhered to a gold chip on 500 nM phorbol-12-myristate-13-acetate (PMA)
injection. After injecting 500 nM PMA, a slow elevation of reflection intensity was seen at cell regions. No
reflection intensity change is observed in non-cell regions. (d) Representative columns indicating the
percentages of SPR-responded cells in the presence (red) or absence (blue) of 100 nM staurosporine.
Reprinted from ref. 6, with permission from Elsevier
a
3
28
Reflection intensity / %
Δ Reflection intensity / %
No cell region (1)
6
16
0 5 10 15 20 25 30 4
Time / min
b 2
54 1
Reflection intensity / %
40
5
Fig. 4 Time course of the reflection intensity change for undifferentiated (a) and differentiated (b) PC12 cells at
individual cell regions on muscarine stimulation with the 2D-SPR imager. After the application of muscarine,
slow and fluctuating elevation of reflection intensity was seen at cell regions. No reflection intensity change is
observed at the non-cell region. (c) Dependence of reflection intensity change at the undifferentiated cell
regions and the differentiated cell regions (2 days treatment, 4 days treatment) on the concentration of
muscarine as a stimulant. The PC12 cells are treated with 80 ng/ml NGF for 2 or 4 days. Reprinted from ref. 7,
with permission from Elsevier
4 Notes
References
1. Suzuki M, Ohshima T, Hane S, Iribe Y, Tobita T 4. Horii M, Shinohara H, Irebe Y, Suzuki M
(2007) Multiscale 2D-SPR biosensing for cell (2007) Application of high resolution 2D-SPR
chips. J Robot Mechatron 19:5519–5523 imager to living cell-based allergen sensing. In:
2. Homola J (2008) Surface plasmon resonance Viovy J-L, Tabeling P, Descroix S, Malaquin L
sensors for detection of chemical and biological (eds) The proceedings of lTAS 2007 conference.
species. Chem Rev 108(2):462–493 Chemical and biological microsystems society,
3. Yanase Y, Hiragun T, Ishii K, Kawaguchi T, Paris, pp 451–453
Yanase T, Kawai M, Sakamoto K, Hide M 5. Horii M, Shinohara H, Irebe Y, Suzuki M
(2014) Surface plasmon resonance for cell- (2011) Living cell-based allergen sensing using
based clinical diagnosis. Sensors 14 a high resolution two-dimensional surface plas-
(3):4948–4959 mon resonance imager. Analyst 136:2706–2711
46 Tanveer Ahmad Mir and Hiroaki Shinohara
6. Shinohara H, Sakai Y, Mir TA (2013) Real-time by 2D-SPR sensor: an approach to study neuro-
monitoring of intracellular signal transduction in nal differentiation. In: Proceeding of the 14th
PC12 cells by two-dimensional surface plasmon international meeting on chemical sensors. doi:
resonance imager. Anal Biochem 441:185–189 10.5162/IMCS 2012/P1.1.18
7. Mir TA, Shinohara H (2013) Two-dimensional 9. Mir TA and Shinohara H (2012) 2D-SPR bio-
surface plasmon resonance imager: an approach sensor detects the intracellular signal transduc-
to study neuronal differentiation. Anal Biochem tion in PC12 cells at single cell level. In:
443:46–51 Proceeding of the sixth international conference
8. Mir TA and Shinohara H (2012) Real-time on sensing technology. doi: 10.1109/ICSensT.
monitoring of cell response to drug stimulation 2012.6461763
Chapter 4
Abstract
Surface Plasmon resonance (SPR) optical fiber biosensors constitute a miniaturized counterpart to the
bulky prism configuration and offer remote operation in very small volumes of analyte. They are a cost-
effective and relatively straightforward technique to yield in situ (or even possibly in vivo) molecular
detection. They are usually obtained from a gold-coated fiber segment for which the core-guided light is
brought into contact with the surrounding medium, either by etching (or side-polishing) or by using
grating coupling. Recently, SPR generation was achieved in gold-coated tilted fiber Bragg gratings
(TFBGs). These sensors probe the surrounding medium with near-infrared narrowband resonances,
which enhances both the penetration depth of the evanescent field in the external medium and the
wavelength resolution of the interrogation. They constitute the unique configuration able to probe all
the fiber cladding modes individually, with high Q-factors. We use these unique spectral features in our
work to sense proteins and extra-cellular membrane receptors that are both overexpressed in cancerous
tissues. Impressive limit of detection (LOD) and sensitivity are reported, which paves the way for the
further use of such immunosensors for cancer diagnosis.
Key words Surface plasmon resonance, Optical fiber sensors, Bragg gratings, Immunosensing, Cells,
Proteins
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_4,
© Springer Science+Business Media LLC 2017
47
48 Christophe Caucheteur et al.
Fig. 1 Sketch of the Kretschmann-Raether prism approach for plasmonic generation and operating principle of
its interrogation to measure binding of biomolecules on receptors grafted on the gold substrate
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 49
these probes that are inherently polarization selective [9, 10] and
on their subsequent use for biochemical sensing [11, 12].
In our work, we use the well-acknowledged antibody-antigen
affinity mechanism to assess the unique protein detection and
quantification capabilities of gold-coated TFBGs and evaluate
their performances in terms of both sensitivity and limit of detec-
tion (LOD). We have conducted experiments on two kinds of
bioreceptors, e.g., proteins and extracellular membrane receptors
that are both overexpressed in the case of cancer cells. In particular,
we have quantified a certain type of cytokeratins that are secreted by
lung cancer tumors and cells suspensions. Using another functional
film on the same sensor, we have also detected extracellular mem-
brane receptors in native membranes of different human epithelial
cell lines. A differential diagnosis has been demonstrated between
two cell lines, with overexpressed membrane receptors (positive
control) and with a low number of these receptors (negative con-
trol). Such results make an important step forward toward the
demonstration of in vivo diagnosis.
In the remaining of this chapter, we will first describe the
operating principle of the sensor. Then, we will focus on its perfor-
mances in the quantification of proteins and cells in vitro.
2 Materials
2.3 Optical System to 1. Optical vector analyzer (Luna Technologies OVA CTe).
Measure the Spectrum 2. In-line linear polarizer (General Photonics).
of Gold-Coated TFBGs
3. In-house developed microfluidic chamber to immerse gold-
coated TFBGs in controlled liquids.
4. Hand-held Abbe refractometer accurate to 104 RIU (Reichert
AR200).
52 Christophe Caucheteur et al.
3 Methods
3.1 Operating The sensor is obtained from a single-mode optical fiber in which a
Principle of TFBGs photo-imprinted refractive index grating is formed over a short
section of the core, and that is further coated with a bilayer coating:
a very thin metal coating and a suitable biochemical recognition
binding layer.
The glass optical fiber is a 125-μm-thick cylindrical waveguide
made of two concentric layers, the core in the middle surrounded
by a cladding that is thick enough to prevent light propagating in
the core to interact with the fiber surroundings. The refractive
index of the core is slightly higher than that of the cladding to
allow for the guidance of light in the fiber core. Our experiments
are performed on 8 μm core single-mode optical fibers that guide
light into a single optical mode at wavelengths between 1300 and
1650 nm. Such fibers are widely available at low cost (less than 100
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 53
Fig. 2 Sketch of the light coupling mechanism for uniform FBGs (a) and tilted FBGs (b)
3.4 Surface The gold-coated TFBGs are functionalized for biosensing pur-
Functionalization poses. The chemistry involved in this process depends on the target
application. In our case study, it is based on the antigen/antibody
affinity. Whatever the analyte to be detected, a self-assembled
monolayer (SAM) is manufactured, prior to the biomolecules
immobilization.
1. For this, gold-coated TFBGs were first thoroughly rinsed with
ethanol and dried under nitrogen.
2. Gold-coated TFBGs were then immersed in a solution of thiols
(2 mM) dispersed in ethanol. Thiols incubations were usually
done during 18 h at room temperature in a 1-mm-thick capil-
lary tube sealed at both ends to prevent solvent evaporation. At
the end of the incubation, the functionalized gold-coated
TFBGs were removed from the tube and again rinsed with
ethanol (see Note 1).
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 57
Fig. 5 Sketch of a gold-coated tilted fiber Bragg grating used for biosensing
purposes which displays the functionalized photo-inscribed optical fiber inserted
in a packaging
58 Christophe Caucheteur et al.
Fig. 7 Transmitted amplitude spectra for two orthogonal SOPs (radial and
azimuthal polarizations) of a TFBG immersed in salted water (SRI ¼ 1.338)
0
-10
-20
n=1.325
-30
Amplitude [dB]
0
-10
-20
n=1.340
-30
0
-10
-20
n=1.360
-30
1525 1530 1535 1540 1545 1550 1555 1560 1565 1570 1575 1580
Wavelength [nm]
Fig. 8 SPR signature in the transmitted spectrum of gold-coated TFBG for coarse
changes in SRI
1560
wavelength SPR
Linear fit
1555
1550
Wavelength [nm]
1545
1540
1535
Slope=553nm/RIU
R=99.66%
1530
1.32 1.33 1.34 1.35 1.36 1.37
Surrounding refractive index [RIU]
-6
-8
Relative Transmission (dB)
-10
-12
-14
-16
-18
1529.4 1529.6 1529.8 1530.0 1530.2 1530.4 1530.6 1546.0 1546.2 1546.4 1546.6 1546.8 1547.0 1576.8 1577.0 1577.2 1577.4 1577.6
Wavelength (nm)
Fig. 10 Spectra measured during a biosensing experiment (a) with three spectral regions shown in detail: on
the short wavelength side of the SPR (b); near the SPR (+2 mode) (c); and on the long wavelength side (d)
near the SPR and a few nm away, it becomes clear that the presence
of a comb of resonances with widely different sensitivities allows for
very small wavelength shifts to be detected unambiguously with
high precision. Most often, the focus is made on the +2 mode
among the spectral comb, as it appears to be the most sensitive in
terms of both amplitude variation and wavelength shift.
Optical fiber devices have two important advantages over bulky
prism SPR configurations: light propagates essentially without loss
in short lengths of fibers, resulting in very high signal-to-noise
ratios, and interfacing devices to light sources and detectors con-
sists essentially of plugging connectors into widely available fiber
optic instrumentation, instead of having to carefully align optical
beams through imaging systems. In terms of experimentally
demonstrated FOM, TFBGs exhibit a value reaching 5000, which
surpasses all configurations by more than one order of magnitude
[2]. As shown in Fig. 11, this results from the fact that they exhibit
narrow resonance bands (FWHM ~0.1–0.2 nm) compared to even
the best possible theoretical value (~5 nm obtained by calculating
the reflection from the base of a prism in the Kretschmann-Raether
configuration), also keeping in mind that the experimental SPR
FWHM from other fiber configurations all exceed 20 nm and more.
Fig. 11 Comparison between the best theoretical SPR response for 50 nm gold on
silica in the Kretschmann-Raether configuration (thick blue line) and a measured
TFBG-SPR spectrum with the same thickness of gold (thin red line). The arrows
indicate the resonance to be followed in each case
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 63
3.6 Proteins and Up to now, and to the best of our knowledge, there have been a few
Cells Quantification reports where gold-coated TFBGs were used for biochemical sens-
with Gold-Coated TFBG ing. In [23], the probe was associated with aptamers and a mea-
Immunosensors surement of the dissociation constant was demonstrated. In [24],
the probe was used to measure the intracellular density of non-
physiological cells, namely human acute leukemia cells. In [25], the
sensor was integrated into cell culture equipment and was used for
real-time monitoring of cellular response to chemical stimuli
obtained by adjunction of trypsin, serum, and sodium azide. The
corresponding effects—detachment of cells from the surface,
uptake of serum, and inhibition of cellular metabolism,
respectively—were monitored through a shift of the SPR signature
in the transmitted amplitude spectrum of gold-coated TFBGs.
In the following, we summarize our main achievements toward
the demonstration of lab-on-fiber devices with gold-coated TFBGs
suited for cancer diagnosis [26, 27].
3.7 Sensing A diverse range of tumor markers has been associated with lung
Cytokeratins cancer. In this study, we focused on cytokeratins 7 (CK7) that are
particularly useful tools for diagnosis in oncology. In particular,
CK7 profile of lung tumor has proved to be a useful aid in the
differential diagnosis of carcinomas, since primary and metastatic
tumors present different profiles. In fact, primary lung tumors
express cytokeratin 7 (CK7+) while secondary tumors are deficient
in CK7 (CK7). Moreover, it has been demonstrated that cytoker-
atin fragments can be released from malignant cells and conse-
quently CK fragments can be located in blood circulation and are
therefore easily accessible with an optical fiber properly modified.
The cytokeratin 7 antigen detection by the bioreceptor is based
on the specific chemical reaction with its corresponding antibody
(AbCK7) previously immobilized on the optical fiber surface. The
operating principle is based on the recognition of the cytokeratin 7
antigen epitope by the fragment antigen-binding (Fab), a region
present on the cytokeratin 7 antibody. In addition to the full protein
(CK7FP) detection made of 469 amino acids, corresponding to a
large biomolecule, we proposed here to monitor a protein fragment
called cytokeratin 7 peptide (CK7pep) made of only 23 amino acids
to present a comparative study depending on the size of the target.
In fact, the detection of small molecule antigens remains a challenge
in the SPR immunosensing. Most of the SPR biosensors are based on
large molecule detection since SPR response in the presence of small
mass suffers from various disadvantages not encountered with full
protein, such as low signal-to-noise ratio.
1. Detection of CK7 full protein.
(a) Gold-coated TFBGs, preliminary functionalized with
cytokeratin 7 antibodies, have been first connected to the
LUNA and held straight between two clamps, for trans-
mission recording (see Note 4).
64 Christophe Caucheteur et al.
-10 4
(a) (b)
Biosensor Response (dB)
3
-11
Amplitude (dB)
2
-12
-13
0
1E-12 1E-11 1E-10 1E-9 1E-8 1E-7 CK7 peptide CK7FP Control
Concentration CK7FP (g/mL) Samples in PBS
Fig. 12 Amplitude monitoring of the sensitive mode in the presence of CK7FP at different concentrations
diluted in PBS (a). Biosensor response obtained for optical fiber functionalized with AbCK7, in the presence of
CK7 peptide or CK7FP in comparison with OF functionalized without AbCK7 (b)
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 65
-6
(b)
3
-8
Biosensor Response (dB)
Amplitude (dBm)
1E-9 g/mL
2E-9 g/mL
-10 3E-9 g/mL
4E-9 g/mL
2
5E-9 g/mL
1E-8 g/mL
2E-8 g/mL
-12 3E-8 g/mL
4E-8 g/mL
5E-8 g/mL
6E-8 g/mL
7E-8 g/mL
-14 1E-7 g/mL
1
2E-7 g/mL
5E-7 g/mL
1E-6 g/mL
-16
(a) 0
1545.8 1546.0 1546.2 1546.4 1546.6 1546.8 1547.0 CK7 peptide diluted in PBS + serum Control PBS + serum
Wavelength (nm)
Fig. 13 Shift of the sensitive mode during incubation of the optical fiber functionalized with AbCK7 on CK7
peptide at different concentrations diluted in PBS + 10% FBS (a). Biosensor response obtained for optical fiber
functionalized with AbCK7 immersed in PBS + 10% FBS, in the presence or in the absence of CK7 peptide (b)
66 Christophe Caucheteur et al.
3.8 Sensing In this section, we report on the use of gold-coated TFBGs for
Transmembrane selective cellular detection through membrane protein targeting.
Receptors The focus is made on the epithelial growth factor receptor (EGFR),
which is a transmembrane receptor from the 4-tyrosine kinase
receptors family. It is an important biomarker and therapeutic
target that it is over-expressed by numerous cancer cells.
1. Surface functionalization.
The sensor surface selectivity was ensured by bio-
functionalization through a two-step approach.
(a) First, the clean waveguide surface was activated with the
Carboxylic acid-SAM formation reagent (cat. n: C488)
obtained from Dojindo (Japan).
(b) Then, monoclonal mouse immunoglobulin G (IgG)
raised against the human epidermoid carcinoma cell line
(Santa Cruz Biotechnology Inc. and American Type Cul-
ture Collection (USA)) were immobilized on the surface
through carbodiimide covalent biochemistry using Dojin-
do’s amine coupling kit (cat. n: A515-10). This antibody
was diluted in the Dojindo’s reaction buffer to 0.01 μg/
mL for efficient covalent immobilization during 30 min.
The manufacturer’s instructions were followed for each
step for both processes.
2. EGFR detection by SPR-TFBG.
Two cell lines were used: the first one with overexpressed
human epidermal growth factor receptors (EGFRs)—A431
cell line, (EGFR positive, EGFR (+)). The second cell line
was EGFR negative – OCM1 cell line, (EGFR ()).
(a) The culture process was done at 37 C in a humidified
incubator with 5% CO2 atmosphere. No antibiotic was
used and cell cultures were free of mycoplasma and patho-
genic viruses. Cells suspensions were then prepared for
SPR experiments. For this, A431 and OCM1 cells from a
confluent monolayer were detached mechanically by a
gentle scraping of cells from the growth surface into ice-
cold phosphate buffered saline (PBS). Cells were then
washed three times in cold PBS by repeated centrifugation
(at 2500 RPM and 4 C for 3 min). Pellets were then again
suspended to a concentration of 2–5 106 cells/mL.
0.5 mL volumes were used for experiments. A handheld
automated cell counter from Millipore (Scepter 2.0,
PHCC00000) was used to count the cells.
(b) For immunosensing experiments, gold-coated TFBGs were
hold straight between two clamps and were then
approached toward the cells suspensions, using a vertical
translation stage. To demonstrate that a differential
response can be obtained between both cell lines, TFBGs
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 67
Fig. 14 Different assays presenting specific cell interactions. Dark gray bars
represent A431 cells, light gray bars represent OCM1 cells. Blank bars are
washing and rinsing steps for RPMI-1640 cell culture media supplemented
with FBS
68 Christophe Caucheteur et al.
Table 1
Sensor response as a function of the cells concentration, bringing an
estimate about the LOD
4 Notes
References
1. Kretschmann E, Raether H (1968) Radiative 13. Othonos A, Kalli K (1999) Fiber Bragg gratings:
decay of non radiative surface plasmon excited fundamentals and applications in telecommuni-
by light. Z Naturforsch 23:2135 cations and sensing. Artech House, London
2. Caucheteur C, Guo T, Albert J (2015) Review 14. Erdogan T (1997) Fiber grating spectra.
of recent plasmonic fiber optic biochemical J Lightwave Technol 15:1277–1294
sensors: improving the limit of detection. 15. Erdogan T, Sipe JE (1996) Tilted fiber phase
Anal Bioanal Chem 407:3883 gratings. JOSA A 13:296–313
3. Pollet J, Delport F, Janssen KPF, Jans K, Maes 16. Laffont G, Ferdinand P (2001) Tilted short-
G, Pfeiffer H, Wevers M, Lammertyn J (2009) period fiber Bragg grating induced coupling to
Fiber optic SPR biosensing of DNA hybridiza- cladding modes for accurate refractometry.
tion and DNA-protein interactions. Biosens Meas Sci Technol 12:765–772
Bioelectron 25:864–869 17. Caucheteur C, Mégret P (2005) Demodula-
4. Baldini F, Brenci M, Chiavaioli F, Gianetti A, tion technique for weakly tilted fiber Bragg
Trono C (2012) Optical fiber gratings as tools grating refractometer. Photon Technol Lett
for chemical and biochemical sensing. Anal 17:2703–2705
Bioanal Chem 402:109–116 18. Chan CF, Chen C, Jafari A, Laronche A, Thom-
5. Albert J, Shao L-Y, Caucheteur C (2013) son DJ, Albert J (2007) Optical fiber refractom-
Tilted fiber Bragg grating sensors. Laser Pho- eter using narrowband cladding-mode
ton Rev 7:83–108 resonance shifts. Appl Optics 46:1142–1149
6. Sharma AK, Rajan J, Gupta BD (2007) Fiber- 19. Chen C, Albert J (2006) Stain-optic coeffi-
optic sensors based on surface plasmon reso- cients of individual cladding modes of single
nance: a comprehensive review. IEEE Sensors J mode fibre: theory and experiment. Electron
7:1118–1129 Lett 42:1027–1028
7. Offermans P, Shaafsma MC, Rodriguez SRK, 20. White IM, Fan XD (2008) On the performance
Zhang Y, Crego-Calama M, Brongersma SH, quantification of resonant refractive index sen-
Rivas JG (2011) Universal scaling of the figure sors. Opt Express 16:1020–1028
of merit of plasmonic sensors. ACS Nano 21. Shevchenko YY, Chen C, Dakka MA, Albert J
5:5151–5157 (2010) Polarization selective grating excitation
8. Shevchenko YY, Albert J (2007) Plasmon reso- of plasmons in cylindrical optical fibers. Opt
nances in gold-coated tilted fiber Bragg grat- Lett 35:637–639
ings. Opt Lett 32:211–213 22. Caucheteur C, Chen C, Voisin V, Berini P,
9. Caucheteur C, Shevchenko YY, Shao L-Y, Albert J (2011) A thin metal sheath lifts the
Wuilpart M, Albert J (2011) High resolution EH to HE degeneracy in the cladding mode
interrogation of tilted fiber grating SPR sensors refractometric sensitivity of optical fiber sen-
from polarization properties measurement. sors. Appl Phys Lett 99:041118
Opt Express 19:1656–1664 23. Shevchenko Y, Francis TJ, Blair DAD, Walsh R,
10. Caucheteur C, Voisin V, Albert J (2013) DeRosa MC, Albert J (2011) In situ biosensing
Polarized spectral combs probe optical with a surface plasmon resonance fiber grating
fiber surface plasmons. Opt Express aptasensor. Anal Chem 83:7027–7034
21:3055–3066 24. Guo T, Liu F, Liu Y, Chen NK, Guan BO,
11. Albert J, Lepinay S, Caucheteur C, DeRosa Albert J (2014) In situ detection of density
MC (2013) High resolution grating-assisted alteration in non-physiological cells with polar-
surface plasmon resonance fiber optic aptasen- imetric tilted fiber grating sensors. Biosens
sor. Methods 63:239–254 Bioelectron 55:452–458
12. Voisin V, Pilate J, Damman P, Mégret P, Cau- 25. Shevchenko Y, Camci-Unal G, Cuttica DF,
cheteur C (2014) Highly sensitive detection of Dokmeci MR, Albert J, Khademhosseini A
molecular interactions with plasmonic optical (2014) Surface plasmon resonance fiber sensor
fiber grating sensors. Biosens Bioelectron for real-time and label-free monitoring of cellu-
51:249–254 lar behavior. Biosens Bioelectron 56:359–367
Immunosensing with Near-Infrared Plasmonic Optical Fiber Gratings 71
26. Ribaut C, Voisin V, Malachovska V, Dubois V, 27. Malachovska V, Ribaut C, Voisin V, Surin M,
Mégret P, Wattiez R, Caucheteur C (2016) Leclère P, Wattiez R, Caucheteur C (2015)
Small biomolecule immunosensing with plas- Fiber-optic SPR immunosensors tailored to
monic optical fiber grating sensor. Biosens target epithetial cells through membrane
Bioelectron 77:315–322. doi:10.1016/j.bios. receptors. Anal Chem 87:5957–5965
2015.09.019
Chapter 5
Abstract
In spite of the high analytic potential of Magneto Optical Surface Plasmon Resonance (MOSPR) assays,
their applicability to biosensing has been limited due to significant chip stability issues. We present novel
solutions to surpass current limitations of MOSPR sensing assays, based on innovative chip structure,
tailored measurements and improved data analysis methods. The structure of the chip is modified to contain
a thin layer of Co-Au alloy instead of successive layers of homogenous metals with magnetic and plasmonic
properties, as currently used. This new approach presents improved plasmonic and magnetic properties, yet
a structural stability similar to standard Au-SPR chips, allowing for bioaffinity assays in saline solutions.
Moreover, using a custom-designed measurement configuration that allows the acquisition of the SPR
curve, i.e., the reflectivity measured at multiple angles of incidence, instead of the reflectivity value at a
single-incidence angle, a high signal-to-noise ratio is achieved, suitable for detection of minute analyte
concentrations. The proposed structure of the MOSPR sensing chip and the procedure of data analysis
allow for long time assessment in liquid media, a significant advancement over existing MOSPR chips, and
confirm the MOSPR increased sensitivity over standard SPR analyses.
Key words Magneto-optic surface plasmon resonance, Magnetic alloys, Surface plasmon resonance
enhancement, Affinity biosensor, Angle-resolved surface plasmon resonance, Fixed-angle surface
plasmon resonance
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_5,
© Springer Science+Business Media LLC 2017
73
74 Sorin David et al.
n2
θc ¼ ð1Þ
n1
where n2 is the refractive index of the upper medium and n1 is the
refractive index of the lower medium and the light travels from n1
toward n2.
The coupling of the light into the metal film is done via a
grating (Otto configuration) or a prism (Kretschmann configura-
tion) [1]. In the following, only the Kretschmann configuration is
considered.
Depending on the value of the incident light angle θ, the type
of the plasmonic metal, and the refractive indices of the two media,
the light interacts with the free electrons of the metal film and the
energy of the photons is transferred resonantly to these electrons. A
drop in the intensity of the reflected light is recorded by a suitable
detector (e.g., a photodetector array) placed in the path of the
reflected beam—Fig. 1 corresponding to the specific SPR angle.
The dependency of the intensity of the reflected light over the
incidence angle generates the SPR curve that has a dip minimum
corresponding to the SPR angle, θmin (Fig. 1). According to Eq. 2
[2], the SPR angle depends on λ (the wavelength of the incident
light), via n*m, the complex refractive index of the plasmonic layer
and the values of the refractive indices of the media n1 and n2.
sffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
n∗2 n22
sin θmin ¼ ∗2m ; n∗ ¼ nm þ i k m : ð2Þ
nm þ n22 n21 m
2 Materials
2.1 Surface 1. Bovine serum albumin (BSA), human IgG (HIgG), affinity
Chemistry isolated anti-human IgG (AHIgG), N-hydroxysuccinimide
(NHS), 1-ethyl-3-(dimethylaminopropyl) carbodiimide
(EDC), ethilenediaminetetraacetic acid (EDTA), and ethanol-
amine were purchased from Sigma–Aldrich (Germany).
2. (1-mercapto-11-undecyl)hexa(ethylene glycol)carboxylic acid
was purchased from Prochimia Surfaces, Poland (see Note 1).
3. Surfactant P20 was provided by GE Healthcare.
4. Matching oil Immersol 518 F, ne ¼ 1.518 from Carl Zeiss.
5. Aqua regia HCl:HNO3 3:1 mixture (see Note 2).
6. piranha solution H2SO4:H2O2 3:2 mixture (see Note 3) .
3 Methods
13. The Au-Co alloy is thermally evaporated at a rate of 0.5 Å/s from
a single tungsten boat containing corresponding quantities of Au
and Co to yield an alloy of 10% Co—volumetric ratio (588 mg Au
and 27 mg Co). Alternative techniques for producing thin films
of magnetic materials and alloys do exist and may be used for a
better control of the film composition and structure. However,
they have limitations in respect to costs (see Note 7), or material
type (see Note 8) .
3.3 (MO)SPR For classical SPR measurements the response is derived from the
Measurements and SPR curve as a function the angle for which the minimum of the
Data Processing reflectivity is recorded [1]. The reflectivity (Rpp) is obtained by
normalizing the intensity of the measured reflected p-polarized
light versus a reference signal (e.g., obtained using an s-polarized
light or a solution with an SPR dip outside the measurement range
of the device). As pinpointed by Eq. 2, the SPR angle (θmin),
depends on the refractive index of the media in the immediate
vicinity of the sensing surface [1] and is determined most precisely
if the reflectivity dip is the sharpest. Changes at the sensing surface,
accompanied by a modification of the refractive index, cause a shift
in the SPR angle that is used for label-free assessment of biomolec-
ular surface interactions. This is the preferred mode of operation for
real-time measurements having a linear dependence on the refrac-
tive index changes and with the amount of molecules accumulated
Biosensing Based on Magneto-Optical Surface Plasmon Resonance 79
Fig. 2 The magneto optical SPR concept and schematic setup: electromagnet for transverse oscillating
magnetic field application; tailored chips with magneto-plasmonic layers; top microfluidics; all system
components were custom developed and built around an integrated illumination and detection Kretschman
configuration as provided by a Spreeta module
on the surface [1]. Based on the full curve analysis, various data
processing methods were developed to improve sensitivity, robust-
ness against noise [16] and foster reduced time of analysis [17].
Figure 2 outlines the MOSPR concept. In brief, the measure-
ment setup, described in more detail elsewhere [18], comprises:
– A SPR module.
– An external electromagnet with power supply for applying the
alternating transverse magnetic field.
– A Flow Injection Analysis (FIA) type setup as fluidic assembly.
The SPR measurement module is based on a SPREETA sensor
model TSPR1K23 (Texas Instruments USA). The FIA comprises a
flow channel connected to an injection pump using PTFE tubes.
The flow channel is made by micromachining a polyether ether
ketone (PEEK) slab and a gasket of PDMS (Fig. 2). The data
acquisition board is designed to give online access to whole curve
measurements to allow calculation/extraction of the MOSPR data.
80 Sorin David et al.
3.3.1 Extracting the The MOSPR signal is calculated from the reflectivity curves
MOSPR Data recorded in different states of magnetization of the MOSPR chip
and is defined as ΔRpp/Rpp [11]. Here, Rpp is the reflectivity
(standard for SPR assays), obtained when not applying a magnetic
field and ΔRpp represents the difference between the reflectivity
measured at one direction of magnetization (M+) and the reflectiv-
ity measured at the opposite direction of magnetization (M).
Figure 3a indicates the reflectivity curves in respect with magneti-
zation as well as the corresponding ΔRpp, Rpp, Δθ (the difference
between the SPR angle measured at M+ and M respectively) and
θmin (the SPR angle measured with no magnetic field) data.
The MOSPR response is derived from the reflectivity values
corresponding to a selected angle of incidence, as the variation of
the reflectivity Rpp while switching the direction of the in plane
magnetization of the sensor [10]:
ΔRpp Rpp ðM þÞ Rpp ðM Þ
¼ ð4Þ
Rpp ð0Þ Rpp ð0Þ
where Rpp(M+) and Rpp(M) represent the reflectivities for the
direction of the magnetization perpendicular to the propagation
plane of the incident p-polarized light in both ways and Rpp(0)
represents the reflectivity without magnetization. In real measure-
ments, ΔRpp is obtained from the amplitude of the oscillations of
the measured reflectivity (Fig. 3a). The MOSPR curve is defined as
the variation of the ΔRpp/Rpp versus the angle of incidence of the
light.
As shown in Fig. 3b, for noise-free data a change in the refrac-
tive index of the media that yields a small reflectivity change in the
SPR signal, determines a larger change in MOSPR signal. To this
effect, an intermediary smoothing step is applied to the raw data.
3.3.2 Fitting Function of Reflectivity data for the entire angular range (SPR curves) are
SPR Data collected and fitted with a rational polynomial equation to achieve
smoothing.
82 Sorin David et al.
Fig. 3 (a) Representative parameters of the measured SPR curves used to derive the MOSPR data: Rpp the
reflectivity value for a selected incidence angle; ΔRpp corresponds to reflectivity differences between M+ and
M magnetization states, Δθ (the difference between the SPR angles for M+ and M magnetization states)
and θmin (the SPR angle measured with no magnetic field). (b) Schematic plot showing ideal SPR (black) and
MOSPR (blue) curves, as well as their variations corresponding to refractive index n (dotted lines) and n + Δn
(continuous lines), respectively
Fig. 4 Theoretical SPR curve for 50 nm layer of gold on glass with water on top
(black dotted line) and the fit with the phenomenological, polynomial function (red
line)
3.3.3 Sensitivity The MOSPR responses are compared with SPR responses in terms
Comparison of MOSPR and of sensitivity and signal-to-noise ratio when analyzing the reflectiv-
SPR Sensors ity at a given, single angle of incidence.
According to Eq. 4 the MOSPR signal is derived from
ΔRpp(θ)/Rpp(θ) data using the value of the reflectivity
corresponding to a selected angle of incidence θs, chosen to
yield the highest sensitivity. At this angle, the MOSPR curve (i.e.,
ΔRpp/Rpp vs. incidence angle) presents the steepest slope [10]. We
derive the θs angle by analyzing the derivative of the fitted SPR
curve. Time series of full SPR curves are recorded, and then fitted
with Eq. 5 and the reflectivity values (corresponding to θs) are
plotted over time. Further, frequency (Fourier) analysis is per-
formed to extract the amplitude of oscillations (corresponding to
ΔRpp) and their mean (corresponding to Rpp). MOSPR and SPR
reflectivity responses derived from raw measurements directly from
the detector corresponding to θs are presented in Fig. 5a while
reflectivity responses calculated from fitted SPR curves are given
in Fig. 5b. The SNR is calculated as the ratio between the response
(SPR, MOSPR) and the corresponding root mean square deviation
(RMSD calculated on 100 data points). The comparison shows
that fitting the curves (and hence reducing the noise) improves
greatly the sensitivity (Fig. 5b). The sensitivity increase for the
84 Sorin David et al.
Fig. 5 (a) Signal-to-noise ratios for Au—SPR (red), Au-Co alloy—MOSPR (black) and Au-Co-Au tri-layer—
MOSPR (blue) derived from raw reflectivity data for different refractive index solutions. (b) Signal-to-noise ratio
for Au—SPR (red), Au-Co alloy—MOSPR (black) and Au-Co-Au tri-layer—MOSPR (blue) for solutions with
different refractive indices—reflectivity values derived from fitted SPR curves
3.4 MOSPR Chip Stability of the MOSPR chips in physiological solutions is tested by
Stability in Liquid exposure to a saline solution (e.g., HEPES buffer solution—HBS)
Media while applying the oscillatory magnetic field. While the alloy shows
no sign of destabilization (for virtually indefinite exposure, similar
to regular Au sensor chips), in the case of Au-Co-Au tri-layer there
is a significant exfoliation of the metallic film (Fig. 6a). Increase in
the transparency of the film (observed under transmission micros-
copy Fig. 6a) further amplified with longer exposure confirms the
conclusion of film exfoliation. In the same flow conditions and
buffers, no modifications were observed in the cases of plain gold
and/or Au-Co alloy (Fig. 6b).
3.5 Bioaffinity Assay MOSPR assays of bioaffinity interactions are performed for the
alloy-based MOSPR chip. Because of the corrosive effect of the
saline buffers the Au-Co-Au tri-layers cannot be used even after
surface modification and functionalization.
Affinity tests, using a model biomolecular interaction between
human immuno-globulin G (HIgG) and Anti-HIgG in a direct
assay [1], were performed. IgG is used as model analyte without
restricting the generality of the method.
Biosensing Based on Magneto-Optical Surface Plasmon Resonance 85
Fig. 6 Optical image of an Au-Co-Au tri-layer chip showing (a) pronounced corrosion in the areas exposed to
saline solution in contrast with (b) non-corroded film
Fig. 7 (a) Relative reflectivity for several specific injections of Anti-HIgG (concentration range 5–80 nM) and
two regeneration steps on the MOSPR Au-Co alloy chip. (b) Signal-to-noise ratio for specific MOSPR (for Au-Co
alloy—black) and SPR (for Au chip—red) responses for several injections of Anti-HIgG
3.6 Concluding The proposed novel solutions, based on innovative chip structure,
Remarks and tailored measurement and improved data analysis methods,
surpass current limitations of MOSPR sensing assays and have
been demonstrated to: (a) enable improved plasmonic and mag-
netic properties yet a structural stability similar to standard Au-SPR
chips, allowing for bioaffinity assays in saline solutions, (b) detec-
tion of minute analyte concentrations (IgG is used as model analyte
without restricting the generality of the method). The custom-
designed MOSPR measurement configuration allows the quasi
simultaneous acquisition of the whole SPR curve at high signal-
to-noise ratios during long time assessment in liquid media, a
significant advancement over existing MOSPR chips, and confirms
the MOSPR increased sensitivity over standard SPR analyses.
4 Notes
Acknowledgments
The authors thank the Romanian Executive Unit for Higher Edu-
cation, Research, Development and Innovation Funding for fund-
ing through grants PN II-ID-PCCE-2011-2-0075 and PN-II-RU-
PD-2012-3-0467.
References
1. Schasfoort R, Tudos A (2008) Handbook of 11. Regatos D, Fariña D, Calle A et al (2010) Au/
surface plasmon resonance. The Royal Society Fe/Au multilayer transducers for magneto-
of Chemistry, Cambridge optic surface plasmon resonance sensing. J
2. Raether H (1988) Surface Plasmons on Appl Phys 108:054502. doi:10.1063/1.
smooth and rough surfaces and on gratings. 3475711
Springer, Berlin 12. Manera MG, Montagna G, Ferreiro-Vila E et al
3. Incorporated TI (2003) Spreeta on the sensi- (2011) Enhanced gas sensing performance of
tivity of spreeta. Solutions 2003:1–14 TiO2 functionalized magneto-optical SPR sen-
4. Sepúlveda B, Calle A, Lechuga LM, Armelles G sors. J Mater Chem 21:16049. doi:10.1039/
(2006) Highly sensitive detection of biomole- c1jm11937k
cules with the magneto-optic surface-plasmon- 13. Manera MG, Ferreiro-vila E, Cebollada A et al
resonance sensor. Opt Lett 31:1085–1087 (2012) Ethane-bridged Zn porphyrins dimers
5. Regatos D, Sepúlveda B, Fariña D et al (2011) in langmuir-Scha fer thin films: spectroscopic,
Suitable combination of noble/ferromagnetic morphologic, and magneto-optical surface
metal multilayers for enhanced magneto- plasmon resonance characterization. J Phys
plasmonic biosensing. Opt Express Chem C 116:10734–10742
19:8336–8346 14. David S, Polonschii C, Luculescu C et al
6. Armelles G, González-Dı́az J (2008) Localized (2015) Magneto-plasmonic biosensor with
surface plasmon resonance effects on the enhanced analytical response and stability. Bio-
magneto-optical activity of continuous Au/ sens Bioelectron 63:525–532. doi:10.1016/j.
Co/Au trilayers. Opt Express 16:3112–3114 bios.2014.08.004
7. Temnov VV, Armelles G, Woggon U et al 15. Johnsson B, Löfås S, Lindquist G (1991)
(2010) Active magneto-plasmonics in hybrid Immobilization of proteins to a
metal–ferromagnet structures. Nat Photonics carboxymethyldextran-modified gold surface
4:107–111. doi:10.1038/nphoton.2009.265 for biospecific interaction analysis in surface
plasmon resonance sensors. Anal Biochem
8. Belotelov VI, Kreilkamp LE, Akimov IA et al 198:268–277. doi:10.1016/0003-2697(91)
(2013) Plasmon-mediated magneto-optical 90424-R
transparency. Nat Commun 4:2128. doi:10.
1038/ncomms3128 16. Chinowsky TM, Jung LS, Yee SS (1999) Opti-
mal linear data analysis for surface plasmon
9. Zvezdin A, Kotov V (1997) Modern magne- resonance biosensors. Sensors Actuators B
tooptics and magnetooptical materials. IOP Chem 54:89–97
Publishing Ltd, Bristol
17. Zhan S, Wang X, Liu Y (2011) Fast centroid
10. Armelles G, Cebollada A, Garcı́a-Martı́n A, algorithm for determining the surface plasmon
González MU (2013) Magnetoplasmonics: resonance angle using the fixed-boundary
combining magnetic and plasmonic function- method. Meas Sci Technol 22:025201.
alities. Adv Opt Mater 1:10–35. doi:10.1002/ doi:10.1088/0957-0233/22/2/025201
adom.201200011
88 Sorin David et al.
18. David S, Gheorghiu E (2011) Towards an 22. Kahn D (1992) Magnetic properties and struc-
advanced magneto-plasmonic sensing plat- ture of cobalt hardened gold. SUR/FIN’92
form. Adv Top Electr Eng (ATEE), 2011 7th 1:305–341
International symposium 1:12–14 23. Yang K, Clavero C, Skuza J (2010) Surface
19. Gheorghiu M, David S, Polonschii C et al plasmon resonance and magneto-optical
(2014) Label free sensing platform for amyloid enhancement on Au–Co nanocomposite thin
fibrils effect on living cells. Biosens Bioelectron films. J Appl Phys 107:103924
52:89–97. doi:10.1016/j.bios.2013.08.028 24. Mattox D (2010) Handbook of physical vapor
20. Neuert G, Kufer S, Benoit M, Gaub HE deposition (PVD) processing, 2nd edn. Else-
(2005) Modular multichannel surface plasmon vier, Inc., Berlin
spectrometer. Rev Sci Instrum 76:054303. 25. Ferreiro-Vila E, Iglesias M, Paz E et al (2011)
doi:10.1063/1.1899503 Magneto-optical and magnetoplasmonic prop-
21. Polonschii C, David S, Tombelli S (2010) A erties of epitaxial and polycrystalline Au/Fe/
novel low-cost and easy to develop functiona- Au trilayers. Phys Rev B 83:205120. doi:10.
lization platform. Case study: aptamer-based 1103/PhysRevB.83.205120
detection of thrombin by surface plasmon res-
onance. Talanta 80:2157–2164
Chapter 6
Abstract
Localized surface plasmon resonance (LSPR) associated with metal nanostructures has developed into a
highly useful sensor technique. Optical LSPR spectroscopy of nanostructures often shows sharp absorption
and scattering peaks, which can be used to probe several bio-molecular interactions. Here, we report
nanoplasmonic biosensors using LSPR on nanocup arrays (nanoCA) to recognize bio-molecular binding
for biochemical detection. These sensors can be modified to quantify binding of small molecules to proteins
for odorant and explosive detections. Electrochemical LSPR biosensors can also be designed by coupling
electrochemistry and LSPR spectroscopy measurements. Multiple sensing information can be obtained and
electrochemical LSPR property can be investigated for biosensors. In some applications, the electrochemi-
cal LSPR biosensor can be used to quantify immunoreactions and enzymatic activity. The biosensors exhibit
better performance than those of conventional optical LSPR measurements. With multi-transducers, the
nanoplasmonic biosensor can provide a promising approach for bio-detection in environmental monitor-
ing, healthcare diagnostics, and food quality control.
Key words Localized surface plasmon resonance (LSPR), Nanocup arrays (nanoCA), Electrochemis-
try, Biosensor, BSA (bull serum albumin), Thrombin
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_6,
© Springer Science+Business Media LLC 2017
89
90 Diming Zhang et al.
Fig. 1 Schematic of localized surface plasmon resonance (LSPR), where the free
conduction electrons in the metal sphere are driven into oscillation due to
coupling with incident light
2 Materials
2.1 Optical and 1. Halogen cold light source (DT-MINI-2, Ocean Optics Inc.,
Electrochemical Dunedin, Florida, USA).
Measurement System 2. Spectrophotometer (USB2000+, Ocean Optics Inc., Dunedin,
Florida, USA).
3. Three fiber bundles (QP230-0.25-XSR, Ocean Optics Inc.,
Dunedin, Florida, USA).
4. Optical fiber attenuator (FVA-ADP-UV, Ocean Optics Inc.,
Dunedin, Florida, USA).
5. Collimating lens (F230APC-633, Thorlabs Inc., Newton, New
Jersey, USA).
6. Absorb sample pool (SPL-CUV-ABS, Pulei Inc., Hangzhou,
China).
7. Multi-mode microplate reader (SpectraMax Paradigm, Molec-
ular Devices Co., United Sates).
8. Electrochemical workstation (CHI 660E, CH Instruments,
Texas, USA).
92 Diming Zhang et al.
2.2 Acquisition and 1. Phosphate buffer saline (PBS) (see Note 1).
Immobilization of 2. 2-(morpholino)ethanesulfonic acid (MES) buffer (see Note 2).
Proteins
3. 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) solu-
tion (see Note 3).
4. N-hydroxy-succinimide (NHS) solution (see Note 4).
5. Carboxy-poly(ethylene)-thiol (HOOC-PEG-SH, 2 kDa,
Sigma- Aldrich, USA) (see Note 5).
6. Recombinant expressed plasmid pET-Acer-ASP2 (from Center
of Analysis & Measurement, Zhejiang University) (see Note 6).
7. E. coli BL21 (DE3) (from Center of Analysis & Measurement,
Zhejiang University).
8. BamH I endonuclease (TaKaR Co., Dalian, China).
9. Hind III endonuclease (TaKaR Co., Dalian, China).
10. Isopropyl-β-D-thiogalactopyranoside (Aladdin, Shanghai, China).
11. OBPs, Acer-ASP2.
12. TNT-specific peptides (see Note 7).
3 Methods
3.1 Fabrication of the The nanoCA device can be fabricated for use in LSPR spectroscopy
nanoCA for monitoring binding events occurring on the surface of the
device [14, 17]. The nanostructures were imprinted by nanocone
template made on quartz substrate and deposited with gold nano-
particles by following steps, as shown in Fig. 2.
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 93
3.2 Optical Detection Periodic nanostructures have prominent optical features such as
for LSPR Spectroscopy absorption and transmission peaks in the visible spectrum. These
features were elicited by plasmon resonance and can be utilized in
optical sensors. The optical detection can be performed using the
following steps.
1. As shown in Fig. 3a, the nanoCA chips were first made into
small rounds with diameters of 1 cm by hole puncher. Then the
rounds can be immobilized on the bottom of wells by transpar-
ent epoxy resin adhesive, leaving 6 h for the adhesive to solidify.
LSPR spectroscopy was performed using the microplate reader
for multi-mode detection in high throughput.
2. A normal transmission model of the microplate reader was
applied to measure the spectra of nanoCA. Light was emitted
from the light source on the bottom of wells of the plate,
delivered through nanoCA chips and received by the spectrom-
eter. The reader detected LSPR spectroscopy in individual wells
of the plate. The lamp and spectrograph can move from one
well to other wells by a mechanical driver. The scanning range
for spectroscopy was set from 300 to 900 nm and the step was
fixed at 1 nm.
3. During measurement, 10 μl sample solutions were often added
by pipette on the surface of the chip in the wells of the plate.
The small volume of the analyte formed a thin liquid layer and
reduced interference from the solution itself.
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 95
Fig. 4 Nanoplasmonic biosensor system for bio- and chemical detection. (a) Schematic diagram of the optical
detection using a 96-well plate instrument in transmission mode. (b) Transmission spectra of nanoCA in the
presence of NaCl at different concentrations (0.1%, 0.2%, 0.3%, 0.4%, and 0.5%). The resonance wavelength
shifts with increasing concentration
3.3 Acquisition of In sensing property test, the nanoCA device showed a good perfor-
Bio-sensitive Material mance for RIU change on the surface. However, the nanoCA
device was only a physical transducer sensitive to refractive index
changes, rather than a real sensor with high selectivity to special
biochemical analytes. In our work, several olfactory proteins, such
as OBPs and peptides, were used to modify nanoCA for bio- and
chemical detections.
96 Diming Zhang et al.
3.3.1 Expression and Oderant binding proteins (OBPs) are an important class of sensing
Purification of OBPs proteins in biological olfactory systems. OBPs have significant
binding affinities to various biochemical molecules. Distinct from
membrane proteins or receptors, OBPs are soluble, can be
expressed at low cost, purified easily, and maintain bioactivity
in vitro [21, 22]. Thus, OBPs are useful as biosensors and can be
obtained by the following steps:
– The recombinant OBPs of Acer-ASP2 were cloned from full-
length cDNA of adult worker bees, Apis cerana cerana. The
recombinant plasmid pET-Acer-ASP2 was expressed and trans-
formed into E. coli BL21 (DE3) competent cells after 450 bp
fragments were excised with BamH I and Hind III from the
pGEM-Acer-ASP2 plasmid.
– The cells were grown in Luria–Bertani broth (including 30 μg/
ml kanamycin) at 37 C with 1.5 mM isopropyl-β-D-thiogalac-
topyranoside to induce expression of the protein. After 5 h at
28 C, the bacterial cells were harvested and lysed by sonication
and centrifuged into crude cell extracts into pellet and superna-
tant (3 ml, 1740 g, 10 min).
– The OBP was then collected from the cells. The pellets of crude
cell extracts containing recombinant proteins were precipitated
in 1.5 M urea in ddH2O and freeze-dried.
– Finally, the protein was resuspended (500 μg/ml) in phosphate
buffered saline (PBS; pH ¼ 7.4) and stored at 4 C for biosensor
experiments. More details can be found in the previous study
about expression of the OBPs from honeybee [23].
3.3.2 Synthesis of Peptides are short protein fragments composed of a chain of amino
Peptide acids. Peptides can be designed based on known structures of
protein binding sites, synthesized with chemical methods, and
purified to obtain specific sequences. They are useful in designing
artificial receptors to mimic molecular recognition between pro-
teins and analytes. Thus, peptides are ideal candidates for biosen-
sing materials and for biosensor fabrication.
– TNT-specific peptide was chemically synthesized based on the
reported bio-sensitive sequence (WHWQRPLMPVSI) as olfac-
tory protein for TNT [24].
– The peptide (CLVPRGSC) can be cleaved by thrombin, and was
synthesized for use in thrombin detection.
– Chemical synthesis of peptides was performed by standard solid-
phase peptide synthesis (SPPS) using BOC chemistry, with step-
wise addition of protected amino acids to a growing peptide
chain. The synthesis work was performed by Genscript company
(Nanjing, China). Peptides were stored as freeze-dried powders
(see Note 8).
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 97
3.5 Olfactory Protein The interaction between small molecules and olfactory proteins
Modified Sensor Using immobilized on nanoCA surface can affect the refractive index in
LSPR Spectroscopy the metal-dielectric interface of nanocup sidewalls and thus give
rise to resonance wavelength shift in LSPR spectroscopy. The
wavelength shift can then be used to quantify small molecules
involved in binding events, and bio-modified nanoCA can be
used as a nanoplasmonic biosensor for bio- and chemical detections
[15, 16].
98 Diming Zhang et al.
3.5.1 LSPR Spectroscopy The nanoCA can be used as a biosensor by monitoring interactions
Analysis for Monitoring of between small molecules and olfactory proteins. For instance, sev-
Small Molecule Binding to eral chemical molecules can specifically bind into hydrophobic
Protein pockets of OBPs immobilized on the nanoCA surface, which can
modulate protein conformation and change relative dielectric con-
stants of the bio-coating [15] (Fig. 6a).
– The OBPs were immobilized on the surface of the nanoCA
device by self-assembly to obtain OBP-modified nanoCA for
monitoring of molecular binding (see Subheading 3.4). For
odorant measurement one kind of aromatic odorant, β-ionone,
was used as a model molecule to test responses of the bio-
nanoCA with OBPs. Ten microliter β-ionone solutions at differ-
ent concentrations ranging from 10 nM to 10 Mm can be added
in wells of the 96-well plates by pipette (see Note 10).
Fig. 6 Bio-nanoCA with olfactory proteins as nanoplasmonic biosensors for odorant and explosive detection.
(a) Molecular docking of the binding pocket of the OBPs, Acer-ASP2. (b) Transmission spectrum of OBPs-
modified nanoCA for β-ionone. (c) Dose-dependent profiles of bio-nanoCA with OBPs in resonance wavelength
shift of transmission peak for β-ionone and TNT. (d) Dose-dependent profiles of bio-nanoCA with TNT-specific
peptide for β-ionone and TNT. (mean SD, n ¼ 15)
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 99
3.5.2 Nanoplasmonic Recent studies reported that trained honeybees are able use their
Biosensor for Explosive sensing proteins to find nitro-explosives, and various OBPs have
Detection been used to modify biosensors for explosive detection [25–27].
We have explored the binding of nitro-explosive molecules to the
OBPs of AcerASP2 by the bio-nanoCA.
– Molecular docking of TNT and the OBP was carried out to
show affinity of these two molecules. Like β-ionone, TNT mole-
cules can enter protein cavities, interact with amino acid resi-
dues, and form a complex with OBPs. This process might elicit
OBP conformation changes like those in β-ionone binding,
thereby modulating the LSPR on the nanoCA in the optical
measurement.
– The OBPs were immobilized on the surface of the nanoCA
device by self-assembly to obtain OBP-modified nanoCA for
monitoring of molecular binding (see Subheading 3.4). Ten
microliter TNT solutions at different concentrations can be
added in wells of the 96-well plates by pipette. Then, optical
detection can be performed as described above (see Subheading
3.2). Figure 6c showed responses of the nanoCA with the OBPs
to TNT at increasing concentrations. The nanoCA can monitor
the binding of TNT molecules to the OBPs. However, the
responses were nonlinear in concentration range from 107 to
105 M, and the nanoCA cannot distinguish TNT and
β-ionone. It indicated that the nanoCA with OBPs was not a
good biosensor strategy for explosive detection of TNT.
– The TNT-specific peptide was immobilized on the surface of the
nanoCA device by self-assembly to obtain peptide-modified
nanoCA for monitoring of molecular binding (see Subheading
3.4). Ten microliter TNT solutions at different concentrations
can be added in wells of the 96-well plates by pipette. Then,
optical detection can be performed as described above (see Sub-
heading 3.2). As shown in Fig. 6d, the nanoCA with the pep-
tides was high selective to TNT, while the nanoCA showed no
response to β-ionone.
100 Diming Zhang et al.
3.6 Electrochemical LSPR is not only influenced by refractive index change on the
LSPR Biosensor for nanostructure surface, but also by potential and current density
Protein Analysis on the surface of a metal film [14]. Thus, LSPR sensors can be
combined with electrochemistry to implement synchronized elec-
trical and optical responses. The combination of electrochemistry
and optical SPR measurement can provide a multitude of informa-
tion from different signal transductions and provide novel
approaches to elucidate detailed processes for electrochemical
reactions.
Fig. 7 Electrochemical LSPR spectroscopy measurement. (a) Schematic of experimental apparatus for
synchronous electrochemical and optical measurement. (b) Transmission LSPR spectroscopy of nanoCA
with different surface currents. (c) Synchronous measurement of LSPR and electrochemistry in CV scanning
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 101
Fig. 9 Electrochemical LSPR detection for specific binding of proteins. (a) The transmission spectrum of
nanoCA with PBS, 100 μg/ml BSA and BSA plus synchronous CV scanning, 100 μg/ml HAS and HSA plus CV
scanning. (b) Statistic for shifts in dip wavelength of HSA, HSA plus CV, BSA and BSA plus CV (mean SD,
n ¼ 6)
3.6.3 Electrochemical The electrochemical LSPR spectroscopy can also be used in throm-
LSPR Spectroscopy as bin detection [28]. The detection can be performed by the follow-
Biosensor for Enzymatic ing steps.
Activity
– As shown in Fig. 10a, polyethylene glycol (PEG), peptide
(CLVPRGSC), and bovine serum albumin (BSA) were immo-
bilized on surface of nanoCA with the self-assembly (methods,
see Subheading 3.4), while peptides were used as probes for
thrombin.
– One milliliter thrombin solutions at different concentrations can
be added in wells by pipette. Thrombin can cleave specific sites
of peptide sequences and remove BSA from the surface of
nanoCA, thereby modulating LSPR transmission spectra.
– Spectrographic measurement was coupled with electrochemical
voltage at 3 V to dissociative BSA from the nanoCA surface and
increase LSPR wavelength shift responses. The measurement
apparatus was the same as described above (see Subheading 3.6).
104 Diming Zhang et al.
Fig. 10 Electrochemical LSPR sensor for thrombin detection. (a) Design of electrochemical LSPR sensor for
thrombin using self-immobilization of protein on nanoCA. (b) LSPR responses of wavelength shifts of the
nanoCA for thrombin detections with (+) and without () electrochemical enhancement, electrochemical
voltage (EV)
The results for thrombin detection are shown in Fig. 10b. The
presence of thrombin can selectively catalyze cleavages at Arg-Gly
bonds of peptides, remove BSA from the nanoCA sidewall, and
generate time-dependent shifts in resonance wavelength of
nanoCA (see Notes 10 and 11).
– The transmission spectrum can be analyzed by LSPR wavelength
shifts with electrochemical enhancement. As expected, as the
electrochemical voltage is increased LSPR wavelength shifts
significantly. There was stronger voltage enhancement in the
presence of thrombin at high concentrations. Indeed, the elec-
trochemically enhanced LSPR biosensor had higher sensitivity
and shorter response time than the sensor utilizing LSPR alone.
This provides a promising approach for designing more sensitive
and rapid LSPR sensors.
4 Notes
Acknowledgments
References
1. Homola J (2008) Surface plasmon resonance olfactory proteins from honeybee as biosensor
sensors for detection of chemical and biological for chemical detection. Sensor Actuat B Chem
species. Chem Rev 108:462–493 221:341–349
2. Vollmer F, Arnold S (2008) Whispering-gal- 16. Zhang D, Zhang Q, Lu Y, et al. (2016) Peptide
lery-mode biosensing: label-free detection functionalized nanoplasmonic sensor for
down to single molecules. Nat Methods explosive detection. Nano-Micro Lett 8:36–43
5:591–596 17. Gartia MR, Hsiao A, Pokhriyal A et al (2013)
3. Nair RV, Vijaya R (2010) Photonic crystal sen- Colorimetric plasmon resonance imaging using
sors: an overview. Prog Quant Electron nano lycurgus cup arrays. Adv Opt Mater
34:89–134 1:68–76
4. Saha K, Agasti SS, Kim C et al (2012) Gold 18. Stewart ME, Mack NH, Malyarchuk V et al
nanoparticles in chemical and biological sens- (2006) Quantitative multispectral biosensing
ing. Chem Rev 112:2739–2779 and 1D imaging using quasi-3D plasmonic
5. Willets KA, Van Duyne RP (2007) Localized crystals. Proc Natl Acad Sci U S A
surface plasmon resonance spectroscopy and 103:17143–17148
sensing. Annu Rev Phys Chem 58:267–297 19. Kuznetsov AI, Evlyukhin AB, Goncalves MR
6. Mayer KM, Hafner JH (2011) Localized sur- et al (2011) Laser fabrication of large-scale
face plasmon resonance sensors. Chem Rev nanoparticle arrays for sensing applications.
111:3828–3857 ACS Nano 5:4843–4849
7. Gao HW, Henzie J, Odom TW (2006) Direct 20. Kee JS, Lim SY, Perera AP et al (2013) Plas-
evidence for surface plasmon-mediated monic nanohole arrays for monitoring growth
enhanced light transmission through metallic of bacteria and antibiotic susceptibility test.
nanohole arrays. Nano Lett 6:2104–2108 Sensor Actuat B Chem 182:576–583
8. Anker JN, Hall WP, Lyandres O et al (2008) 21. Lu Y, Li H, Zhuang S et al (2014) Olfactory
Biosensing with plasmonic nanosensors. Nat biosensor using odorant-binding proteins from
Mater 7:442–453 honeybee: Ligands of floral odors and phero-
9. Stewart ME, Anderton CR, Thompson LB mones detection by electrochemical imped-
et al (2008) Nanostructured plasmonic sen- ance. Sensor Actuat B Chem 193:420–427
sors. Chem Rev 108:494–521 22. Lu Y, Yao Y, Zhang Q et al (2015) Olfactory
10. Cao J, Sun T, Grattan KT (2014) Gold biosensor for insect semiochemicals analysis by
nanorod-based localized surface plasmon reso- impedance sensing of odorant-binding pro-
nance biosensors: a review. Sens Actuators B teins on interdigitated electrodes. Biosens
195:332–351 Bioelectron 67:662–669
11. Cheng XR, Wallace GQ, Lagugné-Labarthet F 23. Li H-L, Zhang Y-L, Gao Q-K et al (2008)
et al (2015) Au nanostructured surfaces for Molecular identification of cDNA, immunolo-
electrochemical and localized surface plasmon calization, and expression of a putative
resonance-based monitoring of α-synuclein–s- odorant-binding protein from an Asian honey
mall molecule interactions. ACS Appl Mater bee, Apis cerana cerana. J Chem Ecol
Interfaces 7:4081–4088 34:1593–1601
12. Boltasseva A (2009) Plasmonic components 24. Jaworski JW, Raorane D, Huh JH et al (2008)
fabrication via nanoimprint. J Opt A Pure Evolutionary screening of biomimetic coatings
Appl Opt 11:114001 for selective detection of explosives. Langmuir
24:4938–4943
13. Pimpin A, Srituravanich W (2011) Review on
micro-and nanolithography techniques and 25. Smith RG, D’Souza N, Nicklin S (2008) A
their applications. Eng J 16:37–56 review of biosensors and biologically-inspired
systems for explosives detection. Analyst
14. Zhang D, Lu Y, Jiang J et al (2015) Nanoplas- 133:571–584
monic biosensor: coupling electrochemistry to
localized surface plasmon resonance spectros- 26. Ramoni R, Bellucci S, Grycznyski I et al (2007)
copy on nanocup arrays. Biosens Bioelectron The protein scaffold of the lipocalin odorant-
67:237–242 binding protein is suitable for the design of
new biosensors for the detection of explosive
15. Zhang D, Lu Y, Zhang Q et al (2015) Nano- components. J Phys Condens Matter
plasmonic monitoring of odorants binding to 19:395012
Nanoplasmonic Biosensor Using Localized Surface Plasmon Resonance. . . 107
Abstract
Biosensor for the detection of virus was developed by utilizing plasmonic peak shift phenomenon of the
gold nanoparticles and viral infection mechanism of hemagglutinin on virus and sialic acid on animal cells.
The plasmonic peak of the colloidal gold nanoparticles changes with the aggregation of the particles due to
the plasmonic interaction between nearby particles and the color of the colloidal nanoparticle solution
changes from wine red to purple. Sialic acid reduced and stabilized colloidal gold nanoparticle aggregation
is induced by the addition of viral particles in the solution due to the hemagglutinin-sialic acid interaction.
In this work, sialic acid reduced and stabilized gold nanoparticles (d ¼ 20.1 1.8 nm) were synthesized by a
simple one-pot, green method without chemically modifying sialic acid. The gold nanoparticles showed
target-specific aggregation with viral particles via hemagglutinin-sialic acid binding. A linear correlation was
observed between the change in optical density and dilution of chemically inactivated influenza B virus
species. The detection limit of the virus dilution (hemagglutinination assay titer, 512) was shown to be
0.156 vol% and the upper limit of the linearity can be extended with the use of more sialic acid-gold
nanoparticles.
Key words Biosensor, Sialic acid, Gold nanoparticle, Viral detection, Colorimetric measurement
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_7,
© Springer Science+Business Media LLC 2017
109
110 Changwon Lee et al.
Fig. 1 Schematic illustration of surface plasmon resonance of gold nanoparticles due to collective oscillation
of surface electrons by incident light of appropriate wavelengths
Fig. 2 Detection scheme of viral particles based on the aggregation of SA-AuNPs on the virus surface through
sialic acid-hemagglutinin binding
2 Materials
2.2 Sialic Acid and 1. 1.25 mM N-acetylneuraminic acid (SA) (Catalogue No. A2388,
Gelatin Gold Sigma-Aldrich, St. Louis, MO) was prepared in DI water.
Nanoparticle Synthesis 2. 0.1 wt% gelatin (Catalogue No. 53028, Sigma-Aldrich, St.
Louis, MO) was prepared in DI water.
3. 1 M sodium hydroxide (Catalogue No. S5881, Sigma-Aldrich,
St. Louis, MO) was prepared in DI water.
4. 0.02 M chloroauric acid (HAuCl4) (Catalogue No. 254169,
Sigma-Aldrich, St. Louis, MO) was prepared in DI water.
112 Changwon Lee et al.
2.3 Influenza Virus 1. Dulbecco’s Modified Eagle Media (DMEM) (Catalogue No.
Deactivation 11965, Life Technology, Grand Island, NY) was used for the
culture of virus.
2. Fetal Bovine Serum (FBS) (Catalogue No. 16000-044, Life
Technology, Grand Island, NY) was added to 10% in DMEM.
3. β-propiolactone (Catalogue No. P5648, Sigma-Aldrich, St.
Louis, MO) was added to 0.05% of final concentration in the
viral solution.
3 Methods
3.1 Sialic Acid Gold 1. Ten milliliter of 1.25 mM sialic acid (SA) solution was prepared
Nanoparticle in DI water at room temperature. The SA solution was mixed
(SA-AuNP) Synthesis with 250 μL of 0.02 M HAuCl4 followed by the addition of
50 μL 1 M NaOH in 20 mL glass vial (see Note 1).
2. The mixture was then stirred and heated for 80 C at 20 g
for 15 min on a heating magnetic stirrer. The color of the
solution changed from yellow to a dark red wine.
3. After the solution was cooled to room temperature, the gold
nanoparticles were washed twice by centrifugation at
4500 g) using a bench-top centrifuge (Eppendorf 5424)
for 20 min. After the centrifugation, supernatant was removed
and the dark red pellet was suspended in DI water and stored
until further use.
3.2 Influenza Virus 1. Virus solution was provided in 10% FBS containing DMEM
Inactivation and it was inactivated by the addition of β-propiolactone (final
concentration of 0.05%) for 1 h at 37 C.
2. Inactivated virus was stored at 20 C until further use.
3.3 Particle Size Particle size analysis was performed on a Nanotrac particle size
Analysis analyzer (Microtrac) with a built-in liquid sample holder. The
concentration of the sample was adjusted for the optimal measure-
ment condition. Three 30-s measurements were averaged for parti-
cle size determination, with results shown in Fig. 3c.
A 0.8 B 0.25
0.7
0.2
0.6
0.5 Increase
0.15
over time
OD
0.4
OD
0.3 0.1
0.2
0.05
0.1
0 0
400 500 600 700 800 0 5 10 15
Wavelength (nm) Time (min)
C D100 20
100 50
90 90
80 40 80
70 70
%Passing
%Channel
%Channel
%Passing
60 30 60
50 50 10
40 20 40
30 30
20 10 20
10 10
0 0 0 0
0.1 1 10 100 1,000 10,000 0.000 0.001 0.01 0.1 1 10
Size (Nanometers) Size (Microns)
Fig. 3 Typical results of the UV-Vis spectroscopic results and particle size analysis results of SA-AuNP before
and after virus incubation. UV-Vis absorption spectra measured at 0, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and 12 min
after the addition of 1.25 vol% influenza B/Victoria. (a) Typical UV-Vis spectral change of SA-AuNP solution after
the addition of the virus, (b) change of OD610 over time, (c) particle size analysis result measured before the virus
addition, and (d) particle size analysis result measured after the addition of 1.25 vol% influenza B/Victoria
3.5 UV-Vis Five hundred microliter of the solution containing SA-AuNP with or
Measurements without virus was added into the 1 cm path length quartz cuvette
and placed in the cuvette holder of the UV-Vis spectrometer (Ocean
Optics USB 4000 UV-Vis spectrometer). Then the UV-Vis spec-
trum was measured immediately. Results are shown in Fig. 3a, b.
3.6 Particle Size Particle size of the SA-AuNP with virus was measured in the same
Analysis way as described in Subheading 3.3. In short, the solution contain-
Measurements of SA- ing virus incubated SA-AuNP was placed in the built-in liquid sample
AuNP with Virus holder of the particle size analyzer (Microtrac). The sample concen-
tration was adjusted for the optimum result, as shown in Fig. 3d.
3.7 Colorimetric 1. Five hundred microliter of SA-AuNP solution was mixed with
Detection of Viral different dilutions of influenza virus solution and immediately
Particles added to a 1-cm path length quartz cuvette.
114 Changwon Lee et al.
OD610
OD610
2 min
0.4 0.1
3 min
0.4 0.05
4 min 0.00
0.0
0 10 20 30 5 min 0 1 2 3 4 5
0.3 Time (min) 6 min 0.3 Time (min)
OD
7 min
OD
8 min 0 min
0.2 0.2 1 min
2 min
3 min
Increase
0.1 0.1 Increase 4 min
over time 5 min
over time
0.0 0.0
300 400 500 600 700 800 300 400 500 600 700 800
Wavelength (nm) Wavelength (nm)
0.20
c d
0.15
delta (OD610 )
0.10 y=0.104x+0.0003
2
R =0.9915
0.05
0.00
0 2 4 6
Virus (v%)
Fig. 4 (a) Absorption spectra of 0.4 nM SA-AuNPs over time after the addition of 2.5 vol% influenza B/Victoria
solution; inset, OD610 value change. (b) Absorption spectra of 0.4 nM SA-AuNPs over time after the addition of
0.156 vol% virus solution; inset, change in OD610. (c) Change in OD610 value after reaching plateau for
different dilutions of virus. A linear correlation is observed for dilutions <1.25 vol% virus. (d) Photograph of
SA-AuNPs with different dilutions of virus, 5, 2.5, 1.25, 0.625, 0.3125, 0.156, and 0 vol% from left to right,
respectively
Plasmonics-Based Detection of Virus Using Sialic Acid Functionalized Gold Nanoparticles 115
0.30 0.20
A B
0.25
0.15
delta (OD610)
0.20
delta (OD610)
0.15 0.10
y=0.0957x+0.0003
y=0.0974x+0.0183 2
0.10 2 R =0.9970
R =0.9999
0.05
0.05
0.00 0.00
0 2 4 6 0 2 4 6
Virus (v%) Virus (v%)
Fig. 5 (a) Change in OD610 measured for 0.8 nM SA-AuNP solution after the addition of influenza B/Victoria. A
linear correlation is observed for dilutions <2.5 vol% virus. (b) Change in OD610 measured for 0.4 nM SA-AuNP
solution after the addition of influenza B/Yamagata. A linear correlation is observed for dilutions <1.25 vol%
virus
3.8 Increasing the To increase the linear range of ΔOD vs. concentration of added
Linear Dynamic Range virus solution, we used a higher concentration of SA-AuNP solu-
of Virus Measurements tion (0.8 nM) to conduct the similar tests, with results shown in
Fig. 5a. It was observed that the linear range of ΔOD vs. concen-
tration of added virus solution was extended to 2.5 vol% of virus in
the solution. These results illustrate the importance of using a large
amount of SA-AuNPs in the solution for virus detection when
applying this scheme. We note that this method can also detect
another virus subtype, influenza B/Yamagata. Results obtained
from the influenza B/Yamagata solutions are similar to those of
influenza B/Victoria and shown in Fig. 5b.
In conclusion, we demonstrate a simple method to synthesize
gold nanoparticles using sialic acid as both the reducing agent and
the stabilizing agent. Experimental results support the hypothesis
that sialic acid molecules on SA-AuNP surface interact with viral
envelope protein hemagglutinin, causing a colorimetric change of
the SA-AuNP solution. The resulting SA-AuNPs can readily detect
influenza B virus (HA titer of 512) diluted to 0.156 vol%. The
method is effective for different influenza B lineages (Victoria and
Yamagata) .
4 Notes
Acknowledgment
References
1. Zhong Z, Patskovskyy S, Bouvrette P, Luong properties of gold nanoparticles. J Am Chem
JHT, Gedanken A (2004) The surface chemis- Soc 130:8038–8043
try of Au colloids and their interactions with 10. Lee C, Gaston MA, Weiss AA, Zhang P (2013)
functional amino acids. J Phys Chem B Colorimetric viral detection based on sialic acid
108:4046–4052 stabilized gold nanoparticles. Biosens Bioelec-
2. Link S, El-Sayed MA (1999) Size and temper- tron 42:236–241
ature dependence of the plasmon absorption of 11. Sauter NK, Hanson JE, Glick GD, Brown JH,
colloidal gold nanoparticles. J Phys Chem B Crowther RL, Park SJ, Skehel JJ, Wiley DC
103:4212–4217 (1992) Binding of influenza virus hemaggluti-
3. Kreibig U, Genzel L (1985) Optical absorption nin to analogs of its cell-surface receptor, sialic
of small metallic particles. Surf Sci acid: analysis by proton nuclear magnetic reso-
156:678–700 nance spectroscopy and X-ray crystallography.
4. Elghanian R, Storhoff JJ, Mucic RC, Letsinger Biochemistry 31:9609–9621
RL, Mirkin CA (1997) Selective colorimetric 12. Varghese JN, Colman PM, van Donkelaar A,
detection of polynucleotides based on the Blick TJ, Sahasrabudhe A, McKimm-Breschkin
distance-dependent optical properties of gold JL (1997) Structural evidence for a second
nanoparticles. Science 277:1078–1081 sialic acid binding site in avian influenza virus
5. Storhoff JJ, Marla SS, Bao P, Hagenow S, neuraminidases. Proc Natl Acad Sci U S A
Mehta H, Lucas A, Garimella V, Patno T, 94:11808–11812
Buckingham W, Cork W, M€ uller UR (2004) 13. Weis W, Brown JH, Cusack S, Paulson JC,
Gold nanoparticle-based detection of genomic Skehel JJ, Wiley DC (1988) Structure of the
DNA targets on microarrays using a novel opti- influenza virus haemagglutinin complexed
cal detection system. Biosens Bioelectron with its receptor, sialic acid. Nature
19:875–883 333:426–431
6. Thanh NTK, Rosenzweig Z (2002) Develop- 14. Suzuki T, Portner A, Scroggs RA, Uchikawa
ment of an aggregation-based immunoassay for M, Koyama N, Matsuo K, Suzuki Y, Takimoto
anti-protein a using gold nanoparticles. Anal T (2001) Receptor specificities of human
Chem 74:1624–1628 respiroviruses. J Virol 75:4604–4613
7. Pavlov V, Xiao Y, Shlyahovsky B, Willner I 15. Vlasak R, Luytjes W, Spaan W, Palese P (1988)
(2004) Aptamer-functionalized Au nanoparti- Human and bovine coronaviruses recognize
cles for the amplified optical detection of sialic acid-containing receptors similar to
thrombin. J Am Chem Soc 126:11768–11769 those of influenza C viruses. Proc Natl Acad
8. Medley CD, Smith JE, Tang Z, Wu Y, Bam- Sci U S A 85:4526–4529
rungsap S, Tan W (2008) Gold nanoparticle- 16. Kunkel F, Herrler G (1993) Structural and
based colorimetric assay for the direct detec- functional analysis of the surface protein of
tion of cancerous cells. Anal Chem human coronavirus OC43. Virology
80:1067–1072 195:195–202
9. Darbha GK, Singh AK, Rai US, Yu E, Yu H, 17. Uncapher CR, DeWitt CM, Colonno RJ
Chandra Ray P (2008) Selective detection of (1991) The major and minor group receptor
mercury (II) ion using nonlinear optical families contain all but one human rhinovirus
serotype. Virology 180:814–817
Chapter 8
Abstract
Two-dimensional surface plasmon resonance (2D-SPR) imaging, which provides a real-time, sensitive, and
high-throughput analysis of surface events in a two dimensional manner, is a valuable tool for studying
biomolecular interactions and biochemical reactions without using any tag labels. The sensing principle of
2D-SPR includes angular, wavelength, and phase interrogation. In this chapter, the 2D-SPR imaging
technique is applied for sensing a target microRNA by its corresponding oligonucleotide probes, with
sequence complementarity, immobilized on the gold SPR sensing surface. However, the low SPR signal due
to intrinsic properties such as low molecular weight and quantity (pico-nanomolar) of the microRNA in
clinical samples limits the direct detection of microRNA. Therefore, we developed a biosensing technique
known as MARS (MicroRNA-RNase-SPR) assay, which utilizes RNase H to digest the microRNA probes
enzymatically for fast signal amplification, i.e., in order to increase both the SPR signal and readout speed
without the need for pre-amplification of target cDNA by polymerase chain reaction (PCR). Practically, we
targeted microRNA hsa-miR-29a-3p, whose signature correlates to influenza infection, for rapid screening
of influenza A (H1N1) patients from throat swab samples.
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_8,
© Springer Science+Business Media LLC 2017
117
118 Ho Pui Ho et al.
A B
Reflected light
Intensity
Sample medium
Angle /
Prism C Wavelength
Phase
Monochromatic or Resonance slope
broadband light source Reflected light
Angle /
Wavelength
Fig. 1 Angular, wavelength, and phase interrogation schemes are illustrated. SPR sensing surface exhibits the
SPR phenomenon under optical wave using monochromatic or broadband light source. The SPR phenomenon
is observed from the analysis of reflected light (a). In angular interrogation, incident angle of monochromatic
light (fixed wavelength) is scanned to identify the absorption dip of the reflected light. In wavelength
interrogation the spectrum of incident light at a fixed angle results in an absorption dip at a particular
wavelength of the spectral reflected light (b). In phase interrogation, SPR signal extraction of reflected light
search for maximum resonance slope, which corresponds to absorption dip in above two schemes (c). Any
bimolecular event on the sensing surface will lead to a shift in the absorption dip, and analysis of the shift in
absorption dip generates the SPR signal readout
5⬘ 3⬘5⬘ cDNA
5⬘ 3⬘ A B
3⬘ C
Mature
Stem-looped
microRNA
primer Intact
hsa-miR-29a-3p D
microRNA
probe
Streptavidin RNase H
addition Digested
MicroRNA microRNA
probes probe
D RNase H
action
Fig. 2 Workflow for microRNA sensing in the MARS assay. First, mature microRNA is converted into cDNA by
stem-loop primers (top, A: Stem-loop primer hybridization; B: Reverse transcription; C: Heat separation/
denature; D: cDNA addition for sensing). On the SPR gold sensing surface (bottom), biotinylated-microRNA
probes with spacer C6 are immobilized through thio-linkage. Unbound region on the sensing surface is
covered with the blocker PEG 1000-SH. Streptavidins are then attached to the immobilized probes through the
biotin-avidin interaction. Target cDNAs with complementary nucleotide sequence are hybridized to the probes
to generate RNA–cDNA hybrids that increase the SPR signal. RNase H is added to digest the RNA probes and to
release the hybridized cDNAs from the RNA–cDNA hybrids, The released cDNAs then binds to the new probes
for further RNase H digestion, leading to the repeated cycle of RNase H reaction for signal amplification as
indicated by forward and backward semicircular arrows. Depletion of the RNA probes on the sensing surface
causes a decrease in SPR signal. Reprinted with permission from ref. 8
MicroRNA-RNase-SPR Biosensing 121
2 Materials
3 Methods
3.1 2D SPR Imager 1. SPRchip (GWC Technologies) from the sealed package is
Setup and Detection freshly opened, rinsed with RNase-free distilled water three
Workflow times, and then air dried before use.
2. MicroRNA probes (final concentration 1 μM, 0.2 μL each) are
blotted on the designated position of the SPR gold sensing
plate (optional with an injector) and incubated at 4 C for 16 h
(see Note 4).
3. Uncoated site of the SPR gold sensing plate is blocked with
PEG 1000-SH (final concentration 1 mM) at 25 C for 2 h.
MicroRNA-RNase-SPR Biosensing 123
4. The prism is cleaned with absolute ethanol three times and then
air dried. Pump tubing and connection tubing are pre-rinsed
with RNase AWAY (Life Technologies) decontamination solu-
tion and then with RNase- and DNase-free distilled water (Life
Technologies) at 200 μL/min before use.
5. The gold sensing plate is inserted with the prism with refractive
index matching oil and placed into the SPR machine for SPR
signal detection.
6. The external pump is connected to SPR machine, sensing
surface is rinsed with SPR working buffer with a flow rate of
200 μL/min for 10 min (see Note 5).
7. Streptavidin working solution is added at 200 μL/min for
3 min.
8. The sensing surface is rinsed with SPR working buffer at
50 μL/min until a steady baseline with intensity fluctuation
less than 0.01 arbitrary unit is obtained.
9. SPR intensity reading is set to zero.
10. Sample cDNA or artificial ssDNA is added at 50 μL/min for
10 min.
11. Biosensing surface is rinsed with SPR working buffer at 50 μL/
min for 10 min.
12. RNase H working solution is added at 50 μL/min for 15 min.
13. Biosensing surface is rinsed with SPR working buffer at 50 μL/
min until a steady baseline is obtained.
3.2 SPR Sensorgram Specificity is important for this biomedical application since we aim
and Detection to apply it for rapid screening of multiple microRNA signatures.
Specificity This relies on the ability of the recognition element microRNA
probe to specifically capture the target, as well as the specificity of
RNase H to digest the probe for signal output. To evaluate this
specificity, microRNA probes of hsa-miR-29a-3p and hsa-miR-
181-5p, and single-stranded DNA (ssDNA) probe for miR-29a-
3p 3p, are immobilized on different regions of the sensing array, as
illustrated in Fig. 3. cDNAs of hsa-miR-29a-3p and hsa-miR-181-
5p, RNase H are added subsequently at the time points indicated in
the sensorgram of Fig. 3. There is a small increase in signal of
microRNA probe and ssDNA hsa-miR-29a-3p only after the injec-
tion of cDNA of miR-29a-3p, indicating the hybridization between
the microRNA probe and target cDNA occurs with high specificity.
A large decrease in signal of microRNA probe hsa-miR-29a-3p after
RNase H addition indicates that RNase H acts only on the RNA
probes from the RNA–DNA hybrid due to the interaction between
cDNA and microRNA probe, but not DNA–DNA hybrid, due to
the interaction between cDNA and ssDNA probe. Subsequent
injections of cDNA of hsa-miR-181-5p and RNase H cause a
124 Ho Pui Ho et al.
1
0.5
0
Intensity (Arbitrary Unit)
−0.5
−1
−1.5
−2
−2.5
−3
−3.5
0 1000 2000 3000 4000 5000 6000
Time (second)
Fig. 3 Specificity of microRNA probe on target cDNA detection. The microRNA probes hsa-miR-29a-3p (blue),
hsa-miR-181-5p (green), ssDNA probe miR-29a-3p (red), or PEG 1000-SH blocker only (purple) are immo-
bilized on the surface of a SPR chip as shown in the upper panel. The cDNA of hsa-miR-29a-3p and hsa-miR-
181-5p (100 nM) and RNase H (60 U/mL) are added at the time points indicated in the lower panel. The signal
change is compared with the blank control (purple) to reveal the specificity for probe–target binding and
RNase H action on RNA–cDNA and ssDNA–cDNA hybrids. Reprinted with permission from ref. 8
0.1
Buf fer
0
Intensity (Arbitrary Unit)
−0.1
−0.2 H1N1
−0.3 10 nM ssDNA
−0.4
−0.5 Healthy
−0.6
0 100 200 300 400 500 600 700 800 900
Time (second)
Fig. 4 Screening throat swab samples by MARS assay. The cDNA from healthy control and H1N1-infected
samples are added to the SPR sensing surface for hsa-miR-29a-3p detection. Synthesized ssDNA (final
concentration at 10 nM) and buffer are also used in the MRSA assay as the standard and reference. The
concentration of hsa-miR-29a-3p in a patient sample could be determined from the standard curve of known
cDNA concentration of hsa-miR-29a-3p
4 Notes
pH
−1
−1.5
***
Fig. 5 pH effect on RNase H activity in MARS assay. The RNase H activity under
different conditions is measured in the MARS assay by detecting the SPR
intensity signal change after addition of the cDNA of hsa-miR-29a-3p
(100 nM) on sensing surface with immobilized microRNA probes. SPR signal
change is recorded under different pH values in SPR working buffer. Results are
mean SEM from three independent experiments and analyzed by Student’s t-
test, where ***p-value <0.001
Acknowledgment
Week of
incubation
0, 1, 2
3
4
5
No probe
control
Time (second)
Intensity (Arbitrary Unit)
Week of
incubation 0 1 2 3 4 5 No probe
control
Fig. 6 Stability of the microRNA probes on SPR surface. The microRNA probes immobilized on the SPR gold
sensing surface are stored at 4 C for the times indicated and subjected to SPR detection on target cDNA
hybridization, which increase intensity of the SPR signal. Results in the upper panel are illustrated as bar
charts in the lower panel, with mean SEM from three independent experiments, where *p<0.05 compared
to SPR intensity signal at 0 weeks of incubation. Reprinted with permission from ref. 8
References
1. Kabashin AV, Patskovsky S, Grigorenko AN plasmon resonance differential phase imaging.
(2009) Phase and amplitude sensitivities in sur- Biosens Bioelectron 24:606–612
face plasmon resonance bio and chemical sens- 6. Tambyah PA, Sepramaniam S, Ali JM et al
ing. Opt Express 17:21191–21204 (2013) MicroRNAs in circulation are altered in
2. Wong CL, Chen GCK, Ng BK et al (2011) response to influenza A virus infection in
Multiplex spectral surface plasmon resonance humans. PLoS One 8:e76811
imaging (SPRI) sensor based on the polarization 7. Loo JFC, Wang SS, Peng F et al (2015) A non-
control scheme. Opt Express 19:18965–18978 PCR SPR platform using RNase H to detect
3. Shao Y, Li Y, Gu D et al (2013) Wavelength- MicroRNA 29a-3p from throat swabs. Analyst
multiplexing phase-sensitive surface plasmon 140:4566–4575
imaging sensor. Opt Lett 38:1370–1372 8. Peng F, He H, Loo JFC et al (2016) Identifica-
4. Wong CL, Ho HP, Suen YK et al (2007) Two tion of microRNAs in throat swab as the bio-
dimensional biosensor arrays based on SPR markers for diagnosis of influenza. Int J Med Sci
phase imaging. Appl Optics 46:2325–2332 13:77–84
5. Wong CL, Ho HP, Suen YK et al (2008) Real-
time protein biosensor arrays based on surface
Chapter 9
Abstract
Gold nanorod (GNR) based label-free sensing has been attractive due to its unique property of localized
surface plasmon resonance (LSPR). Compared to bulk gold, the SPR of GNRs is more sensitive to the
refractive index change caused by biological binding in the close proximity. Numerous studies have
reported biological detection in solution based GNR probes. However, the biosensing has the intrinsic
problems of fluctuating readings and short storage time due to nanoparticle aggregation. In contrast, a
chip-based nanorod biosensor is a more robust and reliable platform. We have developed a nanoplasmonic
biosensor in a chip format by immobilizing functionalized GNRs on a (3-mercaptopropyl)trimethoxysilane
modified glass substrate. The covalent Au-S bond ensures a strong GNR deposition on the substrate. This
biochip exhibits a high sensitivity and stability when exposed to physiological buffer with high ionic
strength. Another advantage of GNR as optical transducer is its LSPR peak dependence on the aspect
ratio, which provides an ideal multiplexed detection mechanism. GNRs of different sizes that exhibit
distinct SPR peaks are combined and deposited on designated spots of a glass substrate. The spectral shift
of the respective peaks upon the biological binding are monitored for simultaneous detection of specific
analytes. Coupled with a microplate reader, this spatially resolved GNR array biochip results in a high-
throughput assay of samples as well as multiplexed detection in each sample. Since most biological
molecules such as antibodies and DNA can be linked to GNR using previously reported surface chemistry
protocol, the label-free nanosensor demonstrated here is an effective tool for protein/DNA array analysis,
especially for detection of disease biomarkers.
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_9,
© Springer Science+Business Media LLC 2017
129
130 Zhong Mei et al.
Fig. 1 (a) UV–Vis spectra of synthesized GNRs; (b) TEM images of GNRs. The longitudinal plasmon peak is
tunable by varying rod aspect ratio
Fig. 2 Schematic of GNR biofunctionalization and design of label-free, multiplexed biosensing by GNRs of
different aspect ratios. (a) Thiolation of antibody, followed by GNR functionalization. (b) Fabrication of
multiplex GNR biochip. (c) Optical detection
sensor. When the antigens (human IgG and rabbit IgG) are present
in the test samples, they will specifically bind to their respective
antibodies (i.e., anti-human IgG and anti-rabbit IgG). Upon this
binding, pronounced red shifts at the respective dominate LSPR
bands are monitored by UV–Vis absorbance measurements. The
magnitude of the red shift in the distinct LSPR peak wavelength is
directly proportional to the amount of antigen binding onto the
GNR probes.
2 Materials
Table 1
Growth conditions for GNRs with respective longitudinal SPR wavelengths
3.1.3 Antigen Detection 1. Position the GNR-modified glass chip into a UV cuvette
(2.5 mL, 12.5 12.5 45 mm; BrandTech Scientific,
Wertheim, Germany) and insert the cuvette to the cuvette
holder of the Beckman-Coulter UV-NIR spectrophotometer,
as shown in Fig. 3. Measure the UV–Vis absorption spectra of
GNRs on glass as the baseline.
136 Zhong Mei et al.
Fig. 4 Detection of human IgG targets by anti-human IgG modified GNRs biochip. The black curve is the
absorption spectrum of anti-IgG modified GNRs biochip (baseline). The other six colored curves are absorp-
tions spectra of the GNR/anti-IgG chip incubated in the target human IgG samples of varying concentrations
(a–f: 10–60 nM). The peak wavelength at ~840 nm shows a red shift after IgG binding to the GNR/anti-IgG
chip. A higher concentration of IgG resulted in a larger red shift. Calibration curve of the red-shift versus IgG
concentration shows the longitudinal plasmon shift as a function of human IgG concentration
3.2 Nanoarray Based The methods for gold nanorod synthesis and functionlization are
Multiplexed similar as described above. To achieve multiplexed biosensing,
Biosensing for Multiple GNRs with varying sizes are mixed and deposited on one substrate.
Antigens Here, we take two-antibody detection for example.
1. GNRs with a LSPR wavelength at 840 nm are functionalized
with anti-human IgG, while GNRs with a LSPR at 630 nm are
functionalized with anti-rabbit IgG.
2. Microscopy glass slides (300 100 1.0 mm; Fisher Scientific,
Pittsburgh, PA) are treated with Piranha and MPTMS solution
as described above.
3. 10 μL of antibody-modified GNRs with mixed sizes are
dropped onto designated spots on the glass slides and incu-
bated for 2 h. The detection spots are arranged to mimic the
layout of a 96 well plate to accommodate an absorption reading
by the BioTek plate reader in a high-throughput fashion
(Fig. 5).
4. Wash the substrate with water three times and dry it with
nitrogen gas.
5. To perform a multiplexed detection, a sample solution (10 μL)
of mixed human IgG and rabbit IgG are dropped onto the
nanosensing spots and incubated for 30 min until equilibrium.
6. The glass substrate is attached to the bottom of a 96-well plate
for absorbance measurement. The UV–Vis absorbance mode is
chosen with a scanning range from 400 to 999 nm. As the light
source scans the spots from well to well, the corresponding
UV–Vis spectra are displayed in the same layout and ready for
analysis. Figure 6 is the result of multiplexed biosensing of
human and rabbit IgG samples at various concentrations
(Note 7).
4 Notes
Fig. 5 Representative pictures of (a) antibody functionalized GNR array on glass substrates which is expanded
to a high-throughput biosensing chip. Each dark spot, containing two sizes of functionalized GNRs, works as
an individual sensing platform for detecting two antigens simultaneously in one sample. Thus, this biochip can
detect 12 samples at one time; (b) Reverse side and (c) front side of attaching the chip to the bottom of a 96-
well plate frame for accommodating the reading on microplate reader (d)
Fig. 6 Multiplexed biosensing of human and rabbit IgG samples at various concentrations. (a) UV–Vis spectra
of the GNR biochip before and after detection of six various concentrations of mixed IgG. The shorter
wavelength around 630 nm is dedicated for rabbit IgG assay, while the longer one around 840 nm for
human IgG assay; (b) Plotting curve (black) of wavelength shift around 630 nm versus the concentration of
rabbit IgG; (c) Plotting curve (black) wavelength shift around 840 nm versus the concentration of human IgG.
Both curves suggest that the nanorod chip shows a linear response to the analyte (rabbit or human IgG) in the
range from 10 to 60 nM. For comparison, the calibration curves of single GNR based biochip mentioned in
Subheading 3.1 are also included (red). Interestingly, the multiplexed biosensor has an increased sensitivity
than single biosensor for human IgG detection
Fig. 7 Local electromagnetic field simulation of a vertical GNR array. (a) model of GNR array; (b) local field of
side view in the cross section along the y–z plane; (c) local field of the top view along the z direction. The
rainbow bar shows the intensity. A redder color indicates a more intensified local field
nanorod is much stronger than that on the side facets. And the
CTAB density of tips is lower than the sides, which ensures a
less steric hindrance for the tip modification [18]. For the
future perspective, controlling the orientation of the GNR
array assembly on the substrate is the current focus. For
instance, if GNRs are orderly organized to form a vertically
standing array, a large amount of homogeneous hotspots will
be created at the GNR surface due to the plasmon coupling
effect. Figure 7 is the local electromagnetic field simulation of a
vertical nanoarray consisting of seven GNRs using COMSOL
5.0. The incident light with an intensity of 1.0 108 V/m
propagates along z-direction, passing through the GNR array.
The simulation results show that the GNR array exhibit highly
uniform and enhanced local field on the tips. This unique
property is desired for biomedical applications such as nanor-
eactor and ultrasensitive SERS sensor [19].
Gold Nanorod Array Biochip for Label-Free, Multiplexed Biological Detection 141
References
1. Giri S, Trewyn BG, Lin VS (2007) Mesoporous nonspecific tumor marker, based on surface
silica nanomaterial-based biotechnological and plasmon resonance. Biosens Bioelectron 19
biomedical delivery systems. Nanomedicine (9):999–1005
(Lond) 2(1):99–111 12. Sim HR, Wark AW, Lee HJ (2010) Attomolar
2. Lee SH, Sung JH, Park TH (2012) detection of protein biomarkers using biofunc-
Nanomaterial-based biosensor as an emerging tionalized gold nanorods with surface plasmon
tool for biomedical applications. Ann Biomed resonance. Analyst 135(10):2528–2532
Eng 40(6):1384–1397 13. Ye X et al (2013) Using binary surfactant mix-
3. Oyelere AK et al (2007) Peptide-conjugated tures to simultaneously improve the dimen-
gold nanorods for nuclear targeting. Bioconjug sional tunability and monodispersity in the
Chem 18(5):1490–1497 seeded growth of gold nanorods. Nano Lett
4. Chylek P (1986) Absorption and scattering of 13(2):765–771
light by small particles. Appl Opt 25(18):3166 14. Gulati A, Liao H, Hafner JH (2006) Monitor-
5. Bennett HS, Rosasco GJ (1978) Resonances in ing gold nanorod synthesis by localized surface
the efficiency factors for absorption: Mie scat- plasmon resonance. J Phys Chem B 110
tering theory. Appl Optics 17(4):491–493 (45):22323–22327
6. Upadhyayula VK (2012) Functionalized gold 15. Wang Y, Tang L (2015) Multiplexed gold
nanoparticle supported sensory mechanisms nanorod array biochip for multi-sample analy-
applied in detection of chemical and biological sis. Biosens Bioelectron 67:18–24
threat agents: a review. Anal Chim Acta 16. Wang X et al (2015) Gold nanorod biochip
715:1–18 functionalization by antibody thiolation.
7. Yu L, Andriola A (2010) Quantitative gold Talanta 136:1–8
nanoparticle analysis methods: a review. 17. Wang Y, Tang L (2013) Chemisorption assem-
Talanta 82(3):869–875 bly of Au nanorods on mercaptosilanized glass
8. Dreaden EC et al (2012) The golden age: gold substrate for label-free nanoplasmon biochip.
nanoparticles for biomedicine. Chem Soc Rev Anal Chim Acta 796:122–129
41(7):2740–2779 18. Orendorff CJ, Murphy CJ (2006) Quantita-
9. Anker JN et al (2008) Biosensing with plasmo- tion of metal content in the silver-assisted
nic nanosensors. Nat Mater 7(6):442–453 growth of gold nanorods. J Phys Chem B 110
10. Jain PK et al (2006) Calculated absorption and (9):3990–3994
scattering properties of gold nanoparticles of 19. Lim DK et al (2011) Highly uniform and
different size, shape, and composition: applica- reproducible surface-enhanced Raman scatter-
tions in biological imaging and biomedicine. J ing from DNA-tailorable nanoparticles with 1-
Phys Chem B 110(14):7238–7248 nm interior gap. Nat Nanotechnol 6
11. Chou SF et al (2004) Development of an (7):452–460
immunosensor for human ferritin, a
Chapter 10
Abstract
The invasion of cancer cells through their surrounding extracellular matrices is the first critical step to
metastasis, a devastating event to cancer patients. However, in vitro cancer cell invasion is mostly studied
using two-dimensional (2D) models. Three-dimensional (3D) multicellular spheroids may offer an advan-
tageous cell model for cancer research and oncology drug discovery. This chapter describes a label-free, real-
time, and single-cell approach to quantify the invasion of 3D spheroid colon cancer cells through Matrigel
using a spatially resolved resonant waveguide grating imager.
Key words Adhesion, Colorectal cancer cell, Dynamic mass redistribution, Extracellular matrix,
Invasion, Migration, Multicellular spheroid, PTEN, Resonant waveguide grating
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_10,
© Springer Science+Business Media LLC 2017
143
144 Nicole K. Febles et al.
1.1 Sensor For the label-free single cell 3D2 invasion assay, there are four
Configurations and critical components, including resonant waveguide grating
Detection Schemes (RGW) biosensor microplate, Matrigel, spheroids of cancer cells,
and RWG imager (Fig. 1).
The biosensor microplate, commercially known as Epic®
384well cell culture compatible microplate from Corning
Incorporated, is used directly. Within the bottom of each well of
the microplate, there is a RWG biosensor. The biosensor itself
consists of three layers—a glass substrate, a grating structure, and
a waveguide thin film with high refractive index (Fig. 1a). Under
illumination with a light source light at a specific wavelength is
coupled into the waveguide through diffraction, creating a surface
Label-Free Single Cell 3D2 Invasion 145
Fig. 1 Spatially resolved resonant waveguide grating (RWG) imager for label-free single cell monitoring. (a)
The principle of RWG biosensor for monitoring the invasion and adhesion of cancer cells. (b) The instrument.
(c) Optical setup of the imager, which consists of three components: a tunable light source to sweep the
wavelength range, an array of optical components to guide and expand light path leading to illuminate at a
normal incident angle all biosensors within a plate, and a CMOS camera to record the resonant wavelength
image. (d) A false-colored DMR image of a 3 4 biosensor array. All biosensors were pre-coated with 10 μL
0.1 mg/mL Matrigel. The image was obtained 24 h after spheroids of HT29 cells in the absence and presence
of vandetanib at different doses were placed on the top Matrigel surface. False color bar scale: 500 (blue) to
2000 pm (red). The pattern and area of adherent cells is indicated by the green-to-red spot, while the signal
intensity is indicated by the false color scale—red: high positive signal; green: moderate positive signal; blue:
negative signal. The well corresponding to the second left in the top row did not receive any spheroid as the
negative control. Condition of spheroid formation: 40K per well seeding density, 4 days culture. The adhesion
area and signal was found to be decreased as the dose of vandetanib increases
2 Materials
2.1 Tissue Culture 1. Human colorectal adenocarcinoma HT-29 cell line (ATCC®
Medium and Cell Line HTB-38™, American Type Cell Culture, Manassas, VA, USA).
2. The isogenic colorectal carcinoma (CRC) cell lines, HCT116-
WT and HCT116-PTEN/ cells (Horizon Discovery Ltd.,
Cambridge, UK) (see Note 1).
3. McCoy’s 5A medium (Catalog #16600, Life Technologies,
Carlsbad CA, USA).
4. Complete medium for HT29: McCoy’s 5A modified medium
supplemented with 10% fetal bovine serum, 4.5 g/L glucose,
2 mM glutamine, and 1 Pennstrep.
5. Complete medium for the isogenic cell lines: McCoy’s 5A
medium supplemented with 10% fetal bovine serum, 4.5 g/L
glucose, 1.5 mM glutamine, and 1 Pennstrep.
6. Serum-free medium for the isogenic cell lines: McCoy’s 5A
medium supplemented with 4.5 g/L glucose, 1.5 mM gluta-
mine, and 1 Pennstrep.
7. Pennstrep antibiotic solution 100: 10,000 units penicillin/
mL, 10,000 μg streptomycin/mL.
148 Nicole K. Febles et al.
3 Methods
3.1 3D Spheroid Cell This section describes the protocol to form uniform spheroids of
Culture three CRC cell lines by directly culturing an equal number of cells in
each well of a 96-well ULA round bottomed microplate (Fig. 2a, b).
The spheroids formed can be transitional (meaning that it can
grow in size over time) or stable (meaning that it stops growing).
For the 3D2 invasion assay, mechanically stable spheroids are
preferred since it is important to minimize potential interference
with the assay results of mechanically dissociated cells from the
Label-Free Single Cell 3D2 Invasion 149
Fig. 2 Growth pattern of spheroidal cells of HCT116 and its isogenic PTEN null cell line in 96-well ULA round
bottomed plate. (a) ULA microplate. (b) schematic drawing showing how a spheroid is formed within a ULA
round bottomed well. (c, d) Daily light microscopic images of HCT116 WT cells (c), or HCT116 PTEN/ cells
(d) at Day 0 to Day 5 (i–v, respectively). Seeding density: 1000 cells per well; scale bar: 500 μm. (e, f) The
diameter of spheroidal clusters of both cell lines as a function of culture duration when seeding density is 1000
(e) or 20,000 (f) cell per well. This figure is adapted from ref. 27 with permission
150 Nicole K. Febles et al.
3.2 Matrigel Coating This section describes the protocol to coat RWG biosensor plates or
Transwell inserts with Matrigel for invasion assays.
1. Precool Matrigel, biosensor plates, Transwell inserts, pipets,
and tips at 4 C before coating. This step is essential to prevent
premature gelation, and to ensure uniform coating.
2. Dilute Matrigel using the complete media to 0.1 mg/mL or
0.2 mg/mL (1 50 or 1 25 dilution, respectively) at 4 C.
3. Add 10 or 20 μL the diluted Matrigel solution to each well of a
384well biosensor microplate, or 100 μL to a Transwell insert
at 4 C.
4. Incubate the microplate or Transwell insert inside an incubator
at 37 C under air/5% CO2 for 24 h to form a 3D gel on the
biosensor surface, or on the Transwell insert.
3.3 Label-Free Single This section describes the protocol to perform label-free single cell
Cell 3D2 Invasion 3D2 invasion assay. This assay starts with placing a single spheroid
Assay onto the top surface of Matrigel, followed by that individual cells
spontaneously transfer from the spheroid to the top Matrigel sur-
face, invade through the matrix, and eventually adhere onto the
biosensor surface (Fig. 3). The RWG imager monitors in real time
the cell adhesion events, which, in turn, are used as an indicator to
determine the invasive potential of a cancer cell line. This assay
effectively mimics the first critical step of cancer metastasis; that is,
the local invasion of cancer cells through surrounding ECM, which
involves adhesion, proteolytic remodeling of the ECM, and migra-
tion [47–50].
This assay enables multiparametric analysis of cell invasion and
adhesion (Fig. 4), The initial adhesion point, designated as the
coordinate of (0, 0), is first identified as the pixelated location at
which the first positive DMR occurs. The lateral distance of any
pixel relative to the initial adhesion point is then calculated. The
maximal DMR at each pixel is extracted from its DMR signal. The
adhesion time at each pixel is also extracted from its DMR as the
time reaches a specific DMR amplitude (e.g., 500 pm for HT29, or
200 pm for the isogenic cell lines). The total adhesion events are
obtained by counting the number of pixels that give rise to a DMR
exceeding a predetermined threshold signal (e.g., 500 pm for
HT29, or 200 pm for the isogenic cell lines). The total adhesion
area is determined using ImageJ as the area that gave rise to a signal
above the background. Correlation analysis can then be performed.
For HCT116, PTEN deletion was found to increase the invasion
distance (Fig. 5a vs. Fig. 5b), and the total adhesion events
(Fig. 5c), suggesting that PTEN deletion increases the invasion
152 Nicole K. Febles et al.
Fig. 3 Label-free single cell 3D2 invasion assay. (a) The assay consists of four critical steps: coating the
biosensor surface with Matrigel; adding medium containing an inhibitor compound to the well; transferring a
spheroid from a ULA round bottomed microplate and placing it onto the top Matrigel surface; and monitoring
the invasion and adhesion in real time. (b–e) The time series DMR images of a biosensor before and after a
single spheroid was placed onto the biosensor surface coated with 10 μL 0.1 mg/mL Matrigel: 0 min (b), 1 h.
(c), 6 h (d), and 24 h (e). Spatial scale bar: 500 μm. Intensity scale bar: false colored (500 to 2000 pm). (f–i)
The time series light microscopic images of a biosensor before and after a single spheroid was placed onto
Matrigel coated biosensor surface: 0 min (f), 1 h. (g), 6 h (h), and 24 h (i). Scale bar: 500 μm. Condition of
spheroid formation: 40K per well seeding density, 4 days culture. (b–i) is adapted with permission from ref.
26. Copyright (2015) American Chemical Society
Fig. 4 Label-free single cell analysis for the invasion and adhesion of HT29 cell spheroids. (a) A DMR image
taken 24 h after a spheroid was placed on the top Matrigel surface. Scale bar: 500 μm. (b) Real-time
population averaged DMR on the Matrigel coated surface. (c) Pixelated real-time DMR signals for the line
indicated in (a). (d) The correlation between the adhesion time and the lateral distance relative to the first
adhesion point. (e) The correlation between the maximal response and the relative distance. (f) The correlation
between the adhesion time and the maximal response. The biosensor was coated with 10 μL 0.1 mg/mL
Matrigel. Condition of spheroid formation: 40K per well seeding density, 4 days culture. This figure is adapted
(b–j) is adapted with permission from ref. 26. Copyright (2015) American Chemical Society
Fig. 5 PTEN deletion increases the invasion rate of CRC cells in spheroid through 3D Matrigel matrix. (a, b)
Correlation analysis between the adhesion time and lateral distance for the parental (a) and PTEN/ (b)
cells. (c) The adhesion events versus cell types. (d) The adhesion time to reach 200 pm under different
conditions. Coating: 0.2 mg/mL Matrigel. Data represent mean s.d. for c, d (n ¼ 3). ***p < 0.001. This
figure is adapted from ref. 27 with permission
3.4 Cell Monolayer This section describes the wound healing assay protocol to detect
Wound Healing Assay the migratory potential of CRC cells. This assay is to first culture
cells to form a monolayer, then physically create a wounded area,
and finally monitor the wound recovery over time using light
microscopy (Fig. 6a). The migratory behavior of cells, one of the
key hallmarks of a metastatic cancer cell, is reflected by the rate of
recovery of the scratch region (Fig. 6b).
1. Harvest cells from T75 flask using 1 trypsin–EDTA.
2. Centrifuge the cells down, remove the supernatant, and resus-
pend the cell pellet in freshly prepared complete cell culture
medium.
3. Add cells to a 6-well plate and grow cells to confluency at 37 C
with 5% CO2.
4. Wash the confluent cells three times with the serum free
medium, and starve the cells overnight.
5. Produce a scratch with a sterile 200 μL pipette tip in a marked
region of the well.
6. Collect immediately light microscopic images of the marked
scratch area.
Fig. 6 2D cell wound recovery assay showing that PTEN deletion increases the
migratory potential of HTC116 cells. (a) Critical steps of the assay. (b) The 2D
migration rate of HCT116 versus HCT116-PTEN/ cells. Data represents
mean s.d. (n ¼ 5). ***p < 0.001. This figure is adapted from ref. 27 with
permission
156 Nicole K. Febles et al.
3.5 Transwell This section describes the Transwell invasion assay protocol to
Invasion Assay of detect the impact of PTEN deletion on the invasive potential of
Individual Cancer Cells HCT116 cells. This assay measures the invasion of individual cells
through Matrigel coated substrates having defined micropores
(Fig. 7a). This assay mimics local invasion of tumor cells through
the ECM or basement membrane. The invasiveness is quantified by
counting the number of invaded cells and normalizing to those in
uncoated Transwell inserts (Fig. 7b, c).
1. Harvest cells from T75 flask using 1 trypsin–EDTA.
2. Centrifuge the cells down, remove the supernatant, and resus-
pend the cell pellet in serum free media.
3. Add 100 μL cell suspension to the top side of the Matrigel
coated Transwell inserts with a cell density of 2.5 104 cells/
mL. Use the uncoated Transwell inserts as the control.
Fig. 7 Transwell invasion assay showing that PTEN deletion increases the invasiveness of the CRC cells. (a)
Critical steps of the assay. (b) Representative light microscopic images of HCT116 versus HCT116-PTEN/
cells adhered on the underside of Transwell inserts 24 h after invasion. (c) The invasion rate through Matrigel
coated Transwell of HCT116 versus HCT116-PTEN/ cells. Data represents mean s.d. (n ¼ 5).
***p < 0.001. This figure is adapted from ref. 27 with permission
Label-Free Single Cell 3D2 Invasion 157
4. Place the inserts having cells to a 24-well plate, each well being
filled with 750 μL of the cell culture media with 5% FBS as a
chemoattractant, to form invasion chambers.
5. Incubate the invasion chambers for 24 h at 37 C with 5% CO2.
6. Remove the inserts and wash twice with 1 phosphate buff-
ered saline.
7. Discard the non-invading cells using a cotton swab moistened
with culture media.
8. Fix the adhered cells in 4% paraformaldehyde, permeablize in
100% methanol, and stain using crystal violet stain.
9. Manually count the number of invading cells on the underside
of the Transwell using a light microscope.
10. Calculate the percentage invasiveness by normalizing the num-
ber of invading cells in the Matrigel coated inserts to that in the
uncoated inserts.
4 Notes
References
1. Steeg PS, Theodorescu D (2008) Metastasis: a 6. Carragher N (2009) Cell migration and inva-
therapeutic target for cancer. Nat Clin Pract sion assays as tools for drug discovery. Clin Exp
Oncol 5:206–219. doi:10.1038/ncponc1066 Metastasis 26:381–397. doi:10.3390/
2. Desgrosellier JS, Cheresh DA (2010) Integrins pharmaceutics3010107
in cancer: biological implications and therapeu- 7. Kramer N, Walzl A, Unger C, Rosner M, Kru-
tic opportunities. Nat Rev Cancer 10:9–22. pitza G, Hengstschl€ager M, Dolznig H (2013)
doi:10.1038/nrc2748 In vitro cell migration and invasion assays.
3. Eccles SA, Box C, Court W (2005) Cell migra- Mutat Res 752:10–24. doi:10.1016/j.mrrev.
tion/invasion assays and their application in 2012.08.001
cancer drug discovery. Biotechnol Annu Rev 8. Quail DF, Maciel TJ, Rogers K, Postovit LM
11:391–421. doi:10.1016/S1387-2656(05) (2012) A unique 3D in vitro cellular invasion
11013-8 assay. J Biomol Screen 17:1088–1095. doi:10.
4. Albini A, Benelli R (2007) The chemoinvasion 1177/1087057112449863
assay: a method to assess tumor and endothelial 9. Chitcholtan K, Asselin E, Parent S, Sykes PH,
cell invasion and its modulation. Nat Protoc Evans JJ (2013) Differences in growth proper-
2:504–511. doi:10.1038/nprot.2006.466 ties of endometrial cancer in three dimensional
5. Kam Y, Guess C, Estrada L, Weidow B, Quar- (3D) culture and 2D cell monolayer. Exp Cell
anta V (2008) A novel circular invasion assay Res 319:75–87. doi:10.1016/j.yexcr.2012.09.
mimics in vivo invasive behavior of cancer cell 012
lines and distinguishes single-cell motility in 10. H€arm€a V, Virtanen J, M€akel€a R, Happonen A,
vitro. BMC Cancer 8:198. doi:10.1186/ Mpindi JP, Knuuttila M, Kohonen P, Lötjönen
1471-2407-8-198 J, Kallioniemi O, Nees M (2010) A
Label-Free Single Cell 3D2 Invasion 159
establishment and analysis of three-dimen- 42. Fang Y (2013) Troubleshooting and deconvo-
sional tumor spheroid-based functional assays luting label-free cell phenotypic assays in drug
for target validation and drug evaluation. BMC discovery. J Pharmacol Toxicol Methods
Biol 10:29. doi:10.1186/1741-7007-10-29 67:69–81. doi:10.1016/j.vascn.2013.01.004
33. Ferrie AM, Deichmann OD, Wu Q, Fang Y 43. Fang Y (2014) Label-free drug discovery.
(2012) High resolution resonant waveguide Front Pharmacol 5:52. doi:10.3389/fphar.
grating imager for cell cluster analysis under 2014.00052
physiological condition. Appl Phys Lett 44. Fang Y (2014) Label-free cell phenotypic drug
100:223701. doi:10.1063/1.4723691 discovery. Comb Chem High Throughput
34. Ferrie AM, Wu Q, Deichmann O, Fang Y Screen 17(7):566–578
(2014) High frequency resonant waveguide 45. Song MS, Salmena L, Pandolfi PP (2012) The
grating imager for assessing drug-induced car- functions and regulation of the PTEN tumour
diotoxicity. Appl Phys Lett 104:183702. suppressor. Nat Rev Mol Cell Biol
doi:10.1063/1.4876095 13:283–296. doi:10.1038/nrm3330
35. Fang Y, Ferrie AM, Fontaine NH, Yuen PK 46. Kasinskas RW, Venkatasubramanian R, Forbes
(2005) Characteristics of dynamic mass NS (2014) Rapid uptake of glucose and lactate,
redistribution of EGF receptor signaling in liv- and not hypoxia, induces apoptosis in three-
ing cells measured with label free optical bio- dimensional tumor tissue culture. Integr Biol
sensors. Anal Chem 77:5720–5725. doi:10. 6:399–410. doi:10.1039/c4ib00001c
1021/ac050887n 47. Hanahan D, Weinberg RA (2011) Hallmarks
36. Fang Y (2011) Label-free biosensors for cell of cancer: the next generation. Cell
biology. Intl J Electrochem 2011:e460850. 144:646–674. doi:10.1016/j.cell.2011.02.
doi:10.4061/2011/460850 013
37. Schröder R, Janssen N, Schmidt J, Kebig A, 48. Valastyan S, Weinberg RA (2011) Tumor
Merten N, Hennen S, M€ uller A, Bl€attermann metastasis: molecular insights and evolving
S, Mohr-Andr€a M, Zahn S, Wenzel J, Smith paradigms. Cell 147:275–292. doi:10.1016/
NJ, Gomeza J, Drewke C, Milligan G, Mohr K, j.cell.2011.09.024
Kostenis E (2010) Deconvolution of complex 49. Stetler-Stevenson WG, Aznavoorian S, Liotta
G protein-coupled receptor signaling in live LA (1993) Tumor cell interactions with the
cells using dynamic mass redistribution mea- extracellular matrix during invasion and metas-
surements. Nat Biotechnol 28:943–949. tasis. Annu Rev Cell Biol 9:541–573
doi:10.1038/nbt.1671
50. Poincloux R, Lizárraga F, Chavrier P (2009)
38. Verrier F, An S, Ferrie AM, Sun H, Kyoung M, Matrix invasion by tumour cells: a focus on
Fang Y, Benkovic SJ (2011) GPCRs regulate MT1-MMP trafficking to invadopodia. J Cell
the assembly of a multienzyme complex for Sci 122:3015–3024. doi:10.1242/jcs.034561
purine biosynthesis. Nat Chem Biol
7:909–915 51. Lee C, Kim JS, Waldman T (2004) PTEN gene
targeting reveals a radiation-induced size
39. Ferrie AM, Wang C, Deng H, Fang Y (2013) checkpoint in human cancer cells. Cancer Res
Label-free optical biosensor with microfluidics 64:6906–6914. doi:10.1158/0008-5472.
identifies an intracellular signalling wave CAN-04-1767
mediated through the β2-adrerengic receptor.
Integr Biol 5:1253–1261 52. Samuels Y, Diaz LA Jr, Schmidt-Kittler O,
Cummins JM, Delong L, Cheong I, Rago C,
40. Li G, Lai F, Fang Y (2012) Modulating cell-cell Huso DL, Lengauer C, Kinzler KW, Vogelstein
communication with a high-throughput label- B, Velculescu VE (2005) Mutant PI3K3CA
free cell assay. J Lab Automation 17:6–15. promotes cell growth and invasion of human
doi:10.1177/2211068211424548 cancer cells. Cancer Cell 7:561–573. doi:10.
41. Pai S, Verrier F, Sun H, Hu H, Ferrie AM, 1016/j.ccr.2005.05.014
Eshraghi A, Fang Y (2012) Dynamic mass 53. Fang Y (2015) Label-free cell phenotypic
redistribution assay decodes differentiation of profiling and screening: techniques, experi-
a neural progenitor stem cell. J Biomol Screen mental design and data assessment. Methods
17:1180–1191. doi:10.1177/ Pharmacol Tox 53:233–252. doi:10.1007/
1087057112455059 978-1-4939-2617-6_2
Chapter 11
Abstract
The bimodal waveguide (BiMW) sensor is a novel common path interferometric transducer based on the
evanescent field detection principle, which in combination with a bio-recognition element allows the direct
detection of biomolecular interactions in a label-free scheme. Due to its inherent high sensitivity it has great
potential to become a powerful analytical tool for monitoring substances of interest in areas such as
environmental control, medical diagnostics and food safety, among others. The BiMW sensor is fabricated
using standard silicon-based technology allowing cost-effective production, and meeting the requirements
of portability and disposability necessary for implementation in a point-of-care (POC) setting.
In this chapter we describe the design and fabrication of the BiMW transducer, as well as its application
for bio-sensing purposes. We show as an example the biosensor capabilities two different applications: (1)
the immunodetection of Irgarol 1051 biocide useful in the environmental field, and (2) the detection of
human growth hormone as used in clinical diagnostics. The detection is performed in real time by
monitoring changes in the intensity pattern of light exiting the BiMW transducer resulting from
antigen–antibody interactions on the surface of the sensor.
Key words Bimodal waveguide interferometry, Evanescent field biosensor, Integrated optics, Immu-
noassay, Biofunctionalization, Biorecognition
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_11,
© Springer Science+Business Media LLC 2017
161
162 Sonia Herranz et al.
2πL
Δφðt Þ ¼ Δneff
λ
where L is the length of the sensing arm, λ the wavelength of the
light, and Δneff the difference between the effective refractive index
of the two propagated modes.
The phase difference, in an experimental interferometric
signal, is determined by the variation between the initial value,
Δφ1, and the final value, Δφ2, given by the number of fringes,
where a complete fringe represents a phase difference of 2π, as
Fig. 1 shows.
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 163
Fig. 2 (a) Scheme of a BiMW interferometric sensor. (b) Longitudinal side view
of a BiMW showing the distribution of the electromagnetic field in several points
2 Materials
3 Methods
Fig. 3 Scheme of the experimental setup employed for the evaluation of the BiMW sensors
where Iup and Idown are the intensities acquired for the up and down
quadrants of the photodetector, respectively.
For the application of the BiMW interferometer as a biosensor
the surface must be modified with a recognition element or
biological receptor which, when immobilized onto the surface,
increases the transducer selectivity for the substance to be moni-
tored. The environmental and clinical diagnostic BiMW interfero-
metric biosensor applications will be illustrated.
Irgarol 1051 is a protective algaecide (biocide, s-triazine
compound) used in antifouling paints to prevent attachment and
growth of biofouling organisms in the aquatic environment. It leaks
out from the paint and by its toxicity Irgarol 1051 prevents accu-
mulation of organisms onto the painted surface (boat hulls or
marine installations). Traces of Irgarol 1051 have been detected
in the marine environment [7, 8], and due to its broad toxicity
(photosystem-II inhibitor), Irgarol 1051 is potentially toxic toward
valuable aquatic species (mainly small aquatic plants), and its pres-
ence in the environment can cause significant environmental
problems. In fact, a number of studies have demonstrated its
negative impact on marine organisms such as corals.
Human growth hormone (hGH) is produced by the pituitary
gland and promotes growth, regulates the activity of vital organs
and helps to preserve the health of the whole organism. Its defi-
ciency commonly affects children and has several negative effects
such as hypoglycemia in newborn infants or stunted growth in
children. hGH deficiency is results from several genetic diseases
such as Turner syndrome and Prader-Willi syndrome. Thus the
hGH level in blood or urine can be used for the diagnosis of
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 169
Fig. 4 Immunoassay formats: (a) Competitive immunoassay format selected for Irgarol 1051 detection (b)
Direct immunoassay format selected for hGH detection. For sensor surface details, see Fig. 7
a b
Amax Amax
100% 100%
90%
Sensor response
Sensor response
80% 80%
50% 50%
20% 20%
Amin 10% Amin
0% 0%
Fig. 5 Standard calibration curves showing the relationship between the analyte concentration ([Analyte],
logarithmic scale) and the sensor response for: (a) competitive and (b) direct immunoassays
Table 1
Layer parameters employed for the evaluation of the single-mode and
bimodal condition
3.1 BiMW Simulation (a) To design a BiMW we need to choose optimal materials,
and Fabrication which ensure a cost-effective fabrication, a further integra-
tion onto a complete LOC platform, and a highly sensitive
3.1.1 BiMW Simulation
device. As the transducers are designed for biosensors appli-
cations, SiO2/Si3N4/SiO2 waveguides are selected [9].
Table 1 summarizes the parameters employed in the calcula-
tion, relative to the materials’ refractive indices and their
thicknesses.
(b) Once the material is defined, core dimensions are analyzed to
ensure the best sensitivity of the device. For that, different
considerations have to be taken into account [4] (see Fig. 6):
l Single-mode behavior (mode TE00 or TM00) in section A
in Fig. 6b.
l Bimodal behavior (mode TE00 and TE10 or TM00 and
TM10) in sections B, C, and D in Fig. 6b.
l Performances achievable in the clean room facilities.
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 171
Fig. 6 (a) Front cross section of the employed rib-waveguide, indicating the main parameters described on the
text. (b) Longitudinal side view of the BiMW device. Light is injected in the single-mode section (A). After an
abrupt step junction, two modes propagate till the end of the device. Regions B and D are covered by silicon
dioxide while C is exposed to the external medium (sensing area). (c) Picture of a chip with 20 BiMW sensors
Table 2
Optimal parameters for core dimensions in a high sensitivity BiMW
Fig. 7 Scheme of the fabrication process of integrated BiMW sensors. (a) A layer
of SiO2 and Si3N4 is deposited on a Si wafer. (b) Step-junction etched in Si3N4
layer for single-mode section definition. (c) Rib structure is defined. (d) Poly-
crystalline deposition over the Si3N4 and etching process to open desired
window on this layer. (e) SiO2 deposition and sensing area opening
3.1.2 BiMW Fabrication Figure 7 summarizes the fabrication process. Below, each step of
Fig. 7 is explained.
(a) The fabrication processing starts with a 4-in. (p-type) Si
wafer with a thickness of 500 μm, over which a 2 μm thick
layer of thermal oxide is grown. Then a 340 nm thick core
layer of Si3N4 is deposited by LPCVD.
(b) The thickness of the Si3N4 is reduced 190 nm to obtain a
150 nm single-mode section using a wet etching process with
75% phosphoric acid at 160 C. A hard mask constituted by a
layer of SiO2 deposited by PECVD and defined by photoli-
thography is employed in this step to protect unexposed
regions.
(c) Using a wet etching process with BHF and a standard pho-
tolithography process, the rib structure of the waveguide
(3 μm in width and 1.5 nm in height) is defined. The low
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 173
3.2 Setup (a) The BiMW chip is placed in an aluminum homemade holder
Configuration as shown in Fig. 8. Into the holder a temperature stabiliza-
tion is included, and the entire system is mounted on a 3-axis
stage. In this way, it is possible to focus the light in the
desired BiMW sensor optimally.
(b) As the total output intensity must be divided in the up and
down quadrant of the photodetector, this one is joined in a
2-axis stage assembly to the aluminum chip holder, to ensure
the right position of the photodetector at the BiMW
Fig. 10 Calibration curve for a BiMW sensor. Insets represent the temporal interferometric signals
corresponding to the entrance of solutions with different refractive indexes
Fig. 11 Functionalization scheme/protocol: (1) cleaning and oxidation; (2) APTES silanization; (3) covalent
attachment of hapten 4e; (4) activation of APTES-modified surface with PDITC, and (5) covalent attachment of
anti-hGH antibody. Sensor surface I: 4e-functionalized surface, used for Irgarol 1051 immunodetection.
Sensor surface II: anti-hGH antibody-functionalized surface, used for hGH immunodetection
3.4.1 Cleaning Procedure (a) Wipe the BiMW chip down using a cleanroom paper soaked
in acetone.
(b) Sequentially rinse the chip with acetone, ethanol, and
deionized water, and dry under nitrogen flow.
(c) Immerse the chip in 1% SDS aqueous solution and sonicate
for 5 min.
(d) Rinse the chip with deionized water and dry again under
nitrogen flow.
178 Sonia Herranz et al.
3.4.2 Oxidation (a) Treat the cleaned BiMW chip with oxygen plasma (100 W, 45
Procedure sccm) for 5 min (see Note 4).
(b) Immerse the chip in 15% HNO3 at 75 C for 25 min.
(c) Rinse generously the chip with deionized water, dry carefully
under nitrogen flow and immediately immerse it in the
silanization solution.
3.4.3 BiMW (a) Clean and oxidize the BiMW chip according to the previous
Interferometer Silanization sections.
Procedure Using APTES (b) Immediately immerse the BiMW chip in a solution of 1%
APTES and incubate at RT for 1 h. Two different solvents
have been evaluated: absolute ethanol and toluene.
(c) Amply rinse the chip with the silanization solvent and
deionized water, sequentially.
(d) Dry the chip thoroughly under nitrogen flow and thermally
treat it in an oven at 110 C for 1 h.
3.4.5 Covalent (a) Prior to the covalent attachment of the anti-hGH antibody,
Immobilization of Anti-hGH the APTES-modified chip (silanization solvent: ethanol)
Antibody must be activated with PDITC, which acts as cross-linker.
After PDITC activation the antibody can be immobilized
through the amine groups present in its structure.
(b) Prepare a 20 mM PDITC solution (5 mL) in anhydrous
DMF containing 1% pyridine.
(c) Immerse the APTES-modified chip in PDITC solution and
incubate in the dark for 2 h.
(d) Wash off unbound reagents by sequentially rinsing the chip
with acetone, ethanol and water, and dry it under nitrogen
stream.
(e) Add 50–100 μL of a solution of anti-hGH antibody in
carbonate buffer onto the PDITC-modified chip, cover it
with a coverslip and incubate overnight, in the dark.
Concentrations must be optimized for each application. For
hGH immunoassay, a 50 μg/mL solution was used.
(f) Rinse the functionalized chip with PBS and carefully dry it
under nitrogen stream.
(g) At this point, the functionalized BiMW chip is positioned on
the experimental apparatus and the PDMS flow-chamber
mounted. Before starting sample evaluation the sensor sur-
face is blocked to avoid nonspecific adsorption and reduce
background signal (see Note 5).
(h) Initiate a continuous flow of PBS and wait until the inter-
ferometric signal stabilizes.
(i) Inject into the flow cell a 2 mg/mL BSA solution in PBS
until the flow channel is full. Then stop the flow and incubate
for 1 h (see Note 6).
(j) The BiMW chip is now functionalized and ready to begin the
sample evaluation. If not used immediately chips must be
dried and stored at 4 C. Before use, the chip is positioned
again on the experimental apparatus and the sensor surface is
conditioning by maintain a continuous flow of PBS for 1 h.
180 Sonia Herranz et al.
3.5 Detection by a (a) Initiate a continuous flow of PBS and wait until stabilization
Competitive of the interferometric signal. This buffer will be used to
Immunoassay perform the Irgarol 1051 immunodetection assay.
3.5.1 Selection of the
(b) Prepare series of solutions (300 μL) with different dilution
Antibody Concentration factors of the antibody (serum As87). For Irgarol 1051
detection, dilutions ranging from 1:10,000 to 1:500 were
used.
(c) Inject into the flow cell the antiserum solutions with a
regeneration step in between, and measure the phase shift
(sensor response). Regeneration consists in disrupting the
receptor–antibody interaction while maintaining the recep-
tor onto the surface with minimal damage, by washing up the
sensor surface with the so-called regeneration solution.
Regeneration solution composition must be empirically opti-
mized (see Note 7). For Irgarol 1051 immunoassay, a 50 mM
NaOH solution was employed.
(d) Construct a plot of sensor response vs. antiserum concentra-
tion. Choose an antibody concentration value (fixed concen-
tration to perform the inhibition assay) below the saturation
limit, able to give high enough response intensity (phase
shift), so that at inhibition, signals would be still clear. Take
into account that by decreasing the antibody concentration, a
better limit of detection can be achieved (see Note 8).
3.5.2 Detection of Irgarol (a) Initiate a continuous flow of PBS (20 μL/min) and wait until
1051 the interferometric signal is stabilized.
(b) Prepare a series of solutions of different concentrations of
Irgarol 1051 in PBS (300 μL, standard solutions). Concen-
trations of the standard solutions to be measure depend on
the limit of detection for each specific application. For Irgarol
1051, the standard solutions were in the ng/L to μg/L
range.
(c) Mix the series of standard solutions with a volume of As87
serum so that the final dilution factor in solution equals that
previously selected. Incubate the mixture for 10 min.
(d) Pump one antibody-Irgarol standard solution into the flow
cell at 20 μL/min. This step produces a shift phase of the
wave (Δφ) due to the interaction of the free antibody with
the immobilized antigen.
(e) Wash off the unbound antibody by pumping PBS for a few
minutes (20 μL/min).
(f) Regenerate the surface by injecting 50 mM NaOH
(20 μL/min, 120 s), and wait until stabilization of the signal
(surface conditioning). At this point, the surface is ready for
the measurement of a second standard solution. In Fig. 12,
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 181
As87 +
As87 Irgarol 1051 NaOH 50 mM As87 NaOH 50 mM
80
60
40
ΔS
ΔS
20
ΔS
S, %
0
–20
–40
–60
NaOH 50 mM
–80
2 12 32 42 72 82
Time, min
Fig. 12 Real monitoring of anti-Irgarol 1051 antibody (As87) detection. The presence of Irgarol 1051 (1 μg/L) in
solution leads to a decrease of the sensor response. The sensor surface was regenerated with 50 mM NaOH
3.6 Detection of hGH 1. Initiate a continuous flow of PBS and wait until the signal is
by a Direct stabilized.
Immunoassay 2. Prepare a series of solutions of different concentrations of hGH
in PBS (400 μL, standard solutions). For hGH, the standard
solutions were in the ng/L to μg/L range.
182 Sonia Herranz et al.
3. Inject into the flow cell the hGH solutions with a regeneration
step in between (regeneration solution: 20 mM HCl), and
measure the phase shift (sensor response).
4. Construct a plot of sensor response vs. hGH concentration and
fit the data to an appropriate curve-fitting model for obtaining
the calibration curve. The hormone hGH usually appears at
ng/L levels in urine, so a LOD at low ng/L is desirable.
5. Inject into the flow cell the sample (unknown hGH concentra-
tion), wait until stabilization of the signal and measure the
sensor response.
6. Estimate the concentration of hGH in the sample by interpo-
lating the measured sensor response in the fitting curve.
3.7 Data Analysis To quantify the concentration of the target analyte in the sample,
the sensor response is interpolated into the calibration curve. In
order to minimize the estimation error and obtain accurate con-
centration values it is extremely important to employ an appropriate
curve-fitting model. The most frequently model used to analyse
immunoassay data is the 4-parameter logistic (4-PL) model, which
fit the data by using the equation:
ðA max Amin Þ
y ¼ A min þ b
1 þ xc
where y is the sensor response, x is the concentration of the target
analyte, Amax and Amin are the asymptotic ends, b is the slope at the
inflection point of the curve and c is the concentration at the
inflection point and represents the half maximal inhibitory concen-
tration (IC50) and the half maximal effective concentration (EC50)
in a competitive and a direct assay, respectively [13] (see Note 10).
Typically, the detection limit (LOD) for a competitive and a direct
assay is calculated as the analyte concentration for which the signal
is inhibited or increase by 10% of sensor signal range (Amax – Amin),
respectively, and the dynamic range (DR) of the biosensor is eval-
uated as the analyte concentrations that produced a sensor signal
between 20 and 80% of (Amax – Amin) (see Fig. 4).
4 Notes
Acknowledgments
References
1. Hunsperguer RG (2009) Integrated optics: the- monoclonal antibodies specific for the human
ory and technology. Springer, New York, NY growth hormone 22 K and 20 K isoforms. J
2. Nishihara H, Haruna M, Suhara T (1989) Clin Endocrinol Metab 81:1613–1618
Optical integrated circuits, McGraw-Hill Opti- 6. Ballesteros, B., Barceló, D., Sanchez-Baeza, F.,
cal and Electro-optical Engineering. Series, Camps, F. and Marco, M.P (1998) Influence of
London the Hapten Design on the Development of a
3. Kozma P, Kehl F, Ehrentreich-Förster E, Competitive ELISA for the Determination of
Stamm C, Bier FF (2014) Integrated planar the Antifouling Agent Irgarol 1051 at Trace
optical waveguide interferometer biosensors: Levels. Anal Chem 70, 4004–4014.
A comparative review. Biosens Bioelectron 7. Ali, H.R, Arifin, M.M., Sheikh, M.A., Shazili,
58:287–307 N.A.M. and Bachok, Z. (2013) Occurrence
4. Zinoviev KE, González-Guerrero AB, Domı́n- and distribution of antifouling biocide
guez C, Lechuga LM (2011) Integrated Irgarol-1051 in coastal waters of Peninsular
bimodal waveguide interferometric biosensor Malaysia. Mar Pollut Bull 70, 253–257.
for label-free analysis. J Light Technol 8. Martı́nez K, Ferrer I, Hernando MD, Fernán-
29:1926–1930 dez-Alba AR, Marcé RM, Borrull F, Barceló D
5. Mellado M, Rodriguez-Frade JM, Kremer L, (2001) Occurrence of Antifouling Biocides in
Martinez-Alonso C (1996) Characterization of
Label-Free Biosensors Based on Bimodal Waveguide (BiMW) Interferometers 185
the Spanish Mediterranean Marine Environ- antifouling Irgarol 1051 and the fungicides
ment. Environ Technol 22:543–552 dichlofluanid and 4-chloro-3-methylphenol
9. Prieto F, Sepúlveda B, Calle A, Llobera A, in water samples. J Chromatogr A 839:
Domı́nguez C, Abad A, Montoya A, Lechuga 253–260
LM (2003) An integrated optical interfero- 13. Dunn J, Wild D (2013) Calibration curve
metric nanodevice based on silicon technology fitting. In: Wild D (ed) The immunoassay
for biosensor applications. Nanotechnology handbook, 4th edn. Elsevier, Oxford, pp
14:907–912 323–336
10. Dante S, Duval D, Fariña D, González-Guer- 14. Fisher MJE (2010) Amine coupling through
rero AB, Lechuga LM (2015) Linear readout EDC/NHS: a practical approach. In: Mol NJ,
of integrated interferometric biosensors using a Fischer MJE (eds) Surface Plasmon Resonance
periodic wavelength modulation. Laser Photon Methods and Protocols, 1st edn. Humana
Rev 9:248–255 Press, Springer New York, pp 55–73
11. Gandhiraman RP, Gubala V, Nam LCH, 15. Gottschalk PG, Dunn JR (2005) The five-
Volcke C, Doyle C, James B, Daniels S, parameter logistic: a characterization and com-
Williams DE (2010) Deposition of chemically parison with the four-parameter logistic. Anal
reactive and repellent sites on biosensor chips Bioanal 343:54–65
for reduced non-specific binding. Colloids Sur- 16. Giraldo J, Vivas NM, Vila E, Badia A (2002)
faces B 79:270–275 Assessing the (a)symmetry of concentration-
12. Peñalver A, Pocurull E, Borrull F, Marcé RM effect curves: empirical versus mechanistic
(1999) Solid-phase microextraction of the models. Pharmacol Ther 95:21–45
Chapter 12
Abstract
Protein microarrays are emerging tools which have become very powerful in multiplexed detection
technologies. A variety of proteins can be immobilized on a sensor chip allowing for multiplexed diagnos-
tics. Therefore, various types of analyte in a small volume of sample can be detected simultaneously. Protein
immobilization is a crucial step for creating a robust and sensitive protein microarray-based detection
system. In order to achieve a successful protein immobilization and preserve the activity of the proteins after
immobilization, DNA-directed immobilization is a promising technique. Here, we present the design and
the use of DNA-directed immobilized (DDI) antibodies in fabrication of robust protein microarrays. We
focus on application of protein microarrays for capturing and detecting nanoparticles such as intact viruses.
Experimental results on Single-particle interferometric reflectance imaging sensor (SP-IRIS) are used to
validate the advantages of the DDI method.
Key words Protein microarrays, DNA-directed antibody immobilization, Label free detection, Single
particle detection, SP-IRIS
1 Introduction
*
These authors contributed equally to this work.
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_12,
© Springer Science+Business Media LLC 2017
187
188 € u
Nese Lortlar Unl € et al.
Fig. 1 Conceptual representation of IRIS detection platform for digital detection of individual viral particles and
antigens labeled with gold nanoparticles
Fig. 4 A schematic representation of SP-IRIS substrate surface with ssDNA spots and the conversion of the
DNA chip into a triplex antibody array through DDI of antibodies. Reprinted with permission from Seymour
et al. Anal. Chem., 2015, 87 (20), pp. 10505–10,512. Copyright 2016 American Chemical Society
2 Materials
2.1 Single-Particle 1. Scientific CCD camera. Retiga 4000R (Qimaging, Corp., Sur-
Interferometric rey, BC, Canada).
Reflectance Imaging 2. Sample illumination source. 530 nm LED source (Thorlabs
Sensor (SP-IRIS) Inc., Newton, NJ, USA).
(Validation Platform: 3. Microscope objectives. 50 0.8 NA Nikon objective for dry
Can Be Replaced by samples or a 40 0.9 NA Nikon objective for samples in liquid
Another Single Particle (Nikon Inc., Melville, NY, USA) (see Note 1).
Detection Method)
4. CRISP (The Continuous Reflection Interface Sampling and
Positioning) autofocus system. MFC-2000 (Applied Scientific
Instrumentation, Eugene, OR, USA).
5. 50:50 Beam splitter (Thorlabs Inc., Newton, NJ, USA).
6. Lenses (Thorlabs Inc., Newton, NJ, USA). Achromatic doub-
lets for the visible spectrum. Catalogue numbers: f1: AC254-
030-A-ML, f2: AC254-050-A-ML and f3: AC254-200-A-ML.
7. Diffuser (Thorlabs Inc., Newton, NJ, USA). N-BK7 Ground
Glass Diffuser, 220 Grit.
8. XY stage (Micronix USA, Santa Ana, CA, USA).
9. Mechanical components for optical setup. Cage system and
lens holders (Thorlabs Inc., Newton, NJ, USA).
10. Computer. Data acquisition and processing.
2.2 Substrate 1. Silicon (Si/SiO2) chips with a patterned thermally grown sili-
Fabrication con dioxide for single particle detection experiments (Silicon
Valley Microelectronics, Santa Clara, CA, USA) (see Note 2).
2. Dimensions of the chips: 10 mm 10 mm 0.5 mm, with a
2.3 mm 2.3 mm active center region for spotting DNA probes.
Fig. 5 The images of an SP-IRIS chip and microfluidic cartridge for in-liquid SP-IRIS measurements (on the
left). Cross section model of the microfluidic cartridge and objective which indicates the fluidic path (in blue),
the sensor (in gray) and the chip and imaging window (in yellow) not to scale (on the right). Adapted with
permission from Sherr et al. ACS Nano, 2016, 10 (2), pp. 2827–2833. Copyright 2016 American Chemical
Society
3 Methods
3.1 DNA Sequence 1. DNA sequences are designed to minimize the hairpin, self-
Design dimer and heterodimer structures to increase the hybridization
efficiency and prevent cross hybridization (see Note 6).
2. Twenty base pair of DNA sequence used as the surface probe
(40 mer) is complementary to the antibody conjugated
sequence used in the antibody-conjugation.
3. Amine modification is introduced at the 50 -end of the surface
probe DNA sequence in order to achieve covalent immobiliza-
tion on the copoly(DMA-NAS-MAPS) polymer coated chip
surface. Table 1 shows the DNA sequences used in this work
for antibody immobilization.
4. Lyophilized oligonucleotides are dissolved in ultrapure water
to have a final concentration of 100 μM. 100 μL aliquots
DNA-Directed Antibody Immobilization for Robust Protein Microarrays. . . 195
Table 1
Model surface probe DNA sequence and its corresponding sequence used in antibody-conjugation
in this work
3.2 Protein 1. The silicon chips are functionalized with the polymer copoly
Microarray Fabrication (DMA-NAS-MAPS). Prior to polymer coating, the chips are
cleaned by sonicating in acetone for 5 min, rinsing in isopro-
panol and deionized water. Then, the chips are dried by blow-
ing nitrogen gas.
2. The chips are cleaned by plasma ashing with oxygen plasma at
300 sccm and 500 W for 10 min.
3. 1% (w/v) copoly(DMA-NAS-MAPS) is dissolved in a mixture
of Solution A2 and water (1:1) by vigorous vortexing.
4. The clean chips are placed in a plastic petri dish and the polymer
solution is poured onto the chips until they are fully covered
with the solution (see Note 7).
5. The chips are incubated in the polymer solution for 30 min at
room temperature while shaking.
6. After functionalization, the chips are rinsed and dipped in
deionized water for 3 min with shaking. This is repeated for
three times. Then, the chips are dried with nitrogen gas.
7. In a vacuum oven, the chips are baked for 15 min at 80 C (see
Note 8).
8. To prepare protein microarrays via DNA-directed immobiliza-
tion technique, surface probe oligonucleotide (Sequence B0 ) is
spotted on the polymer-functionalized chips at an optimum
concentration of 30 μM in sodium phosphate buffer (150 mM,
pH 8.5). In order to determine the effect of probe density, a
varying degree of immobilization for antibody-conjugate was
created and virus capture capacity was determined by spotting
DNA surface probe at different concentrations which are 3, 6,
196 € u
Nese Lortlar Unl € et al.
Fig. 6 IRIS average spot height measurements for direct-antibody and DNA-directed immobilized-antibody
spots. One nanometer of surface thickness corresponds to 1.2 ng/mm2 of antibody density. (a) Schematic
representation of the IRIS chip spotted with three different probe types: antibody, probe DNA and a negative
DNA sequence (not complementary to target antibody-conjugated DNA). (b) IRIS images of the chip at different
stages of the experiment: before hybridization of DNA B-conjugate, after hybridization of the conjugate and
after incubating with VSV virus. (c) IRIS surface height measurements for three different stages of the
experiment
Fig. 7 (a) Schematic representation of the SP-IRIS setup. LED is used for illumination and bright field image is
reflected on a CCD camera. (b) With the help of cross term (highlighted) in the equation, visibility of low-index
nanoparticles is enabled with their enhanced intensity by order of magnitude compared to scattered intensity
(middle term). (c) A representation of multiplexed SP-IRIS chip (bottom) and an actual image of detected
particles on the chip after processing (top). Reprinted with permission from Avci et al. Sensors, 2015, 15,
pp 17649–17665. 2016 Creative Commons (http://creativecommons.org/licenses/by/4.0/)
DNA-Directed Antibody Immobilization for Robust Protein Microarrays. . . 199
ɛp ɛm
α ¼ 4πɛ0 r 3 ð1Þ
ɛp þ 2ɛm
where r is radius of the particle, ɛp and ɛm are particle and
surrounding medium permittivity, respectively [20]. In interfero-
metric imaging techniques, in the intensity recorded at the detec-
tor, strong reference field and weak scattered fields coming from
the particle are mixed and can be expressed with the following
equation:
3.6 Single Particle 1. The flow cell is obtained by assembling the individual layers.
Detection (In Liquid The acrylic layer is placed as the base and then, biosensor chip is
Virus Detection put in the middle of the cartridge. After placing the second
Measurements) layer, the third layer is placed on top providing imaging win-
dow like a polymer cover glass. Then, the fourth and fifth layers
are placed and adaptors are attached to the fifth layer. FEP
tubing is assembled to the adaptors. The syringe pump is
connected via tubing to provide the flow. The flow is directed
in the channels to the chip surface with the precisely designed
microfluidic device as shown in Fig. 5.
2. The flow cell is placed on the sample stage of SP-IRIS under-
neath the objective for image acquisition. DNA-immobilized
SP-IRIS chips are already placed in the microfluidic cartridges.
3. Acquisition software is opened and the camera is turned on.
4. Number of images to be averaged is determined and selected.
Maximum exposure time before saturation of the camera is
selected.
5. First, 13F6-DNA, B-conjugate is flowed at a concentration of
5 μg/mL in PBS with 1% BSA for 1 h to achieve protein
immobilization on the chip.
6. To get pre-incubation particle counts in spots with different
concentration of protein immobilized, SP-IRIS images are
acquired.
DNA-Directed Antibody Immobilization for Robust Protein Microarrays. . . 201
Fig. 9 (a) Left IRIS image showing different surface densities of immobilized antibody-DNA conjugate after
hybridization of the B0 sequence with the 13F6-DNA “B” conjugate. Right image shows 13F6-antibody spots at
different concentrations. (b) Effect of surface antibody density (1 nm ¼ 1.2 ng/mm2) on virus capture
efficiency for both direct antibody and DNA-conjugated antibody immobilization. Average capture virus
densities (n ¼ 6 spots) obtained from SP-IRIS images versus average optical thickness of the antibody
spots obtained from IRIS images were plotted. The ellipses indicate the range of surface antibody heights
where the optimal virus capture occurs. Reprinted with permission from Seymour et al. Anal. Chem., 2015, 87
(20), pp. 10505–10512. Copyright 2016 American Chemical Society
Fig. 10 Comparing captured virus densities on direct-spotted and DNA-tethered antibodies from independently
performed experiments. Each letter represents a data point next to it. Experimental conditions corresponding
to A–G experiments are summarized in the table. Error bars show the variation between six spots for a given
experiment. Reprinted with permission from Seymour et al. Anal. Chem., 2015, 87(20), pp 10505–10512.
Copyright 2016 American Chemical Society
4 Notes
References
1. MacBeath G, Schreiber SL (2000) Printing pro- reversible, and site-selective surface binding of
teins as microarrays for high-throughput func- proteins by means of covalent DNA-
tion determination. Science 289:1760–1763 streptavidin conjugates. Anal Biochem
2. Sun H, Chen GYJ, Yao SQ (2013) Recent 268:54–63
advances in microarray technologies for prote- 14. Ladd J, Boozer C, Yu Q et al (2004) DNA-
omics. Chem Biol 20:685–699 directed protein immobilization on mixed self-
3. Hall DA, Tacek J, Snyder M (2007) Protein assembled monolayers via a streptavidin bridge.
microarray technology. Mech Ageing Dev 128 Langmuir 20:8090–8095
(1):161–167 15. Schroeder H, Adler M, Gergk K et al (2009)
4. Avci O, Lortlar UN, € Yalcin A et al (2015) User configurable microfluidic device for mul-
Interferometric Reflectance imaging sensor tiplexed immunoassays based on DNA-
(IRIS)-a platform technology for multiplexed directed assembly. Anal Chem 81:1275–1279
diagnostics and digital detection. Sensors 15 16. Washburn AL, Gomez J, Bailey RC (2011)
(7):17649–17665 DNA-encoding to improve performance and
5. Schwenk JM, Lindberg J, Sundberg M et al allow parallel evaluation of the binding charac-
(2007) Determination of binding specificities teristics of multiple antibodies in a surface-
in highly multiplexed bead-based antibody bound immunoassay format. Anal Chem
assays for antibody proteomics. Mol Cell Pro- 83:3572–3580
teomics 6:125–132 17. Wacker R, Niemeyer CM (2004) DNA- μFIA-a
6. Cretich M, Daaboul GG, Sola L et al (2015) readily configurable microarray-fluorescence
Digital detection of biomarkers assisted by immunoassay based on DNA-directed immo-
nanoparticles: application to diagnostics. bilization of proteins. Chem Bio Chem
Trends Biotechnol 33(6):343–351 5:453–459
7. Yurt A, Daaboul GG, Connor JH et al (2012) 18. Wacker R, Schroder H, Niemeyer CM (2004)
Single nanoparticle detectors for biological Performance of antibody microarrays fabri-
applications. Nanoscale 4(3):715–726 cated by either DNA-directed immobilization,
8. Walt D (2013) Optical methods for single mol- direct spotting, or streptavidin-biotin attach-
ecule detection and analysis. Anal Chem 85 ment: a comparative study. Anal Biochem
(3):1258–1263 330:281–287
9. Daaboul GG, Lopez CA, Chinnala J et al 19. Seymour E, Daaboul GG, Zhang X et al (2015)
(2014) Digital sensing and sizing of vesicular DNA-directed antibody immobilization for
stomatitis virus pseudotypes in complex media: enhanced detection of single viral pathogens.
a model for ebola and marburg detection. ACS Anal Chem 87(20):10505–10512
Nano 8(6):6047–6055 20. Avci O, Adato R, Yalcin OA et al (2016) Physi-
10. Monroe MR, Daaboul GG, Tuysuzoglu A et al cal modeling of interference enhanced imaging
(2013) Single nanoparticle detection for multi- and characterization of single nanoparticles.
plexed protein diagnostics with attomolar sen- Opt Express 24(6):6094–6114
sitivity in serum and unprocessed whole blood. 21. Pirri G, Damin F, Chiari M et al (2004) Char-
Anal Chem 85(7):3698–3706 acterization of the polymeric adsorbed coating
11. Sevenler D, Lortlar UN,€ € u MS (2015)
Unl€ for DNA microarray glass slides. Anal Chem 76
Nanoparticle biosensing with interferometric (4):1352–1358
reflectance imaging. In: Vestergaard MC, 22. Yalçin A, Damin F, Özkumur E et al (2009)
Kerman K, Hsing I-M, Tamiya E (eds) Nano- Direct observation of conformation of a poly-
biosensors and nanobioanalyses. Springer, meric coating with implications in microarray
Tokyo, pp 81–95 applications. Anal Chem 81(2):625–630
12. Monroe MR, Reddington A, Collins AD et al 23. Romanov V, Davido SN, Miles AR et al (2014)
(2011) Multiplexed method to calibrate and A critical comparison of protein microarray fab-
quantitate fluorescence signal for allergen- rication technologies. Analyst 139
specific IgE. Anal Chem 83(24):9485–9491 (6):1303–1326
13. Niemeyer CM, Boldt L, Ceyhan B et al (1999)
DNA-directed immobilization: efficient,
Chapter 13
Abstract
Reflectometry is classified in comparison to the commercialized refractometric surface plasmon resonance.
The advantages of direct optical detection depend on a sophisticated surface chemistry resulting negligible
nonspecific binding and high loading with recognition sites at the biopolymer sensitive layer of the
transducer. Elaborate details on instrumental realization and surface chemistry are discussed for optimum
application of reflectometric interference spectroscopy (RIfS). A standard protocol for a binding inhibition
assay is given. It overcomes principal problems of any direct optical detection technique.
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_13,
© Springer Science+Business Media LLC 2017
207
208 Guenther Proll et al.
Fig. 1 Scheme of the RIfS detection principle. The left part shows the superimposition of the reflected light
beams and the change in optical thickness during a binding event on the sensor surface. The right part shows
the corresponding change of the characteristic interference spectrum and the resulting binding curve
Reflectometric Interference Spectroscopy 209
2 Materials
2.1 Transducer The RIfS standard transducer consists of a glass substrate (D 263
glass, Schott AG, Germany) (~1 mm thick) coated with a 10 nm
layer of a material with high refractive index (usually Ta2O5 or
Nb2O5), and a top layer of SiO2 (330 nm). As a reference use a
transducer without SiO2 layer. Due to the technical specifications
of the components used for setting up a RIfS measurement system
and the investigated sample types (e.g., liquids or gases), other
transducers might be more beneficial. An ideal RIfS transducer is
designed to create the two reflected light beams of interest at
similar intensities. Therefore, it is recommended to use ray tracing
programs (e.g., FilmWizard by SCI Scientific Computing Interna-
tional or WVASE by J. A. Woollam) to simulate the expected
reflectivities for the visible wavelength range for a given RIfS
setup, taking into account the expected optical properties of the
investigated sample types.
2.4 Parallel Setup 1. White light source 100 W/12 V, Osram, Munich.
2. Lenses, mirror, and positioning optics, Spindler&Hoyer,
Göttingen
3. Polymer light guides (PMAA, n ¼ 1490, coupling element
1 2 (50:50), 1 mm, fiber diameter with SMA 905 fiber
connectors, Microparts, Dortmund.
4. Optical 4--1 multiplexer DiCon VX 500-C, Laser Compo-
nents, Olching.
5. MMS diode row spectrometer with Liliput-PC, ZEISS, Jena.
6. 1900 industrial standard housing by RS Components, Walldorf-
Mörfelden.
7. Commonly Microsoft Windows XP, Vista, Windows 7, Win-
dows 8, Windows 8.1, Windows 10 operating systems.
2.6 Advanced Single 1. Light source providing light of a single color (White light
Setup (See Fig. 4) source 100 W/12 V, Osram, Munich with Bandpassfilter
568 nm).
2. Phodiode based detection unit (Lightwave-Multimeter 2832-
C with Si-photodiodes of the series 818 from Newport).
2.7 Liquid Handling 1. Ten position valve VICI, Valco Europa, Schenkon,
Switzerland.
Reflectometric Interference Spectroscopy 211
2.8 Software 1. MeasureCR for capturing spectras and controlling the systems.
Depending on the selected spectrometer, any other software
control can also be used. As most commercial available spectro-
meters are supplied with ready to use software or plug-ins for
the widely used Lab-View solution (National Instruments Cor-
poration), it is recommended to use one of these solutions.
2. IFZCR for evaluating the interferograms. Any other software
solution capable for performing the same simple mathematical
calculations like Matlab (The MathWorks, Inc.) can be used.
3. MS-Excel (Microsoft) and Origin (MicroCal Inc.) or any
equivalent statistical software packages (open source software
is available) can be used for further processing of measured
data.
4. If an advanced single color setup for reflectometry measure-
ments is used, no spectras have to be analyzed. Therefore,
software only needs to control hardware components. All eval-
uation work can be done with software packages like Matlab or
Origin.
3 Methods
3.1 Setup for RIfS In the standard laboratory setup for RIfS (Fig. 2), the white light is
guided via an optical fiber (1 mm PMMA fiber) to the back of the
transducer mounted in a microfluidic flow cell, which in turn is
attached to a liquid handling system. The reflected light is gathered
in the same waveguide.
The fiber optics used is bifurcated (50:50 ratio), with one tail
leading to the light source and the other to the UV–Vis spectrometer.
Possible light sources are halogen lamps (e.g., 10 W halogen lamp with
fiber in-coupling optics consisting of front surface spherical mirror,
collimating lens, and an infrared absorption filter) or LEDs. For
the spectral detection of the reflected light, diode array spectrometers
are used normally. A gap (approx. 100 μm) between transducer
chip and the fiber output is filled with glycerol (80%) for refractive
index matching. Samples are handled by a flow system (Fig. 3)
(e.g., two peristaltic pumps, inject-load valve, and 6-way valve).
212 Guenther Proll et al.
Carrier P1
Flow D Waste
V1 Loading:
Injection:
Waste P2 V2
Sample
Loop
3.2 Surface All surface chemistry steps are applied to the coated side of the RIfS
Chemistry transducer (see Note 3) if not stated otherwise.
3.2.1 Silanization (a) Transducer chips are cleaned with freshly prepared Piranha
solution (mixture of 30% hydrogen peroxide and concen-
trated sulfuric acid at a ratio of 2:3; caution: hot and aggres-
sive!) for 30 min in an ultrasonic bath to clean the chip and to
generate silanol groups on the transducer surface. As an
alternative, this activation step can be carried out using oxy-
gen plasma.
(b) After rinsing with double distilled water and drying in a
nitrogen stream, the surface is immediately activated by
GOPTS at a surface concentration of approx. 10 μl/cm2 for
1 h in dryness (by assembling two slides face-to-face placed in
a tray with ground joint) followed by cleaning with dry
acetone and drying in a nitrogen stream (see Notes 4 and 5).
(a) Aminodextran.
The coupling of aminodextran as an aqueous solution (1:3)
(approx. 15 μL/cm2) to the silanized surface is carried out by
incubating over night (min 16 h) in a water-saturated atmo-
sphere (see Note 6). After thoroughly rinsing with double
distilled water and drying, the prepared chips are stable for
several months.
(b) Polyethylenglycol.
The coupling of diamino- or dicarboxypoly(ethylene glycol)
(PEG) as a 1 mM solution in dichloromethane (DCM)
(approx. 10 μL/cm2) to the silanized surface is achieved by
incubating overnight at 70 C, followed by thorough rinsing
with Milli-Q water and drying in a nitrogen stream.
(c) Change of functional groups.
For some applications (e.g., immobilization of DNA oligo-
mers with an amino-linker) the follow-up functionalization
via peptide chemistry works better with diaminopoly(ethyl-
ene glycol) treated with 5 M glutaric anhydride (GA) in N,N-
dimethylformamide (DMF) (10 μL/cm2) for 6 h to generate
carboxylic groups on the surface. The same protocol can be
used to modify AMD-surfaces with carboxylic groups.
(d) One step chemistry.
Depending on the expected properties of the biopolymer
layer, complex silanes offering chemically beneficial side
chains like 11-aminoundecyltrimethoxysilane can be used in
a one step procedure combining silanization with biopolymer
coating.
3.2.5 Immobilization of (a) Dissolve 1 mg EMCS per 10 μL DMF and apply this solution
Thiol Terminated Ligands to a transducer modified with a biopolymer offering amino
groups at a surface concentration of 10 μL/cm2 for 6–12 h in
a DMF saturated atmosphere (see Note 6). After rinsing with
DMF (see Note 4), dissolve the ligand at a concentration of
1–3 μM in DMF, and apply this solution to the transducers at
a surface concentration of 10 μL/cm2 for 6–12 h as sandwich
pairs in a DMF saturated atmosphere (see Note 6).
(b) Clean the transducers by rinsing them with DMF and Milli-
Q water and drying them in a nitrogen stream (see Note 4).
3.4 Data Evaluation Measuring the concentration of active antibody in a sample work-
ing at mass-transport limited conditions is essential. Accordingly,
3.4.1 Determination of
the rate of diffusion to the surface is much slower than the binding
the Concentration of Active
reaction to the surface. The reaction kinetics can therefore be
Antibody
neglected. Mass-transport limited conditions can be achieved by a
high binding capacity of the surface and a low antibody concentra-
tion in solution. Under mass-transport limited conditions accord-
ing to first Fick’s law, the resulting binding curve is linear, and its
slope is proportional to the concentration of functional antibody. If
all samples contain the same amount of protein, determined by UV
spectroscopy or Bradford assay, it is possible to calculate the active
antibody concentration in a.u. by the different slopes of the sam-
ples, where the sample with the highest slope is assigned the a.u. 1
per μg used protein.
Γðt Þ ¼ ΓEq 1 ekobs t ð4Þ
with.
K aff c Ab
ΓEq ¼ Γmax , ð5Þ
1 þ K aff c Ab
the equilibrium coverage and.
kobs ¼ ka c Ab þ kd , ð6Þ
the observed rate constant.
The affinity constant Kaff is given by the fraction of the associa-
tion rate constant, ka, and the dissociation rate constant, kd.
To obtain accurate values for ΓEq and kobs, it is necessary that
only the kinetics-controlled part of the signal curve is approxi-
mated, and that the surface and bulk are nearly in equilibrium at
the end of the measurement. Now it is possible to determine ka as
the slope of the straight line of the plot with kobs as ordinate and
active antibody concentration as abscissa. In order to determine
Kaff Eq. 5 can be rewritten as
1 1 1 1
¼ þ : ð7Þ
ΓEq Γmax K aff Γmax c Ab
4 Notes
1. Very thick biolayers (more than approx. 100 nm) lead to mea-
surements out of the linear correlation between change of
optical thickness and shift of the interference spectrum.
2. Correct side of the RIfS transducers: mark the un-coated side
with a diamond pen to avoid mistakes during the surface
chemistry steps. The coated side of the transducers appears
colored because of the light interference.
3. In the case a transducer shows a grey shadow after rinsing and
drying repeat this cleaning procedure. If this does not work,
start the surface chemistry from the beginning (otherwise the
modification will not be homogeneous and will show high
nonspecific binding).
4. All surface chemistry protocols which produce highly reactive
groups are sensitive to humidity (deactivation). This can be
Reflectometric Interference Spectroscopy 219
References
Abstract
Antiretroviral treatment can reduce the death rate of human immunodeficiency virus (HIV) infection, and
its effectiveness is maximized at the early stage of HIV infection. The present protocol demonstrates an early
stage high-content HIV diagnosis based on multicolor concurrent monitoring of CD4, CD8, and CD3
coreceptors and F-actin cytoskeleton using quantum dot (Qdot)–antibody conjugates at the single cell
level. Artificial HIV infection of peripheral blood mononuclear cells (PBMCs) can be achieved by treating
PBMCs with gp120. Using the present methodology, we can determine the CD4–CD8 ratios of normal
PBMCs and artificial HIV-infected PBMCs. In addition, this protocol enables monitoring of structural
changes of actin filament alignments in PBMCs bound to gp120 proteins using the multicolor single cell
imaging system. Overall, this approach presents a new model for accurate early stage HIV diagnosis.
Simultaneously the approach provides information on actin cytoskeleton and subtypes of PBMCs as well
as their CD4–CD8 ratios.
Key words High-content imaging, Quantum dots, HIV diagnosis, PBMCs and actin filament
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_14,
© Springer Science+Business Media LLC 2017
221
222 Annie Agnes Suganya Samson and Joon Myong Song
2 Material
3 Methods
Centrifuge
250g, 10 min, 25° C
Whole blood
Plasma layer
PBMC
Histopaque-1077
Histopaque-1077
RBCs PBMC
3.3 Treatment of Monitoring of the CD4–CD8 ratio can greatly improve HIV diag-
PBMCs with nosis accuracy in early stages of infection, and it is important to
Qdot–Antibody accurately determine this ratio. This segment describes a staining
Conjugates protocol using quantum dot (Qdot)–antibody nanoprobes for
quantitative concurrent monitoring of actincytoskeleton, CD3,
CD4, and CD8 in normal and gp120 treated PBMCs at the level
of single cells.
1. Fix the PBMC or gp120-treated (1 106) cells using 3.7%
formaldehyde for 10 min and then wash twice with 5 mL 1
PBS solution (centrifugation at 250 g for 10 min at 4 C).
2. Place the fixed PBMCs in prechilled acetone for 3 min at
20 C and then wash twice with 1 PBS. (centrifugation at
250 g for 10 min at 4 C).
3. Immerse the PBMCs pellet in 200 μL of 1% BSA aqueous
solution.
4. Add 1 μL of each Qdot–antibody (Mouse-antihuman CD3-
Qdot605, mouse-antihuman CD8-Qdot565, and mouse-
antihuman CD4-Qdot655) stock solutions (1:200 dilutions)
into 200 μL of 1% BSA aqueous solution containing 1 106
normal or gp120-treated PBMCs.
5. Incubate the solution for 1 h at room temperature.
6. After 1 h, wash the Qdot stained cells twice with 1 PBS.
7. Finally, incubate the cells with Alexa Fluor 488 phalloidin in 1
PBS for 20 minto detect F-actin filament in PBMCs.
3.4 Detection of CD3, This section describes a system to simultaneously monitor fluores-
CD4, CD8, and F-Actin cence emission of different wavelengths. PBMCs stained with Alexa
filament in PBMCs Fluor 488 phalloidin, antihuman CD3-Qdot605, antihuman CD8-
Qdot565, and antihuman CD4-Qdot655 can be concurrently
observed using this Hypermulticolor single cell imaging system.
This system provides a scanning rate of 60 wavelengths (nm)/min,
which will enable 60 different cellular images perminute. First,
PBMCs on the sample stage are irradiated by UV using a micro-
scope objective lens to excite all the Qdot–antibody conjugates and
Alexa Fluor 488 phalloidin. Fluorescent emission from the PBMCs
are collected using an identical lens, transmitted through an
acousto-optical transmission filter (AOTF), and images detected
as a function of wavelength using a charge-coupled device (CCD
(Fig. 2).
1. Alexa Fluor 488 phalloidin detects F-actin filament structural
changes of PBMCs at 518 nm. The three quantum dot tagged
antibody conjugates (antihuman CD3-Qdot605, antihuman
CD8-Qdot565, and antihuman CD4-Qdot655) are bound to
CD3, CD4, and CD8 coreceptors expressed in the cellular
membrane of the PBMCs.
a
Blood
Treatment No treatment
HIV-1 gp120
Qdot-anbody
conjugates/Alexa
Fluor®488 phalloidin
Sample
b
12
Computer
1 Sample
2 Objecve lens
Exciter
3
Laser
Beam splier 4
10
11 5
CCD camera
9 Prism
Long-pass filter Focusing
AOTF
Lens
8 7 6
Fig. 2 (a) Multicolor single cell imaging. (b) Hypermulticolor single cell imaging detector. The components of
the imaging system are as follows: (1) sample stage, (2) objective lens, (3) beam splitter, (4) laser, (5) prism,
(6) focusing lens, (7) AOTF, (8) filter, (9) undiffracted light, (10) diffracted light, (11) CCD camera, and (12)
computer
228 Annie Agnes Suganya Samson and Joon Myong Song
3.7 Imaging System As mentioned before, the imaging system consists of various com-
ponents (Fig. 3). A mercury arc lamp acts as an illumination source
for cells (sample) in a fluorescence microscope and the laser beam is
purified by an interference filter, allowing it to pass through the
excitation filter and finally, reflected by a dichroic filter. The filtered
laser beam is focused through a microscope objective lens (20,
40, 60) and then onto the sample platform. The quantum dot
tagged probes in the cells (sample) are induced to emit fluorescence
by the filtered laser beam. The fluorescent emission is collected by
the same objective lens, passes through dichroic filter and comes
out of the side port (ø ¼ 25 mm) of the microscope. In order to
reduce the fluorescence beam diameter, the C-mount lens is
attached to the side port of the fluorescence microscope. Then,
the entire beam passes through the AOTF window (10 10 mm).
The nano crystal in the AOTF splits the fluorescence beam into two
separate beams, the diffracted light (at particular wavelength) and
3.8 Data Analysis Quantitative data analysis can be done using different parameters
like optical density, average gray value, Z position, angle, distance,
area, width, image plane, elapsed time, stage label, wavelength,
region label, intensity S/N, and threshold area. Single cell imaging
cytometry can be used for simultaneous monitoring of cellular
events, that is, expression of different cellular markers. Figure 4a
Fig. 4 (a) Concurrent monitoring of CD3, CD4, CD8 coreceptors and F-actin in PBMCs. The top figures were
obtained from normal PBMCs while the bottom figures were obtained from gp120-treated PBMCs. F-actin
images of normal PBMCs were compared with those of gp120-treated PBMCs. The gp120-treated PBMCs
indicated with arrows showed ununiform distribution of F-actin filament. On the other hand normal
PBMCs showed uniform distribution of F-actin filament. (b) Inhibitory effect of ITK on cytoskeleton structure
of PBMCs treated with gp120 glycoprotein. Top cellular images are PBMCs treated with gp120 glycoprotein
(10 μg/mL) while cellular images at the bottom are PBMCs treated with gp120 glycoprotein (10 μg/mL) and then
ITK inhibitor (10 μM). Gp120 glycoprotein-treated PBMCs show uniform actin cytoskeleton by the ITK inhibitor
treatment
Q-Dot Probes for HIV Diagnosis 231
4 Note
References
1. Chun TW, Engel D, Berrey MM, Shea T, Corey 4. Tcherepanova I, Harris J, Starr A, Cleveland J,
L, Fauci AS (1998) Early establishment of a pool Ketteringham H, Calderhead D, Horvatinovich
of latently infected, resting CD4(+) T cells dur- J, Healey D, Nicolette CA (2008) Multiplex RT-
ing primary HIV-1 infection. Proc Natl Acad Sci PCR amplification of HIV genes to create a
U S A 95(15):8869–8873 completely autologous DC-based immunother-
2. Kahn JO, Walker BD (1998) Acute human apy for the treatment of HIV infection. PLoS
immunodeficiency virus type 1 infection. N One 3(1):e1489. doi:10.1371/journal.pone.
Engl J Med 339(1):33–39. doi:10.1056/ 0001489
NEJM199807023390107 5. Kim MJ, Lee SC, Pal S, Han E, Song JM (2011)
3. Schvachsa N, Turk G, Burgard M, Dilernia D, High-content screening of drug-induced cardi-
Carobene M, Pippo M, Gomez-Carrillo M, otoxicity using quantitative single cell imaging
Rouzioux C, Salomon H (2007) Examination cytometry on microfluidic device. Lab Chip 11
of real-time PCR for HIV-1 RNA and DNA (1):104–114. doi:10.1039/c0lc00110d
quantitation in patients infected with HIV-1 6. Tak YK, Song JM (2013) Early stage high-
BF intersubtype recombinant variants. J Virol content HIV diagnosis based on concurrent
Methods 140(1-2):222–227. doi:10.1016/j. monitoring of actin cytoskeleton, CD3, CD4,
jviromet.2006.11.012 and CD8. Anal Chem 85(9):4273–4278.
doi:10.1021/ac303727e
Chapter 15
Abstract
To improve food safety there is a need to develop simple, low-cost sensitive devices for detection of food-
borne pathogens and their toxins. We describe a simple, low-cost webcam-based detector which can be
used for various optical detection modalities, including fluorescence, chemiluminescence, densitometry,
and colorimetric assays. The portable battery-operated CCD-based detection system consists of four
modules: (1) a webcam to measure and record light emission, (2) a sample plate to perform assays, (3) a
light emitting diode (LED) for illumination, and (4) a portable computer to acquire and analyze images. To
demonstrate the technology, we used a cell based assay for fluorescence detection of the activity of the food
borne Shiga toxin type 2 (Stx2), differentiating between biologically active toxin and inactive toxin which is
not a risk. The assay is based on Shiga toxin inhibition of cell protein synthesis measured through inhibition
of the green fluorescent protein (GFP). In this assay, GFP emits light at 509 nm when excited with a blue
LED equipped with a filter at 486 nm. The emitted light is then detected with a green filter at 535 nm.
Toxin activity is measured through a reduction in the 509 nm emission. In this system the level of detection
(LOD) for Stx2 was 0.1 pg/ml, similar to the LOD of commercial fluorometers. These results demonstrate
the utility and potential of low cost detectors for toxin activity. This approach could be readily adapted to
the detection of other food-borne toxins
Key words LED, CCD, Microbial pathogens, Toxin activity, Shiga toxin, GFP, Webcam, Fluores-
cence, Fluorometer
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_15,
© Springer Science+Business Media LLC 2017
233
234 Reuven Rasooly et al.
1.1 Considerations In both CCD and CMOS devices, photons striking the semicon-
for Optical Detector ducting material generate electron–hole pairs, and the resulting
Selection electric charge is processed into electronic signals. However, the
reading of these charges in each device is different. In CCDs, the
charge from each pixel is sequentially transported across the chip to
an analog-to-digital (A/D) converter, which converts the charge
into a digital value. The use of a single converter for all pixels
increases the uniformity of the output—an important factor in
image quality. However, blooming is a problem in CCD sensors
when a part of the image is over-exposed, causing leakage of elec-
trical charge to adjacent pixels whose signals become corrupted.
The low noise associated with a CCD sensor enables longer expo-
sure times for low-level light detection, provided the CCD has a
low thermal noise level relative to the incoming photonic signal.
This typically necessitates the use of a cooling system in order to
minimize temperature and the subsequent thermal noise for CCD
or CMOS sensors.
In most CMOS devices each pixel has its own charge-to-volt-
age conversion and the sensor may include transistors at each pixel,
noise correction, and digitization circuits to convert the output
signal to digital so that the chip outputs digital bits. When light
hits the sensor, noise may result from some photons hitting the
transistor instead of the pixel. By placing the transistors under the
pixels (i.e., back-side illuminated CMOS sensor) this type of noise
can be reduced. This configuration increases the chip complexity,
reducing the area available for light capture. Moreover, because
each pixel is doing its own conversion, uniformity is reduced,
especially at higher temperatures. Because the output of a CMOS
is massively parallel compared to a CCD, it is possible to achieve
higher image acquisition speeds and more efficient use of energy.
CMOS sensors are also immune to the blooming effect.
CCDs utilize global shutters in which the entire imager is reset
before integration in order to remove any residual signal in the
sensor pixels; the light collection starts and ends at exactly the same
time for all pixels with no motion blur. CMOS devices utilize a
rolling shutter in which light is not collected simultaneously by all
of the pixels. Thus the time that light collection starts and ends is
slightly different for each row; the top row of the imager is the first
one to start and finish collecting light. The row-by-row process of
Low-Cost Charged-Coupled Device (CCD) Based Detectors. . . 235
1.2 Optics for Digital The optics for digital detectors are relatively simple, consisting of a
Detectors lens and filters. A good lens for biodetection has a focal length of
4–12 mm at f1.2. Such a wide aperture, wide-angle lens is capable
of imaging a wide field-of-view (FOV) from a short distance,
providing a more compact device configuration. However, to
achieve this wide FOV at a short distance, the portion of the
image captured along the edges of the lens ends up being curved,
resulting in barrel distortion.
In addition to fixed focal length lenses, zoom lenses have
variable focal lengths enabling them to image both wide and nar-
row FOVs. Their main disadvantage is they are slower, depending
on the focal length used (usually operated at f/2.8–f/5.6), than
comparable fixed focal length lenses (e.g., f/1.2), resulting in
longer exposure times. Extension tubes are needed for greater
magnification to achieve macro (i.e., close-up) imaging. The exten-
sion tube contains no optical elements and is placed between the
sensor and the lens. It moves the lens farther from the sensor image
plane and closer to the imaged surface. Alternatively, reversing the
lens so small objects close to the image sensor can be magnified and
projected onto the image sensor [14] can achieve a macro effect
similar to extension tubes for imaging a very small detection surface
at high magnification. There is a trade-off in using extension tubes:
the farther the lens is from the sensor, the greater the loss of light.
Furthermore, extension tubes may not be the only optical element
affecting light intensity. For fluorometry, emission and excitation
filters are also needed that further reduce the optical signal. There-
fore, the choice of lens, extension tube, and filters must all be
compatible in order to image with enough sensitivity over smaller
surface areas on a detection plate.
1.3 Light Sources for Light sources are critical for optical detection, especially for
Optical Detection fluorescence-based detection. Traditional optical detectors utilize
236 Reuven Rasooly et al.
1.3.3 Screens for The screen of a portable device can also be used for effective
Illumination of Portable illumination. The screen enables high intensity uniform illumina-
Devices tion in red, blue and green. Screens for a portable devices such as a
smartphones can be organic LED (OLED) or liquid crystal display
(LCD). The major difference between the two is the use of a
backlight for an LCD display, where the pixels contain liquid crys-
tals that regulate the wavelengths that pass through the pixel result-
ing in the desired color. One advantage of OLEDs over LCDs is the
individual electrical control of each pixel in an OLED reduces
power consumption, since even a “black” screen for an LCD
requires electrical power to control the orientation of the liquid
crystals to prevent any light from passing through, while the OLED
is naturally black without any electrical power. However, the use of
a single backlight for all pixels in an LCD results in greater
uniformity.
238 Reuven Rasooly et al.
2 Materials
2.3 Biological and 1. Stx2 was obtained from Toxin Technology (Sarasota, FL).
Cell Culture Regents 2. Vero cells: African Green Monkey adult kidney cells (ATCC
CCL-81) were obtained from American Type Culture Collec-
tion (Manassas, VA).
3. HEK293 cells (ATCC CRL-1573) were obtained from Ameri-
can Type Culture Collection (Manassas, VA).
4. Dulbecco’s Modified Eagle’s Medium (DMEM) from Gibco
(Carlsbad, CA).
5. Fetal bovine serum (FBS) (Hyclone, Waltham, MA).
6. Fetal calf serum (FCS) (Hyclone, Waltham, MA).
7. Penicillin and streptomycin from Gibco (Carlsbad, CA).
8. Trypsin (Gibco).
9. 96-well plates (Greiner 655,090 obtained from sigma).
2.4 Plate Assay 1. Black 3.2 mm acrylic (Piedmont Plastics, Beltsville, MD).
Material 2. Clear 0.5 mm polycarbonate film (Piedmont Plastics, Beltsville,
MD).
3. 3 M 9770 Adhesive transfer Tape (Piedmont Plastics, Beltsville,
MD).
4. Epilog Legend CO2 65 W cutter (Epilog, Golden, CO).
3 Methods
3.1 Cell Culture Vero cells were maintained at 37 C in a CO2 incubator in Dulbec-
co’s Modified Eagle’s Medium (DMEM) containing 10% fetal
bovine serum and 100 units/ml of both penicillin and streptomy-
cin. Cells were trypsinized when ready to harvest.
3.2 Preparation of Since most of the virus remains associated with infected cells until
High-Titer Viral Stocks very late in the infection process, high-titer viral stocks were
prepared by concentrating the infected cells. Tissue culture flasks
(175 mm) were seeded with HEK293 cells in DMEM containing
10% FCS and 100 units/ml penicillin/streptomycin. When cells
reached 90% confluency, they were infected with the virus at a
multiplicity of infection (MOI) of 10. When the cytopathic effect
was nearly completed (after 48–72 h) and most of the cells were
rounded but not yet detached, the cells were harvested (they were
240 Reuven Rasooly et al.
3.3 Plaque Assays Plaque assays depend on the ability of the adenovirus to propagate
for Purification and in HEK293 cells. Six 35 mm tissue culture plates were seeded with
Titration of the HEK293 cells. The cells were incubated at 37 C in a CO2 incuba-
Adenovirus tor until the cells were 90% confluent. Serial dilutions (10 8–10 13
of the adenovirus stock) were made in DMEM supplemented with
2% FBS. The diluted virus was added to the cells. After 2 h, the
medium was removed and replaced with 1 Modified Eagle’s
Medium and 1% sea-plaque agarose. The agar overlay was added
to keep the virus localized after the cells had lysed. After 5 days,
plaques were visible, and counted for titer determination after
7 days.
3.4 Cell Transduction The cells were transduced with adenovirus coding for GFP as
described in previous work [23] to produce label free cells that
express the GFP gene used for the assay. Vero cells were plated on
black wells 96-well plates at 1 104 cells in 100 μl of medium per
well. Cells were incubated overnight to allow time for cells to attach
to the plate and then the cells were transduced with Ad-GFP
multiplicity of infection (MOI) of 100.
3.5 Shiga Toxin Transduced cells were treated in triplicate with various amounts of
Activity Assay Shiga toxin for 48 h. The toxin activity is measured through a
decrease in fluorescence because the biologically active toxin inhi-
bits protein synthesis. The controls in these experiments are cells
without toxin. After incubation with toxin, the cell fluorescence
was imaged with a green emission filter, and the signals of the wells
were quantified by the CCD camera operating in a still single frame
mode. Various concentrations of the toxins were employed and the
effect of toxin on the cellular fluorescence was measured.
Low-Cost Charged-Coupled Device (CCD) Based Detectors. . . 241
A B
1 400
w
2
300
Magnitude
3 w
200
4
5
100
B G R
0
6 350 400 450 500 550 600 650 700 750
Wave length (nm)
Fig. 1 Fluorescence detector. (A) The main system elements: CCD detector, LED multi-wavelength illuminator
and cell toxicity assay. The CCD detector components: (1) a camera, (2 ) lens, (3 ) emission filter mounted on
the end of the lens, (4 ) assay plate, (5 ) excitation filter, and (6 ) multi-wavelength LED. (B) The spectra
(measured by a spectrometer) for the (W ) white, (B ) blue, (g) green, and (R ) red LEDs
3.6 CCD Imager The basic configuration of the CCD detector platform is shown
schematically in Fig. 1a along with the spectra of the LED illumi-
nator. The system consists of three main elements (a) the CCD
detector module, (b) the assay chip module, and (c) the illumina-
tion/excitation module (see Note 1). All of these elements are
contained within a black acrylic box designed for portability, recon-
figurability, prevention of light loss and internal light reflection, and
to minimize extraneous signals from ambient light.
3.7 Illumination The multi-wavelength spatial LED illuminator module [20] shown
Modules in Fig. 1a6 comprises a custom built multi-wavelength LED illumi-
nation box with two different types of LED: (1) white, which
generates across the full optical spectrum, and (2) RGB, which
generates at the red, green, and blue wavelengths. The LED illumi-
nator contains four RGB LED strips and four white LED strips,
both 18 LEDs per foot. Each LED emission color is controlled
individually with a switch in the front panel. The top of the box
consists of a diffusion panel (milky white plastic panel), which
assures uniformity of the light and hence even illumination of the
sample chip. The dimensions of the diffusion panel are
8.5 5.5 cm. The LEDs are powered by a 3 V DC battery. It is
important to measure the illumination uniformity across the
measured surface (see Note 5).
3.8 Optical System The optical system includes a Pentax 12 mm f1.2 CCD lens. The
fixed focal length enables the use of a wider aperture. The C mount
fixed focal length lens was connected to the imager. The f1.2 lens
permits operation at very low light levels. To minimize green light
emission, which increases the noise and limits the detection
242 Reuven Rasooly et al.
3.9 Assembling the The assembly of the CCD-based fluorometer based on LED illu-
CCD Detector mination was described in previous work [17, 18, 20]. The CCD
imager (Fig. 1a1) is placed inside the light-sealed black acrylic
enclosure (see Note 4). The camera is mounted in the enclosure
with holes drilled in the top of the enclosure to enable air circula-
tion for cooling the CCD. The lens is mounted onto the camera
body (Fig. 1a2) and a green filter (EmF) is mounted on the lens
(Fig. 1a3). The LOC or detection plate (Fig. 1a4), excitation filter
(ExF) (Fig. 1a5), and the LED (Fig. 1a6) are interchangeable (see
Notes 1–7)
3.10 Light Unlike the monochromatic light emitted from lasers, the multi-
Characteristics of the wavelength LED illumination box provides illumination in the
Multi-Wavelength LED blue, green, red, and white ranges (Fig. 1b), covering a spectrum
of 450–650 nm (red 610–650 nm, green 492–550 nm, and blue
450–495 nm) as described in our previous work [20]. The only
region of the spectrum not represented by the RGB LEDs strip is
the range between 550 and 610 nm. The white LED provide broad
illumination spectra (440–680 nm with lower intensity in the range
472–510 nm). The multi-wavelength LED enables illumination in
narrower spectra by using only one of the R, G, B, or white LEDs,
or combinations of them. Within each LED spectrum, interchange-
able filters can be used to narrow the excitation band (see Note 6).
3.11 Image Images were captured using a camera incorporating a CCD sensor
Capturing with high quantum efficiency and response linearity as well as the
ability to generate high-quality, low noise images. The camera used
in this case employed a monochrome sensor, and fluorescent emis-
sion was detected through a green filter. The images were analyzed
and the data quantified with ImageJ [24, 25]. In PBS we observed
no photobleaching when the cells were exposed for less than 5 min.
The longest exposure time employed in our experiments was 20 s.
The average sensor background signal was subtracted from sample
images. This background signal was recorded by capturing images
with exposure and gain settings identical to those used to image
samples, but with a lens cap in place.
3.12 Image and Data The exposure time for the signal will depend on the type of illumi-
Analysis nation and the light emitted, which is determined empirically (see
Note 8). The spot intensity can be analyzed using any standard
image processing software. For our system, we chose the freeware
ImageJ developed at NIH (see Note 8), which enables 2D image
analysis of the intensity of each spot in a row or column (Fig. 2b), as
Low-Cost Charged-Coupled Device (CCD) Based Detectors. . . 243
B
A
A
I + GFP II
III + IV B
V C
1 2 3
C D 40000
Average Pixel Value (ADU)
35000
30000
R² = 0.8596
25000
20000
A 15000
B 3 10000
C 1 2 0.000001 0.001 1
Shiga Toxin Concentration (ng/mL)
Fig. 2 Fluorescence detection of cell toxicity assay: (a) cell toxicity assay (I ) Vero cells transduced with
adenovirus with GFP (II ) the resultant cells that express the GFP (III ) emit light at 509 nm. A toxin effect on
Vero cells (IV ) will reduce light emission (V ) after the addition of the toxin (VI ) measured by CCD detector
shown in (b). (b) Vero-GFP cells response to low concentrations of Shiga toxin. Vero-GFP cells were treated
with various amounts of Shiga toxin. The signals were detected by the CCD operating in a still single frame
mode, the amount of toxin used 3A control no toxin, 3B 0.01 pg/mL, 3C 0.1 pg/mL, 2A 1 pg/mL, 2B 10 pg/mL,
2C 100 pg/mL, 1A 1 ng/mL, 1B 10 ng/mL, 1C 100 ng/mL. The corresponding ImageJ 3D image is shown in (c).
Average signal brightness (ADU) was plotted against the various Shiga toxin concentrations (d)
well as 3D (Fig. 2c) spatial analysis (2D of the spatial position of the
spots and the third dimension is the intensity of the spots), to
provide visual representation of the image as shown in Fig. 2c.
For spot analysis, the intensity value of every spot was exported to
244 Reuven Rasooly et al.
3.13 Assay Plate A 9-well plate (Fig. 1a6) was fabricated as described in our previous
Fabrication work [21, 26, 27], using black acrylic with one side coated by 3 M
9770 black adhesive transfer tape laser machined to have the
needed array of wells (see Note 10 and 11). A layer of thin polycar-
bonate sheet was attached to the adhesive transfer tape to form the
bottom of the sample wells. To minimize light reflection, the
polycarbonate material used for fabrication did not show detectable
autofluorescence, and all the materials used to fabricate the device
did not appear to inhibit enzymatic reactions. The assay plate
fabrication includes:
1. Micromachining: The fluidics was made of 3.2 mm black
acrylic, micromachined with a computer controlled 65 W Epi-
log Legend CO2 laser system (see Notes 9 and 10). The well
holes are 2 mm, which allows analysis of 20 μl samples. Smaller
holes make the loading of the sample less reproducible (see
Notes 10 and 11).
2. Bonding: The polycarbonate bottom was bonded to the acrylic
with double sided pressure-sensitive adhesive transfer tape (see
Notes 12 and 13) which was bonded to the micromachined
layer so there is no contact between the adhesive and the fluids.
Assembled devices were bonded together and processed with a
laminating machine to eliminate air bubbles and improve uni-
formity of the bonding.
3.14 Detection of Transduced cells were treated with various amounts of Shiga toxin
Active SHIGA Toxin in three replicates. As previously mentioned, the toxin activity is
Using CCD Based measured through decreases in fluorescence using controls without
Optical Detector toxin. The cells were cultured for 48 h and after incubation with
toxin the cell fluorescence was detected with the CCD detector.
The plate containing the cells was illuminated by the LED illumi-
nator using the blue LED with a blue excitation filter. The detec-
tion plate was imaged with a green emission filter, and the signals of
the wells were quantified by the CCD camera operating in a still
single frame mode (Fig. 2b). Various concentrations of the toxins
were employed and the effect of toxin on the cellular fluorescence
was measured as shown in Fig. 2b. The amount of toxin used was
well 3A: control no toxin, 3B: 0.01 pg/mL, 3C: 0.1 pg/mL, 2A:
1 pg/mL, 2B: 10 pg/mL, 2C: 100 pg/mL, 1A: 1 ng/mL, 1B:
10 ng/mL, 1C: 100 ng/mL. The corresponding ImageJ 3D image
is shown in Fig. 2c. Average signal brightness measured by the A/D
unit (ADU) was plotted against the various Shiga toxin concentra-
tions (Fig. 2d). The reduction in cell fluorescence signal is
Low-Cost Charged-Coupled Device (CCD) Based Detectors. . . 245
3.15 Detection Limit To measure lower concentrations of toxin, three replicates of the
of Biologically Active transduced Vero-GFP cells were cultured for 24 h and treated with
Stx2 Using CCD various amounts of Shiga toxin (100 ng to 0.01 pg/mL). The
Measurements signals of the wells were detected by the CCD camera. The image
was quantified with ImageJ. The limit of detection (LOD) was
calculated as the mean pixel value in three control samples (cells
with no toxin) minus three times the standard deviation of these
samples. The plotted LOD is indicated as a horizontal dashed line
(Fig. 3). Concentrations of toxin which yielded a mean signal out of
three replicates that was under this limit were considered to be
detected. Unlike the usual LOD calculation, the standard deviation
was subtracted and not added because the toxin reduces signal.
Thus, the LOD was determined to be 0.1 pg/mL, which is similar
to a commercial fluorimeter.
40000
35000
30000
Average Pixel Value (ADU)
25000
20000
15000
10000
5000
0
0.00000010.000001 0.00001 0.0001 0.001 0.01 0.1 1 10 100
Shiga Toxin Concentration (ng/mL)
Fig. 3 Limit of Shiga toxin detection (LOD). Vero-GFP cells were treated with various amounts of Shiga toxin
(100 ng to 0.01 pg/mL). The signals of the wells were detected by the CCD camera. The image was
quantitated with ImageJ The level of detection (LOD) was calculated as the mean pixel value in three control
samples (cells with no toxin) minus three times the standard deviation of these samples. The LOD is indicated
as a horizontal dashed line. Concentrations of toxin which yielded a mean signal out of three replicates that
was under this limit were considered to be detected
4 Notes
1. Make sure the lens, filters, and sample plate are vertically cen-
tered and aligned.
2. Make sure the arrows on the coated filters are facing the CCD.
3. The filters should be leveled
4. The filters should be always in the same position.
5. The imager enclosure must be light sealed without any light
leaks and use light reflecting material. A long exposure (e.g.,
3 min) can be used to detect light leaks.
6. To measure light uniformity, it is important to measure the
light with a long enough exposure time (e.g., 60 s) to measure
the CCD uniformity when all of the wells are loaded with the
same GFP fluorescence sample. If the lighting is not uniform,
correction values for each well can be calculated and use for
measurement composition.
7. To measure the effectiveness of the filters, it is recommended to
perform two long exposures (e.g., 3 min) without the assay
plate, one with the LED on and one with the LED off. Ideally,
the two measurements should be very similar (the blue filters
pass only blue light which is blocked by the green filters). The
difference between measurements may suggest that the blue
filters do not block all green light and/or the green filters do
not block all blue light.
8. For data analysis, use the ImageJ high contrast visualization,
which will not affect the values but will enable easy selection of
the spots
9. A fully open aperture (e.g., f1.2) will enable shorter exposure
time, but because of the limited depth of field, focusing will be
more limited and the image less sharp. The laser power and
speed for cutting polymers has to be determining empirically. It
is recommended to use the minimum laser power to reduce
overheating or burning the material.
10. The “holes” for the wells on the assay plate are 2 mm in
diameter, which allows analysis of 20 μL samples. Smaller
holes make the loading of the sample less reproducible because
248 Reuven Rasooly et al.
References
1. Ligler FS, Taitt CR, Shriver-Lake LC, Sapsford monitoring ions and metabolites 2: color
KE, Shubin Y, Golden JP (2003) Array biosen- responses to pH, K+ and glucose. Anal Sci
sor for detection of toxins. Anal Bioanal Chem 22:937–941
377:469–477 7. Feldstein MJ, Golden JP, Rowe CA, Maccraith
2. Roda A, Manetta AC, Portanti O et al (2003) A BD, Ligler FS (1999) Array biosensor: optical
rapid and sensitive 384-well microtitre format and fluidics systems. Biomed Microdevices
chemiluminescent enzyme immunoassay for 1:139–153
19-nortestosterone. Luminescence 18:72–78 8. Sohn YS, Goodey A, Anslyn EV, McDevitt JT,
3. Svitel J, Surugiu I, Dzgoev A, Ramanathan K, Shear JB, Neikirk DP (2005) A microbead
Danielsson B (2001) Functionalized surfaces array chemical sensor using capillary-based
for optical biosensors: applications to in vitro sample introduction: toward the development
pesticide residual analysis. J Mater Sci Mater of an “electronic tongue”. Biosens Bioelectron
Med 12:1075–1078 21:303–312
4. Liu Y, Danielsson B (2007) Rapid high 9. Knecht BG, Strasser A, Dietrich R, Martlbauer
throughput assay for fluorimetric detection of E, Niessner R, Weller MG (2004) Automated
doxorubicin—application of nucleic acid-dye microarray system for the simultaneous detec-
bioprobe. Anal Chim Acta 587:47–51 tion of antibiotics in milk. Anal Chem
5. Burkert K, Neumann T, Wang J, Jonas U, 76:646–654
Knoll W, Ottleben H (2007) Automated prep- 10. Balsam J, Bruck HA, Rasooly A (2015)
aration method for colloidal crystal arrays of Smartphone-based fluorescence detector for
monodisperse and binary colloid mixtures by mHealth. Methods Mol Biol 1256:231–245
contact printing with a pintool plotter. Lang- 11. Balsam J, Rasooly R, Bruck HA, Rasooly A
muir 23:3478–3484 (2014) Thousand-fold fluorescent signal
6. Tohda K, Gratzl M (2006) Micro-miniature amplification for mHealth diagnostics. Biosens
autonomous optical sensor array for Bioelectron 51:1–7
Low-Cost Charged-Coupled Device (CCD) Based Detectors. . . 249
12. Balsam J, Bruck HA, Rasooly A (2014) 23. Rasooly R, Do PM (2010) Shiga toxin Stx2 is
Webcam-based flow cytometer using wide- heat-stable and not inactivated by pasteuriza-
field imaging for low cell number detection at tion. Int J Food Microbiol 136:290–294
high throughput. Analyst 139:4322–4329 24. Balsam J, Bruck HA, Kostov Y, Rasooly A
13. Balsam J, Bruck HA, Rasooly A (2013) Ortho- (2012) Image stacking approach to increase
graphic projection capillary array fluorescent sensitivity of fluorescence detection using a
sensor for mHealth. Methods 63:276–281 low cost complementary metal-oxide-semicon-
14. Balsam J, Ossandon M, Bruck HA, Lubensky I, ductor (CMOS) webcam. Sens Actuators B
Rasooly A (2013) Low-cost technologies for 171–172:141–147
medical diagnostics in low-resource settings. 25. Balsam J, Ossandon M, Bruck HA, Rasooly A
Expert Opin Med Diagn 7:243–255 (2012) Modeling and design of microma-
15. Sapsford KE, Taitt CR, Loo N, Ligler FS chined optical Soller collimators for lensless
(2005) Biosensor detection of botulinum tox- CCD-based fluorometry. Analyst
oid A and staphylococcal enterotoxin B in 137:5011–5017
food. Appl Environ Microbiol 71:5590–5592 26. Sapsford KE, Francis J, Sun S, Kostov Y,
16. Golden JP, Floyd-Smith TM, Mott DR, Ligler Rasooly A (2009) Miniaturized 96-well
FS (2007) Target delivery in a microfluidic ELISA chips for staphylococcal enterotoxin B
immunosensor. Biosens Bioelectron detection using portable colorimetric detector.
22:2763–2767 Anal Bioanal Chem 394:499–505
17. Kostov Y, Sergeev N, Wilson S, Herold KE, 27. Sun S, Yang M, Kostov Y, Rasooly A (2010)
Rasooly A (2009) A simple portable electrolu- ELISA-LOC: lab-on-a-chip for enzyme-linked
minescence illumination-based CCD detector. immunodetection. Lab Chip 10:2093–2100
Methods Mol Biol 503:259–272 28. Yang M, Kostov Y, Bruck HA, Rasooly A
18. Sapsford KE, Sun S, Francis J, Sharma S, Kostov (2009) Gold nanoparticle-based enhanced
Y, Rasooly A (2008) A fluorescence detection chemiluminescence immunosensor for detec-
platform using spatial electroluminescent exci- tion of Staphylococcal enterotoxin B (SEB) in
tation for measuring botulinum neurotoxin A food. Int J Food Microbiol 133:265–271
activity. Biosens Bioelectron 24:618–625 29. Yang M, Kostov Y, Bruck HA, Rasooly A
19. Higgins JA, Nasarabadi S, Karns JS et al (2003) (2008) Carbon nanotubes with enhanced
A handheld real time thermal cycler for bacte- chemiluminescence immunoassay for CCD-
rial pathogen detection. Biosens Bioelectron based detection of Staphylococcal enterotoxin
18:1115–1123 B in food. Anal Chem 80:8532–8537
20. Sun S, Francis J, Sapsford KE, Kostov Y, 30. Yang M, Kostov Y, Rasooly A (2008) Carbon
Rasooly A (2010) Multi-wavelength Spatial nanotubes based optical immunodetection of
LED illumination based detector for in vitro Staphylococcal enterotoxin B (SEB) in food.
detection of botulinum neurotoxin A activity. Int J Food Microbiol 127:78–83
Sens Actuators B 146:297–306 31. Irawan R, Tjin SC, Yager P, Zhang D (2005)
21. Sun S, Ossandon M, Kostov Y, Rasooly A Cross-talk problem on a fluorescence multi-
(2009) Lab-on-a-chip for botulinum neuro- channel microfluidic chip system. Biomed
toxin a (BoNT-A) activity analysis. Lab Chip Microdevices 7:205–211
9:3275–3281 32. Hawkins KR, Yager P (2003) Nonlinear
22. Rasooly R, Balsam J, Hernlem BJ, Rasooly A decrease of background fluorescence in poly-
(2015) Sensitive detection of active Shiga toxin mer thin-films – a survey of materials and how
using low cost CCD based optical detector. they can complicate fluorescence detection in
Biosens Bioelectron 68:705–711 microTAS. Lab Chip 3:248–252
Chapter 16
Abstract
Incredible progress continues to be made toward development of low-cost nucleic acid-based diagnostic
solutions suitable for deployment in resource-limited settings. Detection components play a vitally impor-
tant role in these systems, but have proven challenging to adapt for operation in a portable format. Here we
describe efforts aimed at leveraging the capabilities of consumer-class smartphones as a convenient platform
to enable detection of nucleic acid products associated with DNA amplification via the polymerase chain
reaction (PCR). First, we show how fluorescence-based detection can be incorporated into a portable
convective thermocycling system controlled by a smartphone app. Raw images captured by the phone’s
camera are processed to yield real-time amplification data comparable to benchtop instruments. Next, we
leverage smartphone imaging to achieve label-free detection of PCR products by monitoring changes in
electrochemical reactivity of embedded metal electrodes as the target DNA concentration increases during
replication. These advancements make it possible to construct rugged inexpensive nucleic acid detection
components that can be readily embedded in a variety of portable bioanalysis instruments.
Key words PCR, Smartphone, Point of care, Label free detection, Mobile health care, Image analysis
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_16,
© Springer Science+Business Media LLC 2017
251
252 Aashish Priye and Victor M. Ugaz
Fig. 1 Microscale convective flow states enable convective PCR. (a) Thermocycling is performed in a
cylindrical reactor whose top and bottom surfaces are maintained at different fixed temperatures, establishing
a flow field that circulates PCR reagents sequentially through denaturing, annealing, and extension conditions.
(b) Reagents are loaded in cylindrical wells machined into polycarbonate rods and sealed, after which the
reactor is subjected to a vertical temperature gradient. (c) A 3D computational fluid dynamics model coupled
with PCR kinetics yields predictions of DNA replication time scales expressed in terms of a characteristic
generation rate (i.e., DNA doubling events per hour) in a Ra-h/d parametric space. The parametric map reveals
a zone in the chaotic flow regime where accelerated DNA replication is stably achievable over a span of two
orders of magnitude in Ra. Insets originating from the map depict that the convective flow field has a strong
chaotic component in lower aspect ratio geometries as compared to higher aspect ratio geometries (quasi-
periodic flow trajectories). Trajectories were obtained from 3D CFD simulations performed over 5 min of
convective flow with a vertically imposed temperature gradient of 40 C
2 Materials
2.1 Convective PCR 1. Convective PCR reactors/tubes: holes are machined by drilling
Experiments into polycarbonate rods (Amazon Supply; Brand name: Small
Parts; Part #: PlasticRod137).
2. Bovine serum albumin (cat. no. A2153; Sigma-Aldrich).
3. Rain-X Anti-Fog (SOPUS Products).
4. SYBR Green PCR Master Mix (2; Cat. no. 4309155, Life
Technologies).
5. AmpliTaq Gold DNA polymerase (included in the SYBR Green
PCR Master Mix).
6. Forward and Reverse primers (10 μM; 50 -
0 0
CTGAGGCCGGGTTATTCTTG-3 and 5 -CGACTGGC-
CAAGATTAGAGA-30 Integrated DNA Technologies).
7. λ-phage template DNA (1 μg/mL; Cat. no. N3011S, New
England Biolabs).
8. PCR grade water.
9. Aluminum tape (cat. no. PCR-AS-200; Axygen, Inc.) and clear
tape.
10. A smartphone with PCR to go app.
11. Clip on mini-microscope attachment for the phone (60 mini
pocket microscope for iPhone 4; supply).
12. Portable battery pack (Vinsic Tulip battery charger).
13. Optical filter (THORLABS; Ø25 mm OG515 Colored Glass
Filter; Part # FGL515).
14. Microcontroller (Atmega328).
15. Ceramic resistor (Wire wound 10 Ω; Mouser electronics).
16. Temperature sensor (TMP35; Mouser electronics).
17. 5 mm round RGB LED, 5 mW.
18. RN41 Bluetooth module (Microchip).
19. Resistors and capacitors.
20. Polydimethylsiloxane (PDMS).
3 Methods
Fig. 2 A portable convective PCR setup. A smartphone enabled portable convective PCR device. (a) Convective
PCR reactor is placed on an isothermal mini hotplate and is illuminated with an LED excitation source to emit
fluorescence signal from the reactor which is then captured by the phone camera for further analysis. (b) The
phone interfaces with the device’s peripheral components (isothermal heater and LED source) wirelessly via
Bluetooth module coupled with an Arduino microcontroller via the RX and TX lines (Universal asynchronous
receiver/transmitter) (c) Device in operation
3.2 PCR to Go: A The PCR to Go application has been developed for both iOS (Xcode
Fluorescence Analysis 5.0) and Android (Android studio) platforms. It provides a user
Smartphone App friendly GUI that sends commands to the detector/heater system
and seamlessly switches between various in-app functions.
1. Once the app is launched it automatically pairs with the periph-
eral Bluetooth module of the detector system. The user can
then set the heater and LED operation parameters.
2. The analysis can either be carried out in end point or real time
detection modes. In each case, the camera exposure time and
focal plane settings are manually fixed with the sample placed
~5 cm away from the camera lens (see Note 3).
3. A clip on mini microscope attachment (TOMTOP Mini Pocket
Microscope Magnifier Jewelers Loupe for iPhone 4 4S with
LED Light (Amazon.com)), supplemented with the ability to
digitally zoom images via the app’s affine transformation frame-
work, enables a total of 60 magnification to resolve an area of
~1 mm2 without pixel degradation (Fig. 3).
4. In real-time detection mode, the user inputs additional para-
meters such as the frequency of image acquisition and the
number of images to be captured. The excitation LED is illu-
minated for 5 s during which the app synchronizes image
capture.
5. Raw image data from the CMOS sensor are stored as bitmap
data files containing 4 bits per pixel (one each for red, blue,
green, and alpha values) corresponding to standard RGB color
space (sRGB).
6. The user can then define a region of interest within the
acquired image using touch gestures.
7. When the analyze button is tapped, the app extracts the
corresponding sRGB byte matrix from the region of interest.
The alpha channel, which measures transparency, is discarded
as it is constant in all images.
Smartphone-Enabled Detection Strategies for Portable PCR–Based Diagnostics 259
Fig. 3 Smartphone-based fluorescence analysis workflow. RGB bitmap data from the cropped raw images first
undergoes a gamma transformation. The green channel intensity of the pixel matrix is then extracted to
analyze the average fluorescence intensities. In end point detection mode pre- and post-PCR images are
compared, while in real time detection mode averaged intensities from a series of acquired images are fit
using a nonlinear sigmoid function to obtain a qPCR curve
I max
I ¼ þ Io
ðtt 1=2 Þ=k
1þe
Fig. 4 Smartphone-based quantitative fluorescence detection (a) The PCR to Go app presents a simple GUI to
control image acquisition, processing, and data analysis (https://itunes.apple.com/us/app/pcr-to-go/
id909227041?mt¼8). (b) Smartphone-based quantification of a 237 bp target sequence from a λ-phage
DNA template is demonstrated for three initial copy numbers ([DNA]0 ¼ 105, 104, and 103 copies/μL). (c)
Sigmoidal fits are applied to the smartphone acquired real time data, and a standard curve is constructed
using reaction times when fluorescence exceeds a threshold value of 20 units (inset, CT ¼ 9.4, 11.8, and
13.3 min for starting DNA concentrations of [DNA]0 ¼ 105, 104, and 103 copies/μL, respectively). (d) A
benchtop real time PCR instrument (LightCycler 96, Roche) generates comparable results with a nearly
identical standard curve
Fig. 5 Label-free detection via electrode dissolution. (a) Microdevice design incorporating a glass micro-
channel (275 45 μm cross section) bonded to a Si substrate patterned with a Cr microelectrode array. When
a 2.5 V potential is applied, the Cr electrodes electrochemically dissolve at a rate dependent on the chemical
composition of the bulk solution (arrows). (b) The evolution of reflected light intensity from the active electrode
displays composition-dependent behavior. The upper panel compares dissolution kinetics of histidine buffer
(50 mM, pH 7.6), a 100 bp dsDNA ladder (20 μg/mL in 50 mM histidine), a reducing agent (0.06% w/v Na2S2O4
in 50 mM histidine), and at low pH (50 mM histidine, pH 4.4). Pre- and post-reaction PCR reagent mixtures are
compared in the lower panel. (c) Electrodes are large enough to enable dissolution to be directly imaged using
an ordinary smartphone (Apple iPhone 4S). Scale: all electrodes are 50 μm wide horizontally
Fig. 6 Integrated label-free convection PCR detection by imaging local electrode dissolution. (a) Addressable
Cu electrodes are patterned on a glass substrate affixed to the top surface of a cylindrical PCR reactor,
enabling electrochemical dissolution to be viewed from above (drawing not to scale). Anodic dissolution
occurs slowly at neutral pH, but is accelerated under alkaline conditions (images of the anode taken after a 3 V
potential was applied for 1 min). (b) In the pre-PCR solution (above), the anode surface is not visibly changed
when a 3 V potential is applied. Dissolution becomes favored (below) as more negatively charged DNA
molecules are generated during PCR, forming an electrophoretically compacted film at the anode. (c) Images
of the electrodes can be analyzed to quantify anodic dissolution before and after PCR, and compared against
appropriate controls. Scale: all electrodes are 500 μm wideSee Polymerase chain reaction (PCR)
4 Notes
References
1. Ozcan A (2014) Mobile phones democratize 7. Tseng D et al (2010) Lensfree microscopy on a
and cultivate next-generation imaging, diag- cellphone. Lab Chip 10:1787–1792
nostics and measurement tools. Lab Chip 8. Yu H, Tan Y, Cunningham BT (2014) Smart-
14:3187–3194 phone fluorescence spectroscopy. Anal Chem
2. Laksanasopin T et al (2015) A smartphone 86:8805–8813
dongle for diagnosis of infectious diseases at 9. Wei Q et al (2014) Imaging and sizing of single
the point of care. Sci Transl Med 7:273re1 DNA molecules on a mobile phone. ACS Nano
3. Zhu H, Isikman SO, Mudanyali O, Green- 8:12725–12733. doi:10.1021/nn505821y
baum A, Ozcan A (2013) Optical imaging 10. Lee S, Oncescu V, Mancuso M, Mehta S, Erick-
techniques for point-of-care diagnostics. Lab son D (2014) A smartphone platform for the
Chip 13:51–67 quantification of vitamin D levels. Lab Chip
4. Erickson D et al (2014) Smartphone technol- 14:1437–1442
ogy can be transformative to the deployment of 11. Oncescu V, Mancuso M, Erickson D (2014)
lab-on-chip diagnostics. Lab Chip Cholesterol testing on a smartphone. Lab
14:3159–3164 Chip 14:759–763
5. Xu X et al (2015) Advances in smartphone- 12. Oncescu V, O’Dell D, Erickson D (2013)
based point-of-care diagnostics. Proc IEEE Smartphone based health accessory for colori-
103:236–247 metric detection of biomarkers in sweat and
6. Breslauer DN, Maamari RN, Switz NA, Lam saliva. Lab Chip 13:3232–3238
WA, Fletcher DA (2009) Mobile phone based 13. Shen L, Hagen JA, Papautsky I (2012) Point-
clinical microscopy for global health applica- of-care colorimetric detection with a smart-
tions. PLoS One 4:e6320 phone. Lab Chip 12:4240–4243
266 Aashish Priye and Victor M. Ugaz
14. San Park T, Li W, McCracken KE, Yoon J-Y 28. Kopp MU, De Mello AJ, Manz A (1998)
(2013) Smartphone quantifies Salmonella from Chemical amplification: continuous-flow PCR
paper microfluidics. Lab Chip 13:4832–4840 on a chip. Science 280:1046–1048
15. Fronczek CF, San Park T, Harshman DK, Nico- 29. West J et al (2002) Application of magnetohy-
lini AM, Yoon J-Y (2014) Paper microfluidic drodynamic actuation to continuous flow
extraction and direct smartphone-based identi- chemistry. Lab Chip 2:224–230
fication of pathogenic nucleic acids from field 30. Priye A et al (2016) Lab-on-a-drone: toward
and clinical samples. RSC Adv 4:11103–11110 pinpoint deployment of smartphone-enabled
16. Gallegos D et al (2013) Label-free biodetection nucleic acid-based diagnostics for mobile
using a smartphone. Lab Chip 13:2124–2132 health care. Anal Chem 88:4651–4560
17. Huang Y-W, Ugaz VM (2013) Smartphone- 31. Ugaz VM, Krishnan M (2004) Novel convec-
based detection of unlabeled DNA via electro- tive flow based approaches for high-throughput
chemical dissolution. Analyst 138:2522–2526 PCR thermocycling. JALA 9:318–323
18. Krishnan M, Ugaz VM, Burns MA (2002) 32. Skandarajah A, Reber CD, Switz NA, Fletcher
PCR in a Rayleigh-Benard convection cell. Sci- DA (2014) Quantitative imaging with a mobile
ence 298:793–793 phone microscope. PLoS One 9:e96906
19. Priye A, Hassan YA, Ugaz VM (2013) Micro- 33. Liu W, Saint DA (2002) Validation of a quanti-
scale chaotic advection enables robust convective tative method for real time PCR kinetics. Bio-
DNA replication. Anal Chem 85:10536–10541 chem Biophys Res Commun 294:347–353
20. Muddu R, Hassan YA, Ugaz VM (2011) Cha- 34. Rutledge R (2004) Sigmoidal curve-fitting
otically accelerated polymerase chain reaction redefines quantitative real-time PCR with the
by Microscale Rayleigh–Bénard convection. prospective of developing automated high-
Angew Chem Int Ed 50:3048–3052 throughput applications. Nucleic Acids Res
21. Yao D-J, Chen J-R, Ju W-T (2007) Micro–- 32:e178
Rayleigh-Bénard convection polymerase chain 35. Huang Y-W, Shaikh F, Ugaz VM (2011) Tun-
reaction system. JM3 6:043007–043009 able synthesis of encapsulated microbubbles by
22. Braun D, Goddard NL, Libchaber A (2003) coupled electrophoretic stabilization and elec-
Exponential DNA replication by laminar con- trochemical inflation. Angew Chem Int Ed
vection. Phys Rev Lett 91:158103 50:3739–3743
23. Hennig M, Braun D (2005) Convective poly- 36. Shaikh F, Ugaz VM (2006) Collection, focus-
merase chain reaction around micro immersion ing, and metering of DNA in microchannels
heater. Appl Phys Lett 87:183901 using addressable electrode arrays for portable
24. Braun D (2004) PCR by thermal convection. low-power bioanalysis. Proc Natl Acad Sci U S
Mod Phys Lett B 18:775–784 A 103:4825–4830
25. Priye A, Muddu R, Hassan YA, Ugaz VM 37. Sari YA, Ginardi RH, Sarno R (2013) Assess-
(2011) Royal Society of Chemistry, Cambridge ment of color levels in leaf color chart using
smartphone camera with relative calibration.
26. H€ uhmer A, Landers J (2000) Noncontact Inform Syst 2:4
infrared-mediated thermocycling for effective
polymerase chain reaction amplification of DNA 38. Yetisen AK, Martinez-Hurtado J, Garcia-
in nanoliter volumes. Anal Chem 72:5507–5512 Melendrez A, da Cruz Vasconcellos F, Lowe
CR (2014) A smartphone algorithm with inter-
27. Pal R et al (2005) An integrated microfluidic phone repeatability for the analysis of colori-
device for influenza and other genetic analyses. metric tests. Sens Actuators B 196:156–160
Lab Chip 5:1024–1032
Chapter 17
Abstract
There is a need for simple and affordable techniques for cytology for clinical applications, especially for
point-of-care (POC) medical diagnostics in resource-poor settings. However, this often requires adapting
expensive and complex laboratory-based techniques that often require significant power and are too massive
to transport easily. One such technique is flow cytometry, which has great potential for modification due to
the simplicity of the principle of optical tracking of cells. However, it is limited in that regard due to the flow
focusing technique used to isolate cells for optical detection. This technique inherently reduces the flow rate
and is therefore unsuitable for rapid detection of rare cells which require large volume for analysis.
To address these limitations, we developed a low-cost, mobile flow cytometer based on streak imaging. In
our new configuration we utilize a simple webcam for optical detection over a large area associated with a
wide-field flow cell. The new flow cell is capable of larger volume and higher throughput fluorescence
detection of rare cells than the flow cells with hydrodynamic focusing used in conventional flow cytometry.
The webcam is an inexpensive, commercially available system, and for fluorescence analysis we use a 1 W
450 nm blue laser to excite Syto-9 stained cells with emission at 535 nm. We were able to detect low
concentrations of stained cells at high flow rates of 10 mL/min, which is suitable for rapidly analyzing larger
specimen volumes to detect rare cells at appropriate concentration levels. The new rapid detection cap-
abilities, combined with the simplicity and low cost of this device, suggest a potential for clinical POC flow
cytometry in resource-poor settings associated with global health.
Key words POC, Webcam, Flow cytometry, Rare cells, Fluidics, Fluorescence detection
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_17,
© Springer Science+Business Media LLC 2017
267
268 Joshua Balsam et al.
1.1 Point-of-Care The development of POC analytical tools has largely focused on
Cytometry lab-on-a-chip (LOC) microfluidic technologies due to their small
size, simplicity, and low manufacturing cost [4, 5]. In addition,
smartphone cameras are being used effectively as optical detection
systems for mobile POC devices based on LOC platforms [6–14].
For example, an optofluidic fluorescent imaging cytometer using a
smartphone camera with a spatial resolution of 2 μm has been
recently described [9, 13]. However, the flow rate for this system
is 1 μL/min, limiting analyses to small volumes. In addition to
smartphones, standalone inexpensive webcam imaging systems
may also overcome limitations of smartphone cameras. For exam-
ple, smartphone cameras often have limited video capabilities (e.g.,
many phones are limited to 30 fps) and have less versatile optical
systems (e.g., inability to change lenses or directly set imaging
parameters). To overcome these limitations we developed a small,
mobile, and low-cost flow cytometer based on webcam imaging
capable of large volume analysis and rare cell detection for a variety
of POC applications.
The main components of current flow cytometers consist of:
[1] a fluidic system for carrying fluorescently labeled biological
material (e.g., cells), and [2] an optical system for fluorescence
detection. Most current flow cytometers utilize microfluidic
sheathing to focus the cells or particles to a channel whose width
permits only a single cell to pass, enabling narrow-field detection
using photomultipliers or other narrow-field photodetectors. This
technique is known as “hydrodynamic focusing.” In one recent
device a passive hydrodynamic flow focusing was achieved
using chevron grooves imbedded on the walls of the microchannel
[15, 16]. This enabled the sheath fluid to completely surround
the sample stream so that cells could be individually interrogated
by the narrow-field fluorescent detector. However, one limitation
of the focused stream is a low flow rate due to the high hydrody-
namic resistance and pressure constraints of the cell, ultimately
limiting the device to small volumes or long analysis times. In
addition, the commonly used photomultipliers used for detection
are delicate, expensive, and difficult to transport.
In general, current flow cytometers using hydrodynamic focus-
ing flow cells are not suitable for POC applications or for rare cell
detection due to their size, expense, lack of durability, small sample
capacity, and high power requirement. An alternative approach to
the focused microfluidic sheathing used in conventional flow
Streak Imaging Flow Cytometer for Rare Cell Analysis 269
1.2 Streak Imaging Using CCD or CMOS imaging sensors for flow cytometry opens
Flow Cytometry up new possibilities for image analysis. One approach is to image
moving cells at long exposure times such that at a given flow rate a
cell moves a distance X in time t to produce a line of pixels described
as a “streak image.” The dimensions of the streak are directly
related to the size of the cell and its velocity. Unlike conventional
imaging aimed of capturing precise sharp image of the cell using
low flow rate and short exposure, streak imaging enables the use of
higher flow rates and long exposure to screen larger volumes of
specimens.
Figure 1a illustrates a cell image captured as a line generated
using a flow rate of 20 mL/min with a frame rate of 20 FPS. Unlike
a spot that would be generated during regular imaging, the line
pattern associated with exposing many pixels during streak imaging
enables easy identification, especially for low quality detectors with
high background noise. The minimum streak length required is
such that at least one pixel measures the maximum possible signal in
order to maximize signal. This streak length is equal to twice the
length of the cell image in pixels before streaking (L) plus one pixel
(i.e., for a cell image of 3 3 pixels, a streak length of 7 is required).
This effect is shown graphically in Fig. 1b, where a cell of length 3
pixels is seen to move a distance of 4 pixels, thereby producing an
image streak with length 7 pixels with a single bright pixel in its
center. An actual cell streak image is shown in Fig. 1c for a similar
cell image size and similar flow conditions, along with a plot of
actual pixel brightness along the center of the cell streak in Fig. 1d.
Cell streaking such as this results in at least one pixel measuring the
maximum possible signal from the cell.
270 Joshua Balsam et al.
d 20
15
Gray Value
10
0 2 4 6 8
Distance (pixels)
2 Materials
2.1 Cell Culture and 1. THP-1 human monocytes cells, ATCC #TIB-202 (ATCC
Fluorescence Staining Manassas, VA).
2. For cell culturing, RPMI-1640 medium supplemented with
10% (v/v) heat-inactivated FBS, 1% antibiotic–antimycotic
solution, and 2 mM glutamine is used.
3. SYTO-9 dye (Molecular Probes Eugene, OR).
4. The cells are cultured in CO2 incubator.
2.3 Flow Cell Fluid 1. 3 mm clear acrylic sheet (Piedmont Plastics, Beltsville MD).
Delivery, and Imaging 2. Quartz glass microscope slide.
3. 3 M 9770 Adhesive transfer Tape (Piedmont Plastics, Beltsville
MD).
4. Nylon rod (McMaster-Carr, Robbinsville, NJ).
5. Fusion100 syringe pump is used for fluid delivery (Chemyx,
Stafford TX).
6. Epilog Legend CO2 65 W laser cutter (Epilog, Golden CO)
used for flow cell fabrication.
272 Joshua Balsam et al.
3 Methods
3.1 Cell Culture and Fluorescently stained THP-1 human monocytes are used as a
Labeling model to simulate rare CTCs. Though monocytes themselves are
not rare, they are diluted to levels similar to those associated with
rare cells.
1. Cells are cultured with RPMI-1640 medium supplemented
with 10% (v/v) heat-inactivated FBS, 1% antibiotic–antimyco-
tic solution, and 2 mM glutamine (see Note 1) in a 5% CO2
environment at 37 C.
2. Cells are removed from an active culture, pelleted by centrifu-
gation and resuspended in deionized water at an approximate
concentration of 106 cells/mL (staining protocol for Syto-9
dye advises against the use of phosphate buffer solutions).
3. To stain cells, 10 μL of Syto-9 dye (3.34 mM stock concentra-
tion) is added to 1 mL of suspended cells and allowed to rest at
room temperature in the dark for 20 min. Washing of cells to
remove excess dye is not necessary due to the low intrinsic
quantum yield when not bound to nucleic acids (<0.01), and
because of the later dilution of this stock solution by factors of
104–106. One known factor which contributed substantially to
the decreasing counting efficiency at lower concentrations is
the cell staining protocol. The protocol recommended by the
dye manufacturer is followed. However, dye which has entered
the cell but not yet bound to DNA is noted to diffuse out of
cells over time, leading to diminishing cell brightness. Because
all dilutions are prepared simultaneously and then analyzed
sequentially from highest to lowest concentration, lower dilu-
tions exhibited cells with lower fluorescent intensity. Allowing
the cells to equilibrate at a low concentration for several hours
would allow this diffusion process time to complete.
Streak Imaging Flow Cytometer for Rare Cell Analysis 273
3.2 Wide-Field Flow A wide-field flow cytometer is developed that is simple, lightweight,
Cytometer for POC and inexpensive. The components are available commercially, and
Applications the key to the design is the integration of the elements in a compact
configuration that is isolated from ambient light to enhance imag-
ing of the fluorescence signal, and which allows for easy placement
and removal of the wide-field flow cell.
1. The flow cytometer for fluorescence detection (Fig. 2) consists
of four main elements: (1) a webcam used as an optical detec-
tor, (2) a laser excitation source for illumination, (3) a wide-
field flow cell, and (4) an optical stage to hold each module in
alignment for stable and clear imaging.
2. For fluorescence detection, a green emission filter with a center
wavelength of 535 nm and bandwidth of 50 nm is used with a
1 W 450 nm blue laser (see Note 1). The optical elements must
be vertically centered (see Note 2).
3. The optical detector consists of the electronics of a webcam
which is disassembled in order to remove the original lens, and
a 12 mm f/1.2 CCTV lens for improved light collection and
image magnification (see Note 3).
4. The fluidics module consists of a flow cell and a programmable
syringe pump. The flow cytometer platform (Fig. 2) consists of
a stationary platform and a moveable stage for focusing using a
screw mechanism.
274 Joshua Balsam et al.
Flow Cytometer
Flow Cell
In from syringe Out to Waste
pump
Movable stage
12mm
CCTV
Lens
Emission
Filter
Computer
Webcam circuit board and
sensor with USB output
3.3 Wide-Field Flow To maximize residence time of cells in the interrogation window
Cell Fabrication and maximize the number of fluorescent photons captured (i.e., to
increase sensitivity), a new high throughput flow cell is designed
using a wide flow channel that increases volumetric flow rates at
lower flow velocities through an orders of magnitude higher cross-
sectional area than used for hydrodynamic focusing.
1. The flow cell (Fig. 3) consists of a 4–20 mm wide flow channel
that maximizes the volume of fluid in the interrogation win-
dow. Fluorescent labeled samples are injected via an inlet
through the channel, excited by a laser in the channel, and
Streak Imaging Flow Cytometer for Rare Cell Analysis 275
Inlet Inlet
A B D
Field of Field of
View View
Outlet Outlet
Fig. 3 Wide-field flow cell—(a) A schematic of the wide-field flow cell with key elements illuminated with spot
laser. (b) An image from the flow cell illuminated with spot laser. (c) A schematic of the wide-field flow cell
with key elements illuminated with line laser illumination. (d) An image from the flow cell illuminated with line
laser illumination
5. Channel depth is set to keep the flow field within the depth of
field of the lens being used. A Pentax CCTV 12 mm f/1.2 is
operated at approximately f/2.4 to reduce field curvature and
improve depth of field.
6. A Fusion100 syringe pump connected to the flow cell is used
for flow rate control. The maximum flow rate achievable
through this flow cell is 10 mL/min. The adhesive may fail at
higher flow rates due to the dynamic pressures required. The
flow rate in experiments presented in this chapter is limited to
500 μL/min due to the maximum frame rate of the webcam
employed for sensing. For very faint cells the flow rate should
be decreased in order to improve sensitivity. Sensitivity also
increases if exposure time is increased to allow cell images to
form short streaks, with length greater than or equal to
the length of the cell image plus one pixel (detailed in Sub-
heading 3.7).
3.4 Optical System The optical system consists of: (1) the blue laser for fluorophore
for Webcam-Based excitation, (2) a webcam detector, and (3) an excitation filter.
Cytometer Platform 1. Laser Excitation: For fluorescent imaging of a wide-field, a
1 W laser is used for illumination in two modes of illumination,
spot illumination and line illumination:
(a) In spot illumination the laser illuminates the flow cell at an
angle of incidence of approximately 45 (see Note 2) at
this angle the laser illuminate a bright elliptical spot which
covered the width of the flow cell (Fig. 3b). The main
advantage of spot illumination is that it is high intensity,
which is suitable for higher flow rates.
(b) In line illumination, the laser illuminate the edge of the
slide an angle of incidence of approximately 45 and the
light illuminate the flow cell through the slide (Fig. 3d)
(c) The 1 W consumer grade laser is fairly expensive (~$300)
for a device designed for use in a low-resource setting. To
reduce the cost of the laser, a lower power laser with line
generator optics could be used to further focus the laser
spot. This could allow the critical parameter of photon flux
to remain unchanged while reducing overall power; how-
ever, this approach may reduce the area analyzed. Alterna-
tively, high power LEDs could be utilized. This would
require the addition of an excitation filter as well as colli-
mating optics, leading to a more complex and potentially
more expensive configuration.
2. Webcam detector and emission filter: A Sony PlayStation®
Eye webcam is used as the photodetector in this platform. The
Sony PlayStation Eye device was disassembled and the main
circuit board (with attached image sensor and USB cable) is
Streak Imaging Flow Cytometer for Rare Cell Analysis 277
3.5 Configuration of The new imaging flow cytometer, the webcam integrated the sen-
Imaging Flow sor into the bottom of the platform below the flow cell, as shown in
Cytometer Fig. 2. It is connected to a 32-bit Windows-based laptop computer
via a USB2 port. The drivers and software controlling the webcam
are developed and freely distributed by Code Laboratories. The
camera control software (CL-Eye Test) enables setting of several
camera parameters (exposure time, frame rate, and gain), and the
capture of video in uncompressed AVI format. The imaging para-
meters for the camera (exposure time and frame rate) depend on
the brightness of the fluorescently labeled cells and the flow rate (see
Note 7). An example of fluorescence labeled cells is the single video
frame of THP-1 stained with SYTO-9 dye in a wide-field flow cell at
a flow rate of 500 μL/min, as shown in Fig. 4.
3.6 Background Frames may contain a signal of interest (e.g., image of a cell),
Signal and Noise interfering background signal (e.g., autofluorescence), and random
Reduction noise. To enhance detection, the constant component of the inter-
fering signal can be subtracted. For background subtraction, the
median value and the maximum value of each pixel in a video file of
the relevant color channel can be calculated. Figure 5 illustrates the
green channel video frames of a video clip containing 2000 frames
(10.7 s of video) showing a single cell passing through the flow cell
shown in different positions along its path. The median image of all
the frames, shown in Fig. 5a, represents the average background
signal with no information from the cells. The maximum image,
shown in Fig. 5b, indicates the highest signal recorded at each pixel
during the video with the cell position in each frame marked. In
order to improve cell detection, especially for lower SNR images
than shown in this example, the median image is subtracted from
the maximum image (Fig. 5c), which allowed for improved visuali-
zation of cell movement by removing background autofluorescence
from the flow cell.
3.7 Noise Reduction Figure 6a shows an actual cell streak image (circled) with flow
in Streak Mode Images direction indicated. Figure 6b shows a close-up of the cell streak
image containing background noise and showing individual pixels
and. In order to reduce noise each column of pixels is averaged
along the streak length n to produce a single averaged row of pixels,
labeled avg(n) in Fig. 6. Figures 6d–i and ii shows a plot of pixel
278 Joshua Balsam et al.
Fig. 4 Cell imaged using wide-field flow cytometer. (a) Single video frame of
fluorescent labeled THP-1 human monocytes in wide-field flow cell at
500 μL/min, and (b) 3D visualization of A
Fig. 6 Noise reduction in streak mode images. (a) A cell streak image (circled) with flow direction indicated. (b)
Close-up of cell streak image showing individual pixels and background noise. In order to reduce noise, each
column of pixels is averaged over the streak length n to produce a single averaged row of pixels, labeled avg
(n). (c) A plot of pixel values before (i) and after (ii) averaging, showing a 3 improvement in SNR. The plot in (i)
is for the row with the brightest pixel value in (d).
Streak Imaging Flow Cytometer for Rare Cell Analysis 281
3.8 Color Channel When using a color camera, spectral filtering can be employed to
Extraction improve SNR by reducing non-specific background light (e.g.,
excitation light). The webcam CMOS sensor employs a typical
Bayer color filter array in which one half of all pixels have a green
filter, 25% have a red filter, and 25% have a blue filter. The emission
profile of Syto-9 dye used in this work has a large overlap in the
green spectrum resulting in this color channel having the highest
SNR. The blue and red channels typically showed SNRs of one half
and one tenth those of the green channel, respectively. For this
reason the red and blue channels are discarded and only the green
channel is analyzed. The “Split Channels” function in ImageJ is
used to divide a color image into its constituent channels.
3.9 Rare Cell Counting of rare cell events is simulated using dilutions of fluores-
Counting Using cently labeled THP-1 monocytes. Dilutions are prepared as
Streaking Mode described in Subheading 1, and then measured using the wide-
Imaging field flow cytometer in Fig. 2. Figure 7a shows results using the
flow cell depicted in Fig. 3a. Dilutions of 100, 10, and 1 cell per mL
respectively yielded average concentrations of 84, 7.9, and
0.56 cells/mL, with 95% confidence bounds of 61–107, 6.9–9,
and 0.16–0.96 cells/mL. Error bars in Fig. 7 represent a 95%
confidence interval.
Following this set of experiments, flow cell geometry and sys-
tem operating parameters (flow rate, frame rate) are optimized
1000
A 1000
B
Measured Cell Count (cells/mL)
100
Measured Cell Count (cells/mL)
100
10
10
1 1
0.1 0.1
0.01 0.01
100 10 1 100 10 1 0.1
Target Cell Count (cells/mL) Target Cell Count (cells/mL)
Fig. 7 Rare cell counting using streaking mode—(a) Results of counting dilutions of rare cells using line laser
illumination. The large error bars evident at the level of 1 cell/mL prompted further optimization of the flow cell
geometry and operating parameters. (b) Results of counting dilutions of rare cells using an updated flow cell
design to improve image SNR by reducing cell velocity. The field-of-view (FOV) was also improved. Following
this optimization, performance of the wide-field flow cytometer improved substantially at the level of 1 cell/mL,
allowing measurement expansion to as low as 0.1 cell/mL. Measurements made via microscopy
(sparsely filled columns) vs. wide-field flow cytometry (dense fill) were not significantly different at the 95%
confidence level
282 Joshua Balsam et al.
3.10 Factors for In order to optimize the performance of a wide-field flow cyt-
Optimization of Streak ometer, the following method for setting various device parameters
Imaging Flow can be used. This will result in maximized cell image SNR, max-
Cytometer imized sample throughput, and accurate flow sampling.
1. The distance between the imaging lens and the flow cell should
be set such that images of cells are projected onto at most a
3 3 array of pixels. Distance should not be so great that a cell
image is less than one pixel in size. At those distances, photon
flux per pixel begins to diminish and SNR drops quickly. When
cells are imaged on more than one pixel, photon flux per pixel is
a constant and independent of the distance between flow cell
and lens. The largest possible distance should be chosen in
order to maximize the field-of-view (FOV). Set the flow cell
width so that it covers the FOV of the image sensor at this lens-
to-flow cell distance. This allows the largest possible flow cell
width to be used, thereby minimizing flow velocity and max-
imizing SNR at a given flow rate.
2. The relationship between average cell velocity and flow rate
should be determined empirically. The maximum velocity
achievable will be determined later and is a function of cell
image signal-to-noise ratio (SNR) (e.g., higher SNR allows
higher velocity). This in turn depends on the cell staining
Streak Imaging Flow Cytometer for Rare Cell Analysis 283
4 Notes
1. Make sure the lens, filters, and flow cell are vertically centered
and aligned and that the laser illuminates the image area of the
flow cell.
Cell cultures with very high density may result in cell anu-
cleation (ghost cells), cells with low fluorescence signal when
labeled with nucleic acid stain.
2. Make sure the arrows on the coated filters are facing the cam-
era, and that if the filters being used have angular dependence
(e.g., interference filters) the excitation source beam is con-
fined to the correct range of angles.
3. A fast focal ratio (e.g., f/1.2) will enable shorter exposure time,
but focusing will be more difficult as the depth of field will be
reduced. Extension tubes are added between the lens and
sensor to allow for close focusing and enlargement of cell
images. This can result in noticeable image field curvature
(i.e., image corners out of focus while image center is in
focus), so spacing and imaging distance must be optimized.
Focal ratio can be reduced to reduce field curvature.
284 Joshua Balsam et al.
References
1. Balsam J, Bruck HA, Rasooly A (2015) Cell 14. Zhu H, Yaglidere O, Su TW, Tseng D, Ozcan
streak imaging cytometry for rare cell detec- A (2011) Cost-effective and compact wide-
tion. Biosens Bioelectron 64:154–160 field fluorescent imaging on a cell-phone. Lab
2. Balsam J, Bruck HA, Rasooly A (2015) Mobile Chip 11(2):315–322
flow cytometer for mHealth. Methods Mol 15. Golden JP, Kim JS, Erickson JS, Hilliard LR,
Biol 1256:139–153 Howell PB, Anderson GP et al (2009) Multi-
3. Balsam J, Bruck HA, Rasooly A (2014) wavelength microflow cytometer using groove-
Webcam-based flow cytometer using wide- generated sheath flow. Lab Chip 9
field imaging for low cell number detection at (13):1942–1950
high throughput. Analyst 139(17):4322–4329 16. Howell PB Jr, Golden JP, Hilliard LR, Erickson
4. Sun S, Ossandon M, Kostov Y, Rasooly A JS, Mott DR, Ligler FS (2008) Two simple and
(2009) Lab-on-a-chip for botulinum neuro- rugged designs for creating microfluidic sheath
toxin a (BoNT-A) activity analysis. Lab Chip 9 flow. Lab Chip 8(7):1097–1103
(22):3275–3281 17. Taitt CR, Anderson GP, Ligler FS (2005) Eva-
5. Sun S, Yang M, Kostov Y, Rasooly A (2010) nescent wave fluorescence biosensors. Biosens
ELISA-LOC: lab-on-a-chip for enzyme-linked Bioelectron 20(12):2470–2487
immunodetection. Lab Chip 10(16):2093–2100 18. Ngundi MM, Qadri SA, Wallace EV, Moore
6. Wei Q, Qi H, Luo W, Tseng D, Ki SJ, Wan Z MH, Lassman ME, Shriver-Lake LC et al
et al (2013) Fluorescent imaging of single (2006) Detection of deoxynivalenol in foods
nanoparticles and viruses on a smart phone. and indoor air using an array biosensor. Envi-
ACS Nano 7(10):9147–9155 ron Sci Technol 40(7):2352–2356
7. Coskun AF, Nagi R, Sadeghi K, Phillips S, 19. Moreno-Bondi MC, Taitt CR, Shriver-Lake
Ozcan A (2013) Albumin testing in urine LC, Ligler FS (2006) Multiplexed measure-
using a smart-phone. Lab Chip 13 ment of serum antibodies using an array biosen-
(21):4231–4238 sor. Biosens Bioelectron 21(10):1880–1886
8. Navruz I, Coskun AF, Wong J, Mohammad S, 20. Ligler FS, Sapsford KE, Golden JP, Shriver-
Tseng D, Nagi R et al (2013) Smart-phone Lake LC, Taitt CR, Dyer MA et al (2007)
based computational microscopy using multi- The array biosensor: portable, automated sys-
frame contact imaging on a fiber-optic array. tems. Anal Sci 23(1):5–10
Lab Chip 13(20):4015–4023 21. Kostov Y, Sergeev N, Wilson S, Herold KE,
9. Zhu H, Ozcan A (2013) Wide-field fluorescent Rasooly A (2009) A simple portable electrolu-
microscopy and fluorescent imaging flow cyto- minescence illumination-based CCD detector.
metry on a cell-phone. JoVE 74:e50451. Methods Mol Biol 503:259–272
10. Zhu H, Sencan I, Wong J, Dimitrov S, Tseng 22. Sapsford KE, Sun S, Francis J, Sharma S, Kos-
D, Nagashima K et al (2013) Cost-effective tov Y, Rasooly A (2008) A fluorescence detec-
and rapid blood analysis on a cell-phone. Lab tion platform using spatial electroluminescent
Chip 13(7):1282–1288 excitation for measuring botulinum neuro-
11. Zhu H, Sikora U, Ozcan A (2012) Quantum toxin A activity. Biosens Bioelectron 24
dot enabled detection of Escherichia coli using (4):618–625
a cell-phone. Analyst 137(11):2541–2544 23. Sun S, Francis J, Sapsford KE, Kostov Y,
12. Zhu H, Yaglidere O, Su TW, Tseng D, Ozcan Rasooly A (2010) Multi-wavelength Spatial
A (2011) Wide-field fluorescent microscopy on LED illumination based detector for in vitro
a cell-phone. Conf Proc IEEE Eng Med Biol detection of Botulinum Neurotoxin A Activity.
Soc 2011:6801–6804 Sens Actuators B 146(1-8):297–306
13. Zhu H, Mavandadi S, Coskun AF, Yaglidere O, 24. Rasooly A, Bruck HA, Kostov Y (2013) An
Ozcan A (2011) Optofluidic fluorescent imag- ELISA Lab-on-a-Chip (ELISA-LOC). Meth-
ing cytometry on a cell phone. Anal Chem 83 ods Mol Biol 949:451–471
(17):6641–6647
286 Joshua Balsam et al.
25. Rasooly A, Kostov Y, Bruck HA (2013) 27. Balsam J, Rasooly R, Bruck HA, Rasooly A
Charged-coupled device (CCD) detectors for (2014) Thousand-fold fluorescent signal
Lab-on-a Chip (LOC) optical analysis. Meth- amplification for mHealth diagnostics. Biosens
ods Mol Biol 949:365–385 Bioelectron 51:1–7
26. Balsam J, Bruck HA, Rasooly A (2013) Capil- 28. Balsam J, Ossandon M, Bruck HA, Lubensky I,
lary array waveguide amplified fluorescence Rasooly A (2013) Low-cost technologies for
detector for mHealth. Sens Actuators B medical diagnostics in low-resource settings.
186:711–717 Expert Opin Med Diagn 7(3):243–255
Chapter 18
Abstract
A portable kinetics fluorometer is developed to detect viable cells which may be contaminating various
samples. The portable device acts as a single-excitation, single-emission photometer that continuously
measures fluorescence intensity of an indicator dye and plots it. The slope of the plot depends on the
number of colony forming units per milliliter. The device uses resazurin as the indicator dye. Viable cells
reduce resazurin to resorufin, which is more fluorescent. Photodiode is used to detect fluorescence change.
The photodiode generated current proportional to the intensity of the light that reached it, and an op-amp
is used in a transimpedance differential configuration to ensure amplification of the photodiode’s signal. A
microfluidic chip is designed specifically for the device. It acts as a fully enclosed cuvette, which enhances the
resazurin reduction rate. In tests, the E. coli-containing media are injected into the microfluidic chip and the
device is able to detect the presence of E. coli in LB media based on the fluorescence change that occurred in
the indicator dye. The device provides fast, accurate, and inexpensive means to optical detection of the
presence of viable cells and could be used in the field in place of more complex methods, i.e., loop-
meditated isothermal amplification of DNA (LAMP) to detect bacteria in pharmaceutical samples (Jimenez
et al., J Microbiol Methods 41(3):259–265, 2000) or measuring the intrinsic fluorescence of the bacterial
or yeast chromophores (Estes et al., Biosens Bioelectron 18(5):511–519, 2003).
Key words Contamination detection device, Resazurin, Resorufin, E. coli detection, Microfluidic
device, Thermal bonding of PMMA
1 Introduction
Each year thousands of people die and millions are infected due to
food, water, or medicine contamination [1]. In order to prevent
most of these poisonings a rapid, precise, and sensitive microbial
detection method is highly desirable [2, 3]. There are many com-
mon methods to detect pathogens and other biologics [4]. Most of
these methods require well-equipped and environmentally stable
laboratories as well as highly trained staff to handle devices and
reagents or antibodies [2]. These methods are hard to apply in the
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_18,
© Springer Science+Business Media LLC 2017
287
288 Mustafa Al-Adhami et al.
–
a O b
N
+
HO O O
Resazurin
Viable Cells
Reduction
Reaction
HO O O
Resorufin
Fig. 1 (a) The reduction of resazurin to resorufin chemical formula. (b) The color transformation when resazurin
is converted to resorufin
2 Materials
3 Methods
Fig. 4 Spatial positioning of the optics and optoelectronics. ExF Excitation Filter,
EmF Emission Filter, PD Photo diode, LED Light emitting diode
3.2 Microfluidic 1. A PMMA sheet of 1.5 mm in thickness is first cut using any
Cassettes available equipment (CNC mill, laser cutter). In order to
remove the internal stresses from the plastic sheet, it is annealed
using a PID controlled oven [25].
2. After the sheet is annealed, its surface is flattened with sand
paper [26], treated with 90% ethanol and immediately covered
with a 0.2 mm thick PMMA sheet acting as the cover sheet
[27].
3. The ethanol treated sheets are then clamped with a flat surface
clamp and placed in the oven at 55 C for 5 min (Fig. 6). After
the oven, the assembly is taken out to cool down and the
clamps are removed [28].
4. After the microfluidic chip is fabricated, holes are drilled on the
side of the device. These holes are then filled with silicone and
left to cure to act like a septum for the microfluidic chip.
Figure 1b shows one of the chip designs (see Note 4) [11].
a b 23.5mm
PMMA Assembly
PMMA
Ethanol
Vent
Removal of Clamps
Fig. 6 (a) Step sequence to bond two PMMA sheets. (b) Schematics of the microfluidics cassette
Rapid Detection of Microbial Contamination Using a Microfluidic Device 295
3.5 Measurement of 1. Once the cassette is in the device, the control program is
the Resazurin started. It continuously measures and displays the fluorescence
Reduction intensity. The control program also calculates the running
value of the slope of the fluorescence intensity change. If the
slope is close to zero (Fig. 8, squares), there is no
296 Mustafa Al-Adhami et al.
Fig. 7 Experimental protocolfor low CFU detection. The dashed box represents the actual measurement
(reprinted from the PDA Journal of Pharmaceutical Science and technology)
4 Notes
Fig. 8 Fluorescene intensity changes in contaminated vs. non-contaminated media. Triangles: Fluorescene
intensity increase in media contaminated with 10,000 CFU. Squares: fluroscene intensity stays practically
constant in sterile LB media. Both measurements performed at room temperature (25 C). (a.u. refers to
arbitrary unit)
Fig. 9 Correlation between the CFU number and the slope of fluorescence
increase. Samples below 10,000 CFU/mL were verified using agar plates while
the other numbers were estimated
References
1. Pharmtech.com (2015) An overview of rapid of human cells for clinical transplantation. Cell
microbial-detection methods|Pharmaceutical Transplant 15(2):169–174
technology. N.p. 2015. Web. 3 June 2015 17. Nagaoka M, Hagiwara Y, Takemura K, Mura-
2. Hoehl MM et al (2012) Rapid and robust kami Y, Li J, Duncan SA, Akaike T (2008)
detection methods for poison and microbial Design of the artificial acellular feeder layer
contamination. J Agric Food Chem 60 for the efficient propagation of mouse embry-
(25):6349–6358 onic stem cells. J Biol Chem 283
3. Hobson NS, Tothill I, Turner AP (1996) (39):26468–26476
Microbial detection. Biosens Bioelectron 11 18. Longhi MP, Wright K, Lauder SN, Nowell
(5):455–477 MA, Jones GW, Godkin AJ, Jones SA, Galli-
4. Fda.gov (2015) Archived BAM method: rapid more AM (2008) Interleukin-6 is crucial for
methods for detecting foodborne pathogens. recall of influenza-specific memory CD4 T
N.p. Web. 23 July 2015 cells. PLoS Pathog 4(2):e1000006
5. Vogel SJ, Tank M, Goodyear N (2013) Varia- 19. Tanaka TQ, Williamson KC (2011) A malaria
tion in detection limits between bacterial gametocytocidal assay using oxidoreduction
growth phases and precision of an ATP biolu- indicator, alamarBlue. Mol Biochem Parasitol
minescence system. Lett Appl Microbiol 58 177(2):160–163
(4):370–375 Web 20. Hudman DA, Sargentini NJ (2013) Resazurin-
6. Folsome CE (1964) Functional transformation based assay for screening bacteria for radiation
in mammalian cell culture systems. Nature 202 sensitivity. Springerplus 2(1):55
(4936):1023–1024 Web 21. Fields RD, Lancaster MV (1993) Dual-
7. Celsis.com (2010) Quality control—microbial attribute continuous monitoring of cell prolif-
testing: rapid microbiological methods in lean eration/cytotoxicity. Am Biotechnol Lab 11
manufacturing. N.p. Web. 23 July 2015 (4):48–50
8. Pettit AC, Kropski JA, Castilho JL, Schmitz JE, 22. Ahmed SA, Gogal RM Jr, Walsh JE (1994) A
Rauch CA, Mobley BC, Wang XJ, Spires SS, new rapid and simple non-radioactive assay to
Pugh ME (2012) The index case for the fungal monitor and determine the proliferation of
meningitis outbreak in the United States. N lymphocytes: an alternative to [3H]thymidine
Engl J Med 367(22):2119–2125 incorporation assay. J Immunol Methods 170
9. Gurramkonda C et al (2014) Fluorescence- (2):211–224
based method and a device for rapid detection 23. Al-Nasiry S, Geusens N, Hanssens M, Luyten
of microbial contamination. PDA J Pharm Sci C, Pijnenborg R (2007) The use of Alamar
Technol 68(2):164–171 Web Blue assay for quantitative analysis of viability,
10. Jimenez L, Smalls S, Ignar R (2000) Use of migration and invasion of choriocarcinoma
PCR analysis for detecting low levels of bacteria cells. Hum Reprod 22(5):1304–1309
and mold contamination in pharmaceutical 24. Kostov Y et al (2014) Portable system for the
samples. J Microbiol Methods 41(3):259–265 detection of micromolar concentrations of glu-
11. Ncbi.nlm.nih.gov 2015 Polymerase chain reac- cose. Measurement Science and Technology 25
tion (PCR). N.p. Web. 23 July 2015 (2):025701 Web
12. Estes C, Duncan A, Wade B, Lloyd C, Ellis W 25. Henderson RM, Selock N, Rao G 2012 Robust
Jr, Powers L (2003) Reagentless detection of and easy microfluidic connections in acrylic.
microorganisms by intrinsic fluorescence. Bio- Chips and Tips. http://blogs.rsc.org/
sens Bioelectron 18(5):511–519 chipsandtips/2012/04/23/robust-and-easy-
macrofluidic-connections-in-acrylic. Accessed
13. Brown NA (1990) Cell based assays. Develop- 16 Sept 2015
mental toxicity assays in vitro. Anal Proc 27
(9):246 Web. 26. Zhu X, Liu G, Guo Y, Tian Y (2007) Study of
PMMA thermal bonding. Microsystem Tech-
14. Bionity.com (2015) Alamarblue® assay for nology 13:403–407
assessment of cell proliferation using the Fluos-
tar OPTIMA. N.p. Web. 23 July 2015 27. Tran H, Wu W, Lee N (2013) Ethanol and UV-
assisted instantaneous bonding of PMMA
15. Boyce ST, Anderson BA, Rodriguez-Rilo HL assemblies and tuning in bonding reversibility.
(2006) Quantitative assay for quality assurance Sens Actuators B 181:955–962
of human cells for clinical transplantation. Cell
Transplant 15(2):169–174 28. Ng S, Tjeung R, Wang Z, Lu A, Rodriguez I,
Rooij N (2007) Thermally activated solvent
16. Boyce ST, Anderson BA, Rodriguez-Rilo HL bonding of polymers. Microsystem Technol-
(2006) Quantitative assay for quality assurance ogy 14:753–759
Chapter 19
Abstract
Quantum dots (QDs) and upconverting nanoparticles (UCNPs) are luminescent nanoparticles (NPs)
commonly used in bioassays and biosensors as resonance energy transfer (RET) donors. The narrow and
tunable emissions of both QDs and UCNPs make them versatile RET donors that can be paired with a wide
range of acceptors. Ratiometric signal processing that compares donor and acceptor emission in RET-based
transduction offers improved precision, as it accounts for fluctuations in the absolute photoluminescence
(PL) intensities of the donor and acceptor that can result from experimental and instrumental variations.
Immobilizing NPs on a solid support avoids problems such as those that can arise with their aggregation in
solution, and allows for facile layer-by-layer assembly of the interfacial chemistry. Paper is an attractive solid
support for the development of point-of-care diagnostic assays given its ubiquity, low-cost, and intrinsic
fluid transport by capillary action. Integration of nanomaterials with paper-based analytical devices (PADs)
provides avenues to augment the analytical performance of PADs, given the unique optoelectronic proper-
ties of nanomaterials. Herein, we describe methodology for the development of PADs using QDs and
UCNPs as RET donors for optical transduction of nucleic acid hybridization. Immobilization of green-
emitting QDs (gQDs) on imidazole functionalized cellulose paper is described for use as RET donors with
Cy3 molecular dye as acceptors for the detection of SMN1 gene fragment. We also describe the covalent
immobilization of blue-emitting UCNPs on aldehyde modified cellulose paper for use as RET donors with
orange-emitting QDs (oQDs) as acceptors for the detection of HPRT1 gene fragment. The data described
herein is acquired using an epifluorescence microscope, and can also be collected using technology such as a
typical electronic camera.
Key words Quantum dots, Upconverting nanoparticles, Resonance energy transfer, Paper-based
bioassays, Nucleic acid hybridization
*
These authors contributed equally to this work.
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_19,
© Springer Science+Business Media LLC 2017
301
302 Samer Doughan et al.
1 Introduction
1.1 Quantum Dots in Quantum dots (QDs) are colloidal semiconductor nanocrystals
Resonance Energy with diameter in the range of 2–10 nm. [1] The two commonly
Transfer reported structural types of QDs are core QDs (e.g., CdSe, CdTe,
and CdS) and core/shell QDs (e.g., CdSe/ZnS, CdSe/CdS, and
CdSexS1x/ZnS) [1]. The core/shell structural type is prevalent in
bioassay development as the shell passivates the optical properties
of QDs that originate from its core. QDs exhibit robust and unique
optical properties that originate from quantum confinement
effects. These properties include narrow (full-width-at-half-maxi-
mum in the range of 25–40 nm), symmetric and size tunable
emission spectra, greater photostability and brightness than organic
fluorophores, high quantum yields and a large extinction coefficient
over a broad range of wavelengths that stretches from the UV
region to their first exciton peak in the visible region [1]. These
properties are well suited for the use of QDs in optical multiplexing
for the development of bioassays. QDs are commercially available
with numerous surface chemistries allowing for easy conjugation of
biorecognition elements for use in bioassays [1].
QDs are popular RET donors due to their high quantum yield
and tunable photoluminescence (PL) spectra. The surface area of
QDs allows for immobilization of multiple recognition elements,
where a single QD can participate in multiple RET events [1]. Our
group has paired green-emitting QDs (gQDs) and red-emitting
QDs (rQDs) with various molecular dyes as RET acceptors in
nucleic acid hybridization assays [2, 3]. While QDs are popular
RET donors, their use as RET acceptors has been limited to time
gated measurements [4], chemiluminescence resonance energy
transfer (CRET) [5] and bioluminescence resonance energy trans-
fer (BRET) [6]. The broad absorbance band of QDs that stretches
into the UV region of the spectrum makes them susceptible to
direct excitation in the process of exciting the RET donor. We
have recently paired QDs as RET acceptors with UCNP donors
[7, 8]. UCNPs are excited in the near-IR and IR region and emit in
the UV to near-IR region of the spectrum. This permits the excita-
tion of the RET donor without the direct excitation of the QDs.
We have previously shown the utility of UCNP/QD RET pair for
bioassay development using analytes such as proteins [7] and
nucleic acids [8].
1.2 Upconverting UCNPs are luminescent nanocrystals that are typically tens of
Nanoparticles as nanometers in size. They exhibit anti-Stokes emission based on
Donors in Resonance the process of upconversion. Low-energy pump photons are accu-
Energy Transfer mulated in multiple long-lived excited-states of lanthanide ions that
are supported in an inert crystal lattice. The subsequent relaxation
to the ground electronic state results in the emission of radiation of
Paper-Based Nucleic Acid Assay 303
1.5 Paper-Based In this chapter, we describe methods for solid-phase nucleic acid
Solid-Phase Nucleic hybridization assays on a paper-based platform using QDs and
Acid Hybridization UCNPs as donors in RET-based transduction scheme. The paper
Assays Using the gQD/ substrates are patterned using a wax printing method and subse-
Cy3 and UCNP/QD RET quently chemically derivatized with functional groups that are suit-
Pairs able for the immobilization of the donor nanoparticles. In case of
the gQD/Cy3 (donor/acceptor) RET pair (Fig. 1a), the paper
substrates are chemically modified with imidazole groups to allow
immobilization of gQDs that are pre-modified with oligonucleo-
tide probes. Subsequent hybridization of the target strand and the
Cy3-labeled reporter strand brought the Cy3 acceptor dye in close
proximity to the surface of immobilized gQDs to allow RET-
306 Samer Doughan et al.
Fig. 1 Illustration of interfacial chemistry for paper-based solid-phase transduction of nucleic acid hybridiza-
tion using the (a) gQD/Cy3 RET pair and the (b) UCNP/QD RET pair. The paper substrates were patterned using
wax printing and the circular hydrophilic paper zones were derivatized with (a) imidazole groups for the
immobilization of QD–probe oligonucleotide conjugates and (b) aldehyde moieties for immobilization of
UCNPs. Sequential addition and hybridization of target and the (a) Cy3-labeled or the (b) QD-labeled reporter
oligonucleotides provided the necessary proximity for RET sensitized emission from the acceptor (a) Cy3 dye
or (b) oQD upon excitation of donor (a) gQDs (emission maximum at 525 nm) or (b) UCNP with (a) a UV or (b) IR
excitation source. Upon a selective hybridization event, the emission PL shifts from the donor to the acceptor
and the modulation of the donor and the acceptor PL serve as an analytical signal
Paper-Based Nucleic Acid Assay 307
2 Materials
Table 1
Probe, target and reporter oligonucleotide sequences used in the hybridization assays with the
gQD/Cy3 and UCNP/QD RET pairs
3 Methods
3.4 Wax Printing and 1. Using an appropriate software, draw a pattern of the paper
Chemical Modification device that is to be printed on the Whatman® cellulose chro-
of Paper matography paper substrates. We use AutoCAD 2012 software
(see Note 11). For the purpose of the work described in this
3.4.1 Wax Printing of chapter, the dimensions of each paper device is 25 mm 60 mm
Paper Substrates and a single sheet of 20 cm 20 cm Whatman® chromatogra-
phy paper substrate comprised 18 paper devices (arranged in a
6 3 array format). We use paper devices that contain 32
circular zones with a diameter of 5 mm (see Note 12), arranged
in a 4 8 array format. We print on bare paper, and the zones
are surrounded by a filling with black ink.
314 Samer Doughan et al.
3.4.2 Chemical 1. Attach a binder clip to the end of a wax-printed paper substrate
Derivatization of Paper in order to suspend the paper device in air. We do so by
Zones with Aldehyde inserting a micropipette tip in the arms of the binder clip
Functionality such that the bottom of the micropipette tip rests inside one
of the holders of the microcentrifuge tray rack (see Note 15).
2. Prepare 1.4 M solution of LiCl by dissolving 30 mg of LiCl in
500 μL of Milli-Q water.
3. Prepare 94 mM solution of NaIO4 by dissolving 10 mg of
NaIO4 in 500 μL of Milli-Q water.
4. Mix the two solutions prepared in steps 2 and 3 in equal
volumes.
5. Spot a 5 μL aliquot of the solution prepared in step 4 onto each
paper zone and incubate the spotted paper devices inside an
oven set at 50 C for 30 min.
6. Repeat step 5.
7. Wash the paper devices three times with Milli-Q water (see
Note 16).
8. Place the washed paper devices on any form of absorbent
blotting paper in order to wick off the excess water.
9. Dry the paper devices inside a vacuum desiccator.
3.6 Calibration Curve 1. Prepare dilutions of the SMN1 FC or HPRT1 FC target in the
of Target DNA concentration range of 20 nM to 15 μM using 50 mM borate
buffered saline (pH 9.2, 100 mM NaCl) (see Note 18).
2. Prepare a dilution of SMN1 Cy3 reporter at 10–15 μM con-
centration or HPRT1 oQD reporter at 2 μM using borate
buffered saline (BB2).
3. Spot 3 μL of borate buffered saline (BB2) onto the paper zones
that belong to the top row of the paper device (see Note 19).
4. Spot a 3 μL aliquot of SMN1 FC or HPRT1 FC target at
various concentrations onto each of the different rows of the
paper device (excluding the first row, see point 3). In order to
collect replicate measurements, the same concentration of the
target can be spotted onto the paper zones that belong to the
same row of the paper device (see Note 20).
5. Allow the targets/sample solutions to incubate on the paper
device for 1 h (see Note 21).
6. Spot a 3 μL aliquot of 10–15 μM solution of SMN1 Cy3
reporter or 2 μM HPRT1 oQD reporter onto each of the
paper zones that have been subjected to the target/sample
hybridization and allow the reporter solution to incubate on
the paper device for 30 min (see Note 22).
7. Wash the paper substrates for 5 min with BB2 using an orbital
shaker by submerging each paper device inside a 50 mL conical
centrifuge tube containing BB2. Add 1% Tween® 20 by volume
to the wash solution when QDs are used as acceptors.
8. Wick off the excess solution from the paper substrates using an
absorbent blotting paper and dry the paper substrates inside a
vacuum desiccator prior to the data collection (see Note 23).
Paper-Based Nucleic Acid Assay 317
Fig. 2 Solid-phase QD-RET transduction of nucleic acid hybridization on paper substrates using the gQD/Cy3
RET pair. (a) Colored digital image and the corresponding pseudo-colored PL images of gQDs (G channel) and
Cy3 (R channel) after R-G-B splitting of the colored digital image with increasing amount of SMN1 FC TGT. The
amounts of SMN1 FC TGT in (i) to (viii) were 0, 0.94, 1.9, 3.8, 7.5, 15, 30, and 45 pmol, respectively. (b)
Normalized PL spectra acquired using the epifluorescence microscope platform showing the assay response
with increasing amount of SMN1 FC TGT. The amounts of SMN1 FC TGT in (i) to (xii) were 0, 0.057, 0.12, 0.23,
0.47, 0.94, 1.9, 3.8, 7.5, 15, 30, and 45 pmol, respectively. (c) Calibration curves showing the RET ratio
response (red) and the R/G ratio response (black) with increasing amount of SMN1 FC TGT. Figure adapted
with permission from Ref. 21. Copyright 2014 American Chemical Society
Fig. 3 Solid-phase UCNP/QD (donor/acceptor) RET transduction of nucleic acid hybridization on paper
substrates. (a) Pseudo-colored microscope images of UCNPs (blue channel), oQDs (orange channel) and an
overlay of both images with increasing amount of HPRT1 FC target. The amounts of HPRT1 FC target ranged
from 15 fmol to 6 pmol. (b) Calibration curve showing the RET ratio with increasing amount of HPRT1 FC target
4 Notes
29. Make sure the drawn circle fits within the dimensions of the
paper zone. The same drawn circle can be dragged from one
paper zone to another in order to analyze the mean PL inten-
sity from each paper zone. This also ensures that the mean
intensity determined from each of the paper zones represents
the same unit area.
30. The second term in Eq. 2 is a correction factor and accounts for
the crosstalk of the gQDs donor intensity in the red channel.
31. This allows the software to track the locations on the paper
from where the signals are collected as an integrated area for
the image collected with the 455/35 nm filter. This can then
be applied to the image collected with the 560/20 nm filter.
Acknowledgments
References
1. Noor MO et al (2014) Building from the 5. Freeman R, Liu XQ, Willner I (2011) Chemi-
“Ground” up: developing interfacial chemistry luminescent and Chemiluminescence Reso-
for solid-phase nucleic acid hybridization assays nance Energy Transfer (CRET) detection of
based on quantum dots and fluorescence reso- DNA, metal ions, and aptamer-substrate com-
nance energy transfer. Coord Chem Rev plexes using hemin/G-quadruplexes and
263:25–52 CdSe/ZnS quantum dots. J Am Chem Soc
2. Noor MO, Krull UJ (2013) Paper-based solid- 133(30):11597–11604
phase multiplexed nucleic acid hybridization 6. Kumar M et al (2011) A rapid, sensitive, and
assay with tunable dynamic range using immo- selective bioluminescence resonance energy
bilized quantum dots as donors in fluorescence transfer (BRET)-based nucleic acid sensing sys-
resonance energy transfer. Anal Chem 85 tem. Biosens Bioelectron 30(1):133–139
(15):7502–7511 7. Doughan S et al (2014) Solid-phase covalent
3. Noor MO, Tavares AJ, Krull UJ (2013) On- immobilization of upconverting nanoparticles
chip multiplexed solid-phase nucleic acid for biosensing by luminescence resonance
hybridization assay using spatial profiles of energy transfer. ACS Appl Mater Interfaces 6
immobilized quantum dots and fluorescence (16):14061–14068
resonance energy transfer. Anal Chim Acta 8. Doughan S, Uddayasankar U, Krull UJ (2015)
788:148–157 A paper-based resonance energy transfer
4. Algar WR et al (2012) Quantum dots as simul- nucleic acid hybridization assay using upcon-
taneous acceptors and donors in time-gated version nanoparticles as donors and quantum
forster resonance energy transfer relays: charac- dots as acceptors. Anal Chim Acta 878:1–8
terization and biosensing. J Am Chem Soc 134 9. DaCosta MV et al (2014) Lanthanide upcon-
(3):1876–1891 version nanoparticles and applications in
326 Samer Doughan et al.
bioassays and bioimaging: a review. Anal Chim 19. Martinez AW et al (2010) Diagnostics for the
Acta 832:1–33 developing world: microfluidic paper-based
10. Ju Q, Uddayasankar U, Krull U (2014) Paper- analytical devices. Anal Chem 82(1):3–10
based DNA detection using lanthanide-doped 20. Yetisen AK, Akram MS, Lowe CR (2013)
LiYF4 upconversion nanocrystals as bioprobe. Paper-based microfluidic point-of-care diag-
Small 10(19):3912–3917 nostic devices. Lab Chip 13(12):2210–2251
11. Zhou F, Krull UJ (2014) Spectrally matched 21. Noor MO, Krull UJ (2014) Camera-based
duplexed nucleic acid bioassay using two-colors ratiometric fluorescence transduction of
from a single form of upconversion nanoparti- nucleic acid hybridization with reagentless sig-
cle. Anal Chem 86(21):10932–10939 nal amplification on a paper-based platform
12. Algar WR, Tavares AJ, Krull UJ (2010) Beyond using immobilized quantum dots as donors.
labels: a review of the application of quantum Anal Chem 86(20):10331–10339
dots as integrated components of assays, biop- 22. Watterson JH et al (2004) Rapid detection of
robes, and biosensors utilizing optical trans- single nucleotide polymorphisms associated
duction. Anal Chim Acta 673(1):1–25 with spinal muscular atrophy by use of a reus-
13. Algar WR, Krull UJ (2009) Interfacial trans- able fibre-optic biosensor. Nucleic Acids Res
duction of nucleic acid hybridization using 32(2):1–9
immobilized quantum dots as donors in fluo- 23. Noor MO, Shahmuradyan A, Krull UJ (2013)
rescence resonance energy transfer. Langmuir Paper-based solid-phase nucleic acid hybridiza-
25(1):633–638 tion assay using immobilized quantum dots as
14. Algar WR, Krull UJ (2009) Toward a multi- donors in fluorescence resonance energy trans-
plexed solid-phase nucleic acid hybridization fer. Anal Chem 85(3):1860–1867
assay using quantum dots as donors in fluores- 24. Pong BK, Trout BL, Lee JY (2008) Modified
cence resonance energy transfer. Anal Chem 81 ligand-exchange for efficient solubilization of
(10):4113–4120 CdSe/ZnS quantum dots in water: a procedure
15. Robelek R et al (2004) Multiplexed hybridiza- guided by computational studies. Langmuir 24
tion detection of quantum dot-conjugated (10):5270–5276
DNA sequences using surface plasmon 25. Algar WR et al (2011) The controlled display
enhanced fluorescence microscopy and spec- of biomolecules on nanoparticles: a challenge
trometry. Anal Chem 76(20):6160–6165 suited to bioorthogonal chemistry. Bioconjug
16. Martinez AW et al (2007) Patterned paper as a Chem 22(5):825–858
platform for inexpensive, low-volume, portable 26. Sapsford KE et al (2013) Functionalizing
bioassays. Angew Chem Int Ed 46 nanoparticles with biological molecules: devel-
(8):1318–1320 oping chemistries that facilitate nanotechnol-
17. Carrilho E, Martinez AW, Whitesides GM ogy. Chem Rev 113(3):1904–2074
(2009) Understanding wax printing: a simple 27. Algar WR, Krull UJ (2008) Quantum dots as
micropatterning process for paper-based donors in fluorescence resonance energy trans-
microfluidics. Anal Chem 81(16):7091–7095 fer for the bioanalysis of nucleic acids, proteins,
18. Martinez AW et al (2008) FLASH: a rapid and other biological molecules. Anal Bioanal
method for prototyping paper-based microflui- Chem 391(5):1609–1618
dic devices. Lab Chip 8(12):2146–2150
Chapter 20
Abstract
This chapter describes two different methodologies used to improve the analytical performance of colori-
metric paper-based biosensors. Microfluidic paper-based analytical devices (μPADs) have been produced by
a stamping process and CO2 laser ablation and modified, respectively, through an oxidation step and
incorporation of silica nanoparticles on the paper structure. Both methods are employed in order to
overcome the largest problem associated with colorimetric detection, the heterogeneity of the color
distribution in the detection zones. The modification steps are necessary to improve the interaction
between the paper surface and the selected enzymes. The enhanced performance has ensured reliability
for quantitative analysis of clinically relevant compounds.
Key words Oxidation process, Nanoparticle, Colorimetric detection, Clinical diagnostic, Urinalysis
test
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_20,
© Springer Science+Business Media LLC 2017
327
328 Ellen Flávia Moreira Gabriel et al.
2 Materials
2.5 Artificial Urine 1. Artificial urine is prepared at pH 6.0 and consists of 2 mM citric
Sample acid (Sigma-Aldrich; St. Louis, MO, USA), 25 mM sodium
bicarbonate (Sigma-Aldrich; St. Louis, MO, USA), 170 mM
urea (Sigma-Aldrich; St. Louis, MO, USA), 2.5 mM calcium
chloride (E.M. Science; Gibbstown, NJ, USA), 90 mM sodium
chloride (Mallinckrodt Baker; Center Valley, PA, USA), 2 mM
magnesium sulfate (Mallinckrodt Baker; Center Valley, PA,
USA), 10 mM sodium sulfate (Sigma-Aldrich; St. Louis, MO,
USA), 7 mM sodium phosphate monobasic anhydrous (Fisher
Scientific; Waltham, MA, USA), 7 mM sodium phosphate
dibasic anhydrous (Fisher Scientific; Waltham, MA, USA),
and 25 mM ammonium chloride (Sigma-Aldrich; St. Louis,
MO, USA). This solution is stored in the refrigerator (4 C)
until use.
Colorimetric Biosensing on Paper Microfluidic Platforms 331
3 Methods
Fig. 1 Scheme showing the fabrication process of μPADs based on stamping process. In (a), a paraffinized
paper is placed over the native paper surface; in (b), a metal stamp preheated at 150 C is brought into contact
with the layered paper pieces; in (c), it is presented a typical μPAD fabricated by the stamping method. The
optical micrograph in (d) depicts a real image showing the stamped μPAD. Reproduced from ref. 4 with
permission
332 Ellen Flávia Moreira Gabriel et al.
3.1.2 CO2 Laser System 1. Laser cutting is also selected out of the many techniques used
to fabricate μPADs due to its simplicity of being a straightfor-
ward single step and quick process.
2. A schematic representation of the laser cutting step can be seen
in Fig. 2.
3. The μPAD is first designed using CorelDraw™ X6 Software
with dimensions of a 1.25 mm main channel that branches into
three channels of the same width leading to three 5 mm detec-
tion circles on each end.
4. The design is sent to the laser engraver and cut at 600-dpi
resolution using 30% power and 30% speed. A steady flow of
N2 gas impinged at the engraving point to minimize the risk of
ignition.
5. The laser cuts the paper and define the geometry on the surface
by the ablation process.
6. The laser cuts at a rate of approximately 5 s per μPAD.
Fig. 2 Procedure for fabricatin μPADs by CO2 laser process. (a) CO2 laser machine. (b) Scheme showing the
laser cut of a paper sheet to produce paper biosensor devices. All μPADs were cut using the vector mode, at
30% speed (of a maximum linear speed of 1.65 cm/s) and 30% power (of a maximum intensity of 30 W)
Colorimetric Biosensing on Paper Microfluidic Platforms 333
OH
OH
NaIO4 O
O O
O O
O O
HO O
OH
EDC-NHS
OH OH
O Enz-NH2 O O
O
O O
O O
O O
HN O N O
Enz
Fig. 3 Reactional scheme showing the modification process on paper surface with NaIO4 to obtain a paper
biosensor. Adapted from ref. 17 with permission
3.2.2 Incorporation of Silica nanoparticles (SiO2NP) are also employed to modify the
Silica Nanoparticle surface of the paper in order to accomplish a uniform distribution
(SiO2NP) of the enzyme, therefore increasing the color homogeneity in the
detection zones. The SiO2NP incorporated on detection zone of
μPAD can been seen in Fig. 4.
1. First, a silane-coupling reaction is employed to introduce
amine groups onto the surface of the nanoparticles. 3.3 mg of
15 nm SiO2NP are added to a 5% (v/v) solution of APTES in
ethanol.
2. The length of time the nanoparticles remained in contact with
the APTES solution is investigated from 1 to 24 h due to the
variables that can affect the density of the amine groups, and
ultimately the adsorption of the enzymes to the modified
μPAD.
3. The incorporation of SiO2NP on μPADs is realized by two
different procedures: dispensing the nanoparticle suspension
334 Ellen Flávia Moreira Gabriel et al.
Fig. 4 Scheme of modification process with SiO2NP. Scanning electron micrograph images show the silica
incorporated on cellulose surface. The micrograph images was reproduced from ref. 5 with permission
3.3 Enzymatic The enzymatic assays are performed by adding specific color
Procedures reagents for each bioassay on the detection zones. The processes
are described below.
3.3.1 μPADs—Oxidation
Process 1. For the glucose assay, each detection zone is spotted with
0.75 μL of solution 1 and dried at room temperature for
10 min. Then, 0.75 μL of solution 4 is spotted in each detec-
tion zone and allowed to dry at room temperature for 10 min.
2. For the uric acid assay, each detection zone is spotted with
0.75 μL of solution 2 and dried at room temperature for
10 min. Afterwards, 0.75 μL of solution 7 is added in each
detection zone and allowed to dry at room temperature for the
same time used previously.
3.3.2 μPADs—SiO2NP 1. For the lactate assay, each detection zone is spotted with 1 μL
Incorporation of solution 2 and dried at room temperature for 15 min. After-
wards, the zone is spotted with 1 μL of solution 5 and dried for
the same duration as before.
2. For the glucose assay, the detection zone is spotted with 1 μL of
solution 1 and dried at room temperature for 15 min. After-
wards, the zone is spotted with 1 μL of solution 4 and dried for
additional 15 min.
3. For the glutamate assay, the detection zone is spotted with 1 μL
of solution 3 and dried at room temperature for 15 min. Then
Colorimetric Biosensing on Paper Microfluidic Platforms 335
4.1 Oxidation with 1. The oxidation process is used for the conversion of the
NaIO4 hydroxyl groups on paper surface to aldehyde groups. Then,
these groups are chemically activated with an EDC/NHS solu-
tion enabling the covalent coupling with different enzymes.
2. The conversion process is monitored by infrared spectroscopy.
The technique confirmed the presence of aldehyde groups on
paper surface after the oxidation step. As it can be observed in
Fig. 5, the specific band at 1727 cm1, characteristic of alde-
hyde groups, is found only on the oxidized paper.
336 Ellen Flávia Moreira Gabriel et al.
% Reflectance
% Reflectance
40 40
30 30
20 20
10 10
0 0
4000 3500 3000 2500 2000 1500 1000 500 2500 2250 2000 1750 1500 1250 1000
-1 -1
Wavenumbers (cm ) Wavenumbers (cm )
Fig. 5 Characterization of native and oxidized paper by Fourier transform infrared spectroscopy. Reproduced
from ref. 4 with permission
Fig. 6 Optical micrographs showing the improvements on the color uniformity for glucose (left images) and
uric acid assays (right images) before (a, b) and after (c, d) chemical oxidation on paper surface. Reproduced
from ref. 4 with permission
30
195
Color Intensity (AU)
25
180
165 20
150 15
0 5 10 15 20 25
Time (hours)
Fig. 7 Effect of modification time of silica nanoparticles with APTES on the color intensity and gradient.
Reproduced from ref. 5 with permission
338 Ellen Flávia Moreira Gabriel et al.
Fig. 8 Optical images showing the colorimetric assays for glucose, lactate and glutamate assay with and
without nanoparticle. Reproduced from ref. 5 with permission
4.3 Clinical Assay 1. μPADs modified with SiO2NP have been explored for the
with Artificial Urine detection of glucose, lactate, and glutamate in artificial urine
Sample samples.
2. Once demonstrated individual assays on modified μPADs, all
three assays (glutamate, lactate, and glucose) are simultaneously
Colorimetric Biosensing on Paper Microfluidic Platforms 339
Fig. 9 Optical image showing the analysis of an artificial urine sample spiked
with lactate, glucose, and glutamate on the proposed paper based biosensor.
Reproduced from ref. 5 with permission
5 Notes
References
1. Parolo C, Merkoci A (2013) Paper-based 11. Coltro WKT, de Santis Neves R, de Jesus
nanobiosensors for diagnostics. Chem Soc Motheo A, Da Silva JAF, Carrilho E (2014)
Rev 42:450–457 Microfluidic devices with integrated dual-
2. Nery EW, Kubota LT (2013) Sensing capacitively coupled contactless conductivity
approaches on paper-based devices: a review. detection to monitor binding events in real
Anal Bioanal Chem 405:7573–7595 time. Sens Actuators B 192:239–246
3. Martinez AW, Phillips ST, Carrilho E, Thomas 12. Delaney TL, Zimin D, Rahm M, Weiss D,
SW III, Sindi H, Whitesides GM (2008) Sim- Wolfbeis OS, Mirsky VM (2007) Capacitive
ple telemedicine for developing regions: cam- detection in ultrathin chemosensors prepared
era phones and paper-based microfluidic by molecularly imprinted grafting photopoly-
devices for real-time, off-site diagnosis. Anal merization. Anal Chem 79:3220–3225
Chem 80:3699–3707 13. Gamby J, Abid J-P, Abid M, Ansermet J-P,
4. Garcia PT, Cardoso TMG, Garcia CD, Car- Girault H (2006) Nanowires network for bio-
rilho E, Coltro WKT (2014) A handheld molecular detection using contactless imped-
stamping process to fabricate microfluidic ance tomoscopy technique. Anal Chem
paper-based analytical devices with chemically 78:5289–5295
modified surface for clinical assays. RSC Adv 14. Lima RS, Piazzetta MH, Gobbi AL,
4:37637–37644 Rodrigues-Filho UP, Nascente PA, Coltro
5. Evans E, Gabriel EFM, Benavidez TE, Coltro WK, Carrilho E (2012) Contactless conductiv-
WKT, Garcia CD (2014) Modification of ity biosensor in microchip containing folic acid
microfluidic paper-based devices with silica as bioreceptor. Lab Chip 12:1963–1966
nanoparticles. Analyst 139:5560–5567 15. Martinez AW, Phillips ST, Butte MJ, White-
6. Gomes HI, Sales MGF (2015) Development sides GM (2007) Patterned paper as a platform
of paper-based color test-strip for drug for inexpensive, low-volume, portable bioas-
detection in aquatic environment: application says. Angew Chem Int Ed 46:1318–1320
to oxytetracycline. Biosens Bioelectron 16. Martinez AW, Phillips ST, Whitesides GM,
65:54–61 Carrilho E (2009) Diagnostics for the develop-
7. Sicard C, Glen C, Aubie B, Wallace D, ing world: microfluidic paper-based analytical
Jahanshahi-Anbuhi S, Pennings K, Daigger devices. Anal Chem 82:3–10
GT, Pelton R, Brennan JD, Filipe CD (2015) 17. Pelton R (2009) Bioactive paper provides a
Tools for water quality monitoring and low-cost platform for diagnostics. Trends Anal
mapping using paper-based sensors and cell Chem 28:925–942
phones. Water Res 70:360–369 18. Martinez AW, Phillips ST, Wiley BJ, Gupta M,
8. Zhang Y, Zuo P, Ye B-C (2015) A low-cost and Whitesides GM (2008) FLASH: a rapid
simple paper-based microfluidic device for method for prototyping paper-based microflui-
simultaneous multiplex determination of dif- dic devices. Lab Chip 8:2146–2150
ferent types of chemical contaminants in food. 19. Klasner S, Price A, Hoeman K, Wilson R, Bell
Biosens Bioelectron 68:14–19 K, Culbertson C (2010) Paper-based micro-
9. Chung J, Kim S, Bernhardt R, Pyun J (2005) fluidic devices for analysis of clinically relevant
Application of SPR biosensor for medical diag- analytes present in urine and saliva. Anal Bioa-
nostics of human hepatitis B virus (hHBV). nal Chem 397:1821–1829
Sens Actuators B 111:416–422 20. Abe K, Suzuki K, Citterio D (2008) Inkjet-
10. Lin Z, Chen L, Zhang G, Liu Q, Qiu B, Cai Z, printed microfluidic multianalyte chemical
Chen G (2012) Label-free aptamer-based elec- sensing paper. Anal Chem 80:6928–6934
trochemical impedance biosensor for 17β- 21. Wang J, Monton MRN, Zhang X, Filipe CDM,
estradiol. Analyst 137:819–822 Pelton R, Brennan JD (2014) Hydrophobic
Colorimetric Biosensing on Paper Microfluidic Platforms 341
sol-gel channel patterning strategies for paper- biosensor for simultaneous determination of
based microfluidics. Lab Chip 14:691–695 glucose and uric acid. Lab Chip 11:1286–1291
22. Carrilho E, Martinez AW, Whitesides GM 32. Wleklinski M, Li Y, Bag S, Sarkar D, Narayanan
(2009) Understanding wax printing: a simple R, Pradeep T, Cooks RG (2015) Zero volt
micropatterning process for paper-based paper spray ionization and its mechanism.
microfluidics. Anal Chem 81:7091–7095 Anal Chem 87:6786–6793
23. Nie J, Liang Y, Zhang Y, Le S, Li D, Zhang S 33. Bag S, Hendricks PI, Reynolds JC, Cooks RG
(2013) One-step patterning of hollow micro- (2015) Biogenic aldehyde determination by
structures in paper by laser cutting to create reactive paper spray ionization mass spectrom-
microfluidic analytical devices. Analyst etry. Anal Chim Acta 860:37–42
138:671–676 34. Apilux A, Siangproh W, Praphairaksit N, Chai-
24. Evans E, Gabriel EFM, Coltro WKT, Garcia lapakul O (2012) Simple and rapid colorimetric
CD (2014) Rational selection of substrates to detection of Hg(II) by a paper-based device
improve color intensity and uniformity on using silver nanoplates. Talanta 97:388–394
microfluidic paper-based analytical devices. 35. Wang H, Li Y-j, Wei J-f, Xu J-r, Wang Y-h,
Analyst 139:2127–2132 Zheng G-x (2014) Paper-based three-
25. Zhang Y, Zhou C, Nie J, Le S, Qin Q, Liu F, Li dimensional microfluidic device for monitoring
Y, Li J (2014) Equipment-free quantitative of heavy metals with a camera cell phone. Anal
measurement for microfluidic paper-based ana- Bioanal Chem 406:2799–2807
lytical devices fabricated using the principles of 36. Salles M, Meloni G, de Araujo W, Paixão T
movable-type printing. Anal Chem (2014) Explosive colorimetric discrimination
86:2005–2012 using a smartphone, paper device and chemo-
26. Dungchai W, Chailapakul O, Henry CS (2009) metrical approach. Anal Methods
Electrochemical detection for paper-based 6:2047–2052
microfluidics. Anal Chem 81:5821–5826 37. Souza FR, Duarte-Junior GF, Garcia PT, Col-
27. Noiphung J, Songjaroen T, Dungchai W, tro WKT (2014) Avaliação de dispositivos de
Henry CS, Chailapakul O, Laiwattanapaisal W captura de imagens digitais para detecção col-
(2013) Electrochemical detection of glucose orimétrica em microzonas impressas. Quim
from whole blood using paper-based microflui- Nova 37:1171–1176
dic devices. Anal Chim Acta 788:39–45 38. Gabriel EFM, Coltro WKT, Garcia CD (2014)
28. Santhiago M, Wydallis JB, Kubota LT, Henry Fast and versatile fabrication of PMMA micro-
CS (2013) Construction and electrochemical chip electrophoretic devices by laser engraving.
characterization of microelectrodes for Electrophoresis 35:2325–2332
improved sensitivity in paper-based analytical 39. Dungchai W, Chailapakul O, Henry CS (2010)
devices. Anal Chem 85:5233–5239 Use of multiple colorimetric indicators for
29. Yamada K, Takaki S, Komuro N, Suzuki K, paper-based microfluidic devices. Anal Chim
Citterio D (2014) An antibody-free microflui- Acta 674:227–233
dic paper-based analytical device for the deter- 40. Lin C-C, Tseng C-C, Chuang T-K, Lee D-S,
mination of tear fluid lactoferrin by Lee G-B (2011) Urine analysis in microfluidic
fluorescence sensitization of Tb3+. Analyst devices. Analyst 136:2669–2688
139:1637–1643 41. Ragginer C, Lechner A, Bernecker C, Horejsi
30. Delaney JL, Hogan CF, Tian J, Shen W (2011) R, Moller R, Wallner-Blazek M, Weiss S, Faze-
Electrogenerated chemiluminescence detec- kas F, Schmidt R, Truschnig-Wilders M, Gru-
tion in paper-based microfluidic sensors. Anal ber HJ (2012) Reduced urinary glutamate
Chem 83:1300–1306 levels are associated with the frequency of
31. Yu J, Ge L, Huang J, Wang S, Ge S (2011) migraine attacks in females. Eur J Neurol
Microfluidic paper-based chemiluminescence 19:1146–1150
Chapter 21
Abstract
A smartphone-based colorimetric reader (SBCR), comprising a Samsung Galaxy SIII mini, a gadget (iPAD
mini, iPAD4, or iPhone 5s) and a custom-made dark hood and base holder assembly, is used for human C-
reactive protein (CRP) immunoassay. A 96-well microtiter plate (MTP) is positioned on the gadget’s
screensaver to provide white light-based bottom illumination only in the specific regions corresponding to
the well’s bottom. The images captured by the smartphone’s back camera are analyzed by a novel image
processing algorithm. Based on one-step kinetics-based human C-reactive protein immunoassay (IA), SBCR
is evaluated and compared with a commercial MTP reader (MTPR). For analysis of CRP spiked in diluted
human whole blood and plasma as well as CRP in clinical plasma samples, SBCR exhibits the same precision,
dynamic range, detection limit, and sensitivity as MTPR for the developed IA (DIA). Considering its
compactness, low cost, advanced features and a remarkable computing power, SBCR is an ideal point-of-
care (POC) colorimetric detection device for the next-generation of cost-effective POC testing (POCT).
Key words Smartphone-based colorimetric reader, Human C-reactive protein, Graphene, One-step
kinetics, Sandwich immunoassay, Monitoring inflammation
1 Introduction
*
These authors are contributed equally to this work.
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_21,
© Springer Science+Business Media LLC 2017
343
344 A.G. Venkatesh et al.
Fig. 1 (a) Schematics of the smartphone-based colorimetric readers (SBCRs) developing using the gadgets’
(iPAD4, iPAD mini, or iPhone 5s) screen-based bottom illumination, Samsung Galaxy SIII mini’s back camera
(5 MP) based imaging, and a custom-made polyamide dark hood and polyamide base holder assembly. (b)
Screensavers used for the screen-based bottom illumination of the 96-well microtiter plate (MTP) in gadgets.
(c) Image processing algorithm employed for the analysis of smartphone-captured colorimetric images
Fig. 2 Typical SBCR assemblies, demonstrating the steps involved for SBCR-based colorimetric readout of
immunoassays, for (a) iPAD4, (b) iPAD mini, and (c) iPhone 5s. The process involves the placement of the
gadget, with an illuminated screensaver, within the engraved cavity of the base holder. The MTP with
colorimetric products is then placed on top of the screensaver after aligning with the respective illumination
spots. Subsequently, the dark hood is placed on top of the base holder and the smartphone imaging is
performed after placing the smartphone in the containment provided at the top of the dark hood and aligning
the smartphone’s back camera with the hole
2 Materials
2.1 Preparation 1. Nunc microwell 96-well polystyrene plates, flat bottom (non-
of Capture Ab-Bound treated), sterile (Catalog No. P7491, Sigma-Aldrich).
MTP 2. Eppendorf microtubes (1.5 mL; Catalog No. Z 606340,
Sigma-Aldrich).
3. Deionized water (18.2 MΩ, DIW). (Direct-Q®3 Water Purifi-
cation System, Millipore).
4. 70 C freezer (operating range 60 to 86 C) (New
Brunswick).
5. 2–8 C refrigerator (Future, UK).
6. Direct-Q®3 water purification system (Millipore, USA).
7. Mini incubator (Labnet Inc., UK).
8. PVC fume cupboard Chemflow range (CSC Ltd.).
9. KOH pellets (99.99%), semiconductor grade (Catalog No.
306568, Sigma-Aldrich).
A Smartphone-Based Colorimetric Reader for Human C-Reactive Protein Immunoassay 347
Fig. 4 The bioanalytical performance of the developed SBCR-based human CRP IA. (a) Detection of CRP in PBS
(10 mM, pH 7.4), diluted human plasma and diluted human whole blood. (b) Experimental process controls.
Anti-CRP1 and anti-CRP2 are capture and detection antibodies, respectively. The control proteins employed
are human serum albumin (HSA), human lipocalin-2 (LCN2), human fetuin A (HFA), interleukin (IL)-1 beta, IL-6,
IL-8, and tumor necrosis factor (TNF)-alpha. (c) Storage stability of anti-human CRP capture Ab-bound
graphene-functionalized MTPs stored at 4 C in PBS for 6 weeks. (d) Determination of batch-to-batch
variability for various preparations of GNP-anti-CRP Ab, as demonstrated for the detection of 9 ng/mL CRP
by the developed graphene-based IA. All experiments are performed in triplicate with the error bars
representing the standard deviation. Reproduced with permission from Elsevier Inc. [6]
Fig. 5 An overlay plot of the developed human CRP IAs based on the use of SBCR and MTPR, and employing
the normalized signals (the signal response obtained for a particular human CRP concentration divided by the
signal response obtained for the highest human CRP concentration). Reproduced with permission from
Elsevier Inc. [6]
16. Blocker BSA in PBS (10), pH 7.4, 10% w/v (Catalog No.
37525, Thermo Scientific).
17. 0.1% BSA in PBS, pH 7.4 is used as the binding buffer.
18. 0.05% Tween® 20 in PBS, pH 7.4 (PBST) is used as the
washing buffer.
2.2 Human CRP IA 1. Recombinant human CRP (Catalog No. 1707-CR, R&D
Systems).
2. Biotinylated mouse anti-human CRP detection Ab (Catalog
No. BAM17072, R&D Systems).
3. Streptavidin-conjugated horseradish peroxidase (SA-HRP,
Catalog No. , R&D Systems).
4. Biotinylated anti-CRP detection Ab conjugated to SA-HRP is
prepared by adding 1 μL of biotinylated anti-CRP detection Ab
(0.5 mg/mL) to 1 μL of SA-HRP to 2998 μL of the binding
buffer followed by 20 min of incubation at RT. The resulting
concentration of biotinylated anti-CRP detection Ab is
0.17 μg/mL, while SA-HRP dilution is 1:3000.
5. 3,30 ,5,50 -Tetramethylbenzidine (TMB) substrate kit (Catalog
No. 34021, Thermo Scientific).
(a) TMB solution (0.4 g/L).
(b) Hydrogen peroxide solution (containing 0.02% v/v
H2O2 in citrate buffer).
350 A.G. Venkatesh et al.
3 Methods
3.2 Human CRP IA 1. CRP spiked diluted human whole blood or serum: The CRP
spiked samples (0.3–81 ng/mL) are prepared by mixing the
desired CRP concentrations in 1:100 diluted human whole
blood/serum.
2. EDTA plasma samples from anonymized patients. The EDTA
plasma samples from anonymized patients are diluted 1:1000
and 1:4000 in the binding buffer, which makes the resulting
CRP concentration in these samples to fall within the DIA
linear range, thereby enabling the detection of an entire patho-
physiological concentration range of human CRP.
3. Biotinylated anti-CRP detection Ab (100 μL, 0.17 μg/mL)
pre-conjugated to SA-HRP and 100 μL of CRP (varying con-
centrations; 0.3–81 ng/mL) are dispensed sequentially to the
Ab-bound and BSA-blocked MTP wells, and incubated for
15 min at 37 C (see Note 11).
4. The sandwich immune complex-bound MTP is washed with
300 μL of PBST five times to remove nonspecifically bound
substances and excess IA reagents.
352 A.G. Venkatesh et al.
3.3 SBCR Analysis 1. The colorimetric readout of the MTP is performed by smart-
phone imaging using our developed SBCR set up (Figs. 1 and 2).
The gadget (iPAD 4, iPAD mini or iPhone 5s) is placed in the
designated groove of the base holder followed by placing the
96-well MTP on the gadget’s screensaver that provides white
light-based bottom illumination only in the specific regions
corresponding to the MTP well’s bottom. The dark box is then
placed on top of the base holder in the alignment groove.
Subsequently, the Samsung SIII mini’s back camera-based imag-
ing of the colorimetric reaction in the MTP is performed by
placing the smartphone inside the designated groove on top of
the dark hood (see Note 15).
2. The desired pixel intensity (PI) of the captured smartphone
image is then determined by our novel image analysis algo-
rithm using Image J 1.48v (http://imagej.nih.gov/ij)
(Fig. 1c). The color channel of the image is split and the pixel
coordinate of an individual MTP well’s center is identified
using the red channel image. A mean of neighboring pixels
(varies based on the resolution of the camera) from the center is
calculated individually for all the channels. The composite
mean PI (CMPI) is derived from the following color-weighted
formula [CMPI ¼ 0.7 MPIBlue + 0.2 MPIGreen + 0.1 MPIRed].
The desired PI is determined as 255 CMPITest Conc CMPI-
Blank. The resulting CMPI is plotted against their respective—
CRP concentrations using the four-parameter logistic based
standard curve analysis in SigmaPlot version 11.2 software.
The determination of CMPI and PI is done in Microsoft
Excel after taking the PI values of red, blue and green channels
of the smartphone-captured image using image J. The concen-
tration of unknown CRP samples is determined on the basis of
their CMPI from the resulting calibration plot. As described in
Subheading 3.2, a standard curve is plotted using various
known concentrations of CRP (0.3–81 ng/mL). A schematic
of the image analysis algorithm, comprising various steps, is
provided in Fig. 6.
A Smartphone-Based Colorimetric Reader for Human C-Reactive Protein Immunoassay 353
Fig. 6 Schematic of the image analysis procedure for the SBCR based colorimetric readout
4 Notes
Acknowledgments
References
1. Vashist SK, Mudanyali O, Schneider EM et al 12. Tappero E, Johnson P (2010) Laboratory eval-
(2014) Cellphone-based devices for bioanaly- uation of neonatal sepsis. Newborn Infant
tical sciences. Anal Bioanal Chem 406: Nurs Rev 10:209–217
3263–3277 13. http://www.nomidalliance.org/downloads/
2. http://www.itu.int/en/ITU-D/Statistics/ comparative_chart_front.pdf. Accessed 8 June
Documents/publications/mis2013/MIS2013_ 2015
without_Annex_4.pdf. Accessed 8 June 2015 14. Danner M, Kasl SV, Abramson JL et al (2003)
3. Vashist SK, Schneider EM, Luong JHT (2014) Association between depression and elevated
Commercial smartphone-based devices and C-reactive protein. Psychosom Med
smart applications for personalized healthcare 65:347–356
monitoring and management. Diagnostics 15. Raison CL, Capuron L, Miller AH (2006)
4:104–128 Cytokines sing the blues: inflammation and
4. Ozcan A (2014) Mobile phones democratize the pathogenesis of depression. Trends Immu-
and cultivate next-generation imaging, diag- nol 27:24–31
nostics and measurement tools. Lab Chip 16. Spitzer C, Barnow S, Volzke H et al (2010)
14:3187–3194 Association of posttraumatic stress disorder
5. Vashist SK, van Oordt T, Schneider EM et al with low-grade elevation of C-reactive protein:
(2015) A smartphone-based colorimetric evidence from the general population. J Psy-
reader for bioanalytical applications using the chiatr Res 44:15–21
screen-based bottom illumination provided by 17. Lin MS, Shih SR, Li HY et al (2007) Serum
gadgets. Biosens Bioelectron 67:248–255 C-reactive protein levels correlates better to
6. Vashist SK, Schneider EM, Zengerle R et al metabolic syndrome defined by Interna-
(2015) Graphene-based rapid and highly- tional Diabetes Federation than by NCEP
sensitive immunoassay for C-reactive protein ATP III in men. Diabetes Res Clin Pract
using a smartphone-based colorimetric reader. 77:286–292
Biosens Bioelectron 66:169–176 18. Morimoto H, Sakata K, Oishi M et al (2013)
7. Marnell L, Mold C, Du Clos TW (2005) C- Effect of high-sensitivity C-reactive protein on
reactive protein: ligands, receptors and role in the development of diabetes as demonstrated
inflammation. Clin Immunol 117:104–111 by pooled logistic-regression analysis of annual
8. Chiesa C, Natale F, Pascone R et al (2011) C health-screening information from male Japa-
reactive protein and procalcitonin: reference nese workers. Diabetes Metab 39:27–33
intervals for preterm and term newborns dur- 19. Testa R, Bonfigli AR, Sirolla C et al (2008) C-
ing the early neonatal period. Clin Chim Acta reactive protein is directly related to plasmino-
412:1053–1059 gen activator inhibitor type 1 (PAI-1) levels in
9. Chiesa C, Panero A, Osborn JF et al (2004) diabetic subjects with the 4G allele at position
Diagnosis of neonatal sepsis: a clinical and lab- 675 of the PAI-1 gene. Nutr Metab Cardio-
oratory challenge. Clin Chem 50:279–287 vasc Dis 18:220–226
10. Dollner H, Vatten L, Austgulen R (2001) Early 20. Kuo HK, Yen CJ, Chen JH et al (2007) Asso-
diagnostic markers for neonatal sepsis: compar- ciation of cardiorespiratory fitness and levels of
ing C-reactive protein, interleukin-6, soluble C-reactive protein: data from the National
tumour necrosis factor receptors and soluble Health and Nutrition Examination Survey
adhesion molecules. J Clin Epidemiol 1999–2002. Int J Cardiol 114:28–33
54:1251–1257 21. Park HE, Cho GY, Chun EJ et al (2012) Can
11. Nguyen-Vermillion A, Juul SE, McPherson RJ C-reactive protein predict cardiovascular events
et al (2011) Time course of C-reactive protein in asymptomatic patients? Analysis based on
and inflammatory mediators after neonatal sur- plaque characterization. Atherosclerosis
gery. J Pediatr 159:121–126 224:201–207
356 A.G. Venkatesh et al.
22. Ridker PM (2003) High-sensitivity C-reactive with a schizophrenia spectrum disorder treated
protein and cardiovascular risk: rationale for in routine medical practice. Eur Psychiatry
screening and primary prevention. Am J Car- 28:161–167
diol 92:17K–22K 24. Wilson AM, Ryan MC, Boyle AJ (2006) The
23. Sicras-Mainar A, Rejas-Gutierrez J, Navarro- novel role of C-reactive protein in cardiovascu-
Artieda R et al (2013) C-reactive protein as a lar disease: risk marker or pathogen. Int J Car-
marker of cardiovascular disease in patients diol 106:291–297
Chapter 22
Abstract
Hepatitis B virus (HBV) can cause viral infection that attacks the liver and it is a major global health problem
that puts people at a high risk of death from cirrhosis of the liver and liver cancer. HBV has infected one-
third of the worldwide population, and 350 million people suffer from chronic HBV infection. For these
reasons, development of an accurate, sensitive, and expedient detection method for diagnosing, monitor-
ing, and assessing therapeutic response of HBV is very necessary and urgent for public health and disease
control. Here we report a new strategy for detection of viral load quantitation of HBV based on colorimet-
ric polymerase chain reaction (PCR) with DNAzyme-containing probe. The special DNAzyme adopting a
G-quadruplex structure exhibited peroxidase-like activity in the presence of hemin to report colorimetric
signal. This method has shown a broad range of linearity and high sensitivity. This study builds an important
foundation to achieve the specific and accurate detection level of HBV DNA with a low-cost and effective
method in helping diagnosing, preventing and protecting human health form HBV all over the world, and
especially in developing countries.
Key words Hepatitis B virus, Molecular diagnosis, Biosensor, G-quadruplex, DNAzyme, Peroxidase-
like activity, Colorimetric PCR
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_22,
© Springer Science+Business Media LLC 2017
357
358 Li Yang et al.
2 Materials
2.3 Amplification 1. HBV DNA template (the DNA is extracted from the HBV
infection clinical serum samples) is dissolved in sterile water,
stored at 20 C.
2. Primers (forward primer: 1.5 μM, reverse primer: 1 μM; San-
gon Biotech, Shanghai, China) are dissolved in sterile water,
stored at 20 C.
3. Probe (32P labeled, 0.6 μM; Sangon Biotech, Shanghai, China)
is dissolved in sterile water, stored at 20 C.
4. 1.2 mM dNTPs (600 μM dUTP, 200 μM dATP, 200 μM
dCTP, 200 μM dGTP; TransGen Biotech, Beijing, China).
5. FastStart Taq DNA polymerase (5 U; Roche).
6. Taq DNA polymerase (5 U; TransGen Biotech, Beijing,
China).
7. UNG (1 U; Takara).
8. MgCl2 (0.5 mM).
9. 1 Taq polymerase buffer (15 mM Tris–HCl (pH 8.2), 30 mM
KCl, 5 mM (NH4)2SO4, 2.5 mM MgCl2, 0.002% BSA; Trans-
Gen Biotech, Beijing, China).
10. C1000 thermal cycler (Bio-Rad).
3 Methods
3.1 The Principle of 1. The principle of our detection strategy is depicted in Fig. 1.
the Colorimetric The DNA probe containing three parts of sequences (A, B, C)
Detection is designed to form a hairpin structure at room temperature.
After denaturation step of PCR, the concentration of probes is
much higher than that of HBV dsDNA templates, so the
chance for probe to hybridize with the targeted HBV single
strand DNA is much higher than that of its complementary
single strand DNA. Then, in the annealing step of PCR, the
loop domain (part C) of the probe hybridizes to the conserved
region of HBV genome, which will be amplified by two pri-
mers. The stem part of the probe contains blocking sequence A
(black) and a DNAzyme sequence B (blue) that could report
the detection result through oxidation reaction [16–20]. The
blocking sequence A is used to prohibit DNAzyme sequence B
fold into catalytic G-quadruplex structure at room temperature
to decrease the background of colorimetric assay [7]. As the
Fig. 1 Strategy of the colorimetric PCR-based detection of HBV. A part of the probe is the blocking sequence, B
represents the sequence for DNAzyme forming G-quadruplex, loop C is the complementary sequence of the
conserved region of HBV genome. (1) Melting of double-stranded DNA and hairpin loop during PCR denatur-
ation. (2) Hybridization of probe and primers to targeted ssDNA respectively during PCR annealing step. (3)
Cleaving of the hybridized probe by DNA polymerase during primers extension. (4) Release of G-quadruplex
sequence after cleavage by DNA polymerase. (5) Horseradish peroxidase-mimicking DNAzymes formation
with the addition of hemin, reacting with ABTS and H2O2. (6) Colorimetric test result
A Novel Colorimetric PCR-Based Biosensor for Detection and Quantification. . . 361
3.2 DNA Extraction 1. Extraction of DNA from the HBV infection and normal clinical
from Clinical Serum serum samples from HBV patients or normal population is
Sample carried out using QIAamp DNA Blood Mini Kit as described
by the manufacturer (Qiagen).
3.3 The Serum 1. 108 copies/mL clinical serum samples are chosen to dilute with
Calibration Curve negative serum by decuple to ten copies successively. These
Preparation samples are extracted with QIAamp DNA Blood Mini Kit
(Qiagen) to build the calibration curve.
3.4 Manual Design of 1. According to our previous procedure, this method requires
Probe and Primers two consecutive PCR steps to achieve the successful detection
of a target DNA, which include the amplification of target
sequence and subsequently the cleavage of probe [7]. In our
experiment, three different hairpin probes (probe-HBV-1,
probe-HBV-2, and probe-HBV-3) and corresponding primers
are designed against the different conserved regions of the
HBV genome including overlapping genes encoding
X-protein, S gene, and X gene region.
2. Under the optimized conditions, Fig. 2 shows that probe-
HBV-2 for S gene region of HBV genome exhibits the best
colorimetric result comparative to that of other probes. Fur-
thermore, this region of HBV genome is rather conserved in
different genotypes of HBV strains and can be extensively and
accurately used to detect all HBV genotypes. Therefore, the
probe-HBV-2 has been proved to be the best fitted probe for
HBV detection based on our method. The selected sequences
are:
Primer-HBV-Forward-2:CCTGGTTATCGCTGGATGTGT,
Primer-HBV-Reverse-2:GGACAAACGGGCAACATACCTT,
Probe-HBV-2: CCCTACCCA TTCATCCTGCTGCTATG
CCTCATCTTCTT TGGGTAGGG CGGGTTGGGAAA -
NH2 (see Note 1).
The sequence marked with box represents the stem part of
Probe-HBV-2. Sequence in the second box would be closed
362 Li Yang et al.
Fig. 2 Colorimetric PCR reaction with three different probes. Tube 1: probe-HBV-
1 with HBV DNA targets; tube 2: probe-HBV-1 without targets; tube 3: probe-
HBV-2 with targets; tube 4: probe-HBV-2 without targets; tube 5: probe-HBV-3
with targets; tube 6: probe-HBV-3 without targets
3.5 One-Step PCR 1. Thaw 10 Taq polymerase buffer, dNTPs, and primers.
2. The PCR is performed in 50 μL volume containing 2 μL HBV
DNA template, 1.5 μM Primer-HBV-Forward-2, 1 μM
Primer-HBV-Reverse-2, 0.6 μM Probe-HBV-2, 1.2 mM
dNTPs (600 μM dUTP, 200 μM dATP, 200 μM dCTP,
200 μM dGTP), 5 U of FastStart Taq DNA polymerase, 1 U
of UNG (see Note 2), 0.5 mM MgCl2, 1 Taq polymerase
buffer (15 mM Tris–HCl (pH 8.2), 30 mM KCl, 5 mM
(NH4)2SO4, 2.5 mM MgCl2, 0.002% BSA).
3. The negative control is prepared without HBV DNA.
4. The amplification conditions are as follows: 20 C for 10 min;
95 C for 2 min followed by 40 cycles of 94 C for 30 s and
60 C for 90 s.
3.6 32P Labeled 1. To verify the probe had been cleaved by DNA polymerase
Selected Probe PCR successfully, Probe-HBV-2 and mark are 50 end-labeled with
Reaction [γ-32P] ATP. The reaction mixture containing oligonucleotides
with 10 μCi [γ-32P] and 10 U of T4 polynucleotide kinase is
incubated for 1 h at 37 C for DNA phosphorylation.
2. The labeled product is purified by 10% denaturing PAGE.
3. The 50 32P labeled probe-HBV-2 is added into the PCR reaction.
4. The PCR product is used for PAGE analysis. As shown in
Fig. 3, in the positive control containing HBV genome, a
A Novel Colorimetric PCR-Based Biosensor for Detection and Quantification. . . 363
Fig. 4 Colorimetric detection of HBV DNA. (a) Photograph of the colorimetric detection of different concentra-
tion of HBV DNA: tube 1, negative control (without HBV DNA); tubes 2–8 containing 10, 102, 103, 104, 105, 106,
107 copies of HBV DNA, successively. (b) Time-dependent colorimetric detection of different concentrations of
HBV DNA. The inset is a calibrated curve of the average absorbance (414 nm) at 3 min plotted against the
number of HBV DNA. The solid line indicates linear least squares fitting between 10 and 107 copies of HBV
DNA, and their formulation is FI ¼ 0.0061 lg(CHBVDNA) + 0.0678 (R2 ¼ 0.9788). The correlation coefficient is
0.9788. The error bars were determined by standard deviation (SD) of the triplicate data
A Novel Colorimetric PCR-Based Biosensor for Detection and Quantification. . . 365
3.8 Comparation 1. TaqMan assay is the most advanced technique for HBV detec-
with TaqMan Assay tion, which is widely used in clinical diagnosis, so we compare
our colorimetric assay with the quantitative TaqMan PCR
assay. Seven different concentrations of clinical HBV serum
samples and seven negative clinical serum samples are measured
with the TaqMan method.
2. Prepared reagents according to the instruction of Hepatitis B
Viral DNA Quantitative Fluorescence Diagnostic Kit.
3. 200 μL of negative control, positive control, quantitative refer-
ence A–D, and the sample under test is respectively added to
seven clean 1.5 ml microcentrifuge tubes, each containing
300 μL of DNA extraction solution. Mix completely by vibrat-
ing for about 10 min.
4. Add 100 μL of DNA extraction solution 2-mix to each tube.
Mix for 10 s, then quiescence at room temperature for 10 min.
5. Place the microcentrifuge tubes into the magnetic bead separa-
tor for 3 min after a short centrifugation, then sucked out the
solution slowly.
6. 600 μL DNA extraction solution 3 and 200 μL DNA extraction
solution 4 are added to each tube. Mix for 5 s.
7. Place the microcentrifuge tubes into the magnetic bead separa-
tor again after a short centrifugation. 3 min later, there are two
layers of the solution. Discard the lower layer and transfer all of
the microcentrifuge tubes to tube rack.
366 Li Yang et al.
a
1.2
NC
1
101 Copies
103 Copies
0
1 2 3 4 5 6 7 8 9 10 11 12 13
T (min)
b
0.8
y = 0.0731x + 0.0319
0.7 R2 = 0.9822
0.6
Absorbance
0.5
0.4
0.3
0.2
0.1
0
101 102 103 104 105 106 107 108
Concentration (copy)
10
Colormetric method
9
Taqman method
8
7
Concentration (1g)
6
9
TaqMan assay (lg of concentration)
y = 0.9091x + 0.8262
5 8 2
R = 0.913
7
4 6
5
4
3
3
2
2 1
0
1 0 1 2 3 4 5 6 7 8 9
Colormetric assay (lg of concentration)
0
1 2 3 4 5 6 7
Sample number
Fig. 6 Comparison of colorimetric and TaqMan assay. Serum samples of consecutive concentration were
measured by colorimetric and TaqMan assay. Red points represent the values of TaqMan assay and blue
points represent the values of colorimetric assay. The inset is the comparison of the values by colorimetric and
TaqMan assay. The concentration of target used for experiment was copies/ml. “Ig” represents the denary
logarithm value of the concentration of target. X-axis represents the lg value of concentration by colorimetric
assay and Y-axis represents the lg value of concentration by TaqMan assay. The correlation coefficient is
0.913
368 Li Yang et al.
4 Notes
Acknowledgments
This study was supported by the financial supports from the CAS
(Hundreds of Talents Program), National Science Foundation of
China (Grant No. 21172215 and No. 21102140), Innovation
Program of the CAS (Grant No. KSCX2-EW-J-22).
References
1. Mahoney FJ (1999) Update on diagnosis, Infect Dis Med Microbiol J Can Mal Infect
management, and prevention of hepatitis B Microbiol Med/AMMI Can. 16:65–72
virus infection. Clin Microbiol Rev 12: 5. Mullis KB, Faloona FA (1987) Specific synthe-
$351–366 sis of DNA in vitro via a polymerase-catalyzed
2. Loeb KR, Jerome KR, Goddard J et al (2000) chain reaction. Methods Enzymol
High-throughput quantitative analysis of hep- 155:335–350
atitis B virus DNA in serum using the TaqMan 6. Lee LG, Connell CR, Bloch W (1993) Allelic
fluorogenic detection system. Hepatology discrimination by nick-translation PCR with
32:626–629 fluorogenic probes. Nucleic Acids Res
3. Gish RG, Locarnini SA (2006) Chronic hepa- 21:3761–3766
titis B: current testing strategies, clinical gas- 7. Du F, Tang Z (2011) Colorimetric detec-
troenterology and hepatology. Off Clin Pract J tion of PCR product with DNAzymes
Am Gastroenterol Assoc 4:666–676 induced by 50 -nuclease activity of DNA
4. Krajden M, McNabb G, Petric M (2005) The polymerases. Chembiochem Eur J Chem
laboratory diagnosis of hepatitis B virus. Can J Biol 12:43–46
A Novel Colorimetric PCR-Based Biosensor for Detection and Quantification. . . 369
8. Holland PM, Abramson RD, Watson R et al 14. Majhi PR, Shafer RH (2006) Characterization
(1991) Detection of specific polymerase chain of an unusual folding pattern in a catalytically
reaction product by utilizing the 50 –30 exonucle- active guanine quadruplex structure. Biopoly-
ase activity of Thermus aquaticus DNA polymer- mers 82:558–569
ase. Proc Natl Acad Sci U S A 88:7276–7280 15. Kong DM, Wu J, Wang N et al (2009) Peroxi-
9. Xiao Y, Pavlov V, Niazov T et al (2004) Catalytic dase activity-structure relationship of the inter-
beacons for the detection of DNA and telome- molecular four-stranded G-quadruplex-hemin
rase activity. J Am Chem Soc 126:7430–7431 complexes and their application in Hg2+ ion
10. Li Y, Sen D (1996) A catalytic DNA for por- detection. Talanta 80:459–465
phyrin metallation. Nat Struct Biol 3:743–747 16. Emilsson GM, Breaker RR (2002) Deoxyribo-
11. Travascio P, Li Y, Sen D (1998) DNA- zymes: new activities and new applications. Cell
enhanced peroxidase activity of a DNA-apta- Mol Life Sci 59:596–607
mer-hemin complex. Chem Biol 5:505–517 17. Baum DA, Silverman SK (2008) Deoxyribo-
12. Travascio P, Witting PK, Mauk AG et al (2001) zymes: useful DNA catalysts in vitro and
The peroxidase activity of a hemin–DNA oligo- in vivo. Cell Mol Life Sci 65:2156–2174
nucleotide complex: free radical damage to spe- 18. Navani NK, Li Y (2006) Nucleic acid aptamers
cific guanine bases of the DNA. J Am Chem and enzymes as sensors. Curr Opin Chem Biol
Soc 123:1337–1348 10:272–281
13. Deng M, Zhang D, Zhou Y et al (2008) Highly 19. Breaker RR (1997) DNA enzymes. Nat Bio-
effective colorimetric and visual detection of technol 15:427–431
nucleic acids using an asymmetrically split per- 20. Liu J, Cao Z, Lu Y (2009) Functional nucleic
oxidase DNAzyme. J Am Chem Soc acid sensors. Chem Rev 109:1948–1998
130:13095–13102
Chapter 23
Abstract
Rapid and precise quantification of the infectivity of HIV is important for molecular virologic studies,
as well as for measuring the activities of antiviral drugs and neutralizing antibodies. An indicator cell line,
a CCD camera, and image-analysis software are used to quantify HIV infectivity. The cells of the P4R5 line,
which express the receptors for HIV infection as well as β-galactosidase under the control of the HIV-1 long
terminal repeat, are infected with HIV and then incubated 2 days later with X-gal to stain the infected cells
blue. Digital images of monolayers of the infected cells are captured using a high resolution CCD video
camera and a macro video zoom lens. A software program is developed to process the images and to count
the blue-stained foci of infection. The described method allows for the rapid quantification of the infected
cells over a wide range of viral inocula with reproducibility, accuracy and at relatively low cost.
Key words Charge-coupled device (CCD), Imaging, HIV, Infectivity, Quantify, Rapid
1 Introduction
Portions reprinted from the Journal of Virological Methods, Vol. 137, J.R. Day, L.E. Martı́nez, R. Šášik,
D.L. Hitchin, M.E. Dueck, D.D. Richman and J.C. Guatelli, A computer-based, image-analysis method to
quantify HIV-1 infection in a single-cycle infectious center assay, pp. 125–133, Copyright 2006, with permission
from Elsevier.
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_23,
© Springer Science+Business Media LLC 2017
371
372 John R. Day
2 Materials
2.1 Imaging 1. Five megapixel CCD color camera with real-time viewing,
Apparatus C-mount optical lens interface, and FireWire IEEE 1394 digi-
tal interface: MicroPublisher 5.0 RTV (Model # MP5.0-RTV-
CLR-10, QImaging, Burnaby, BC, Canada) (see Note 1).
2. Macro video zoom lens: Optem 18–108 mm, f/2.5, C-mount
(Qioptiq Imaging Solutions, Rochester, NY, formerly Thales
Optem, Inc.) (see Note 2).
3. Copy stand with a 1/400 -20 threaded mounting screw (model #
CS-3, Testrite Instrument Company, Inc.). Something similar
would suffice.
4. Fluorescent light box (typically used for viewing X-ray films) (see
Note 3).
5. Camera mount adapter, custom made by our university
machine shop, consisting of a tripod mounting screw (1/400 -
20 thread) and a rectangular piece of aluminum
(2.25 1 0.25 in.) with two drilled holes (see Note 4).
CCD Camera Detection of HIV Infection 373
Use of the adapter requires that the copy stand have a mount-
ing rod that can extend away from the stand.
6. Personal computer for capturing and analyzing images: Win-
dows operating system, 1 available FireWire port (IEEE 1394)
or a FireWire expansion card.
2.3 HIV Infectivity 1. Indicator cells: P4R5 HeLa cells (P4.R5 MAGI cells, AIDS
Assay Research and Reference Reagent Program, Division of AIDS,
NIAID, NIH contributed by Dr. Nathaniel Landau). The
P4R5 cell line contains the β-galactosidase gene under the
control of the HIV-1 promoter region (LTR). The derivation
of the parental P4 line has been reported [7]. P4R5 cells
express on their surface the HIV entry receptors, CD4,
CXCR4, and CCR5.
2. Complete media: Dulbecco’s Modified Eagle Medium
(DMEM; Gibco, Grand Island, NY) supplemented with 10%
fetal bovine serum (FBS; GemCell, Woodland, CA),
100 U/mL penicillin, 100 μg/mL streptomycin (pen/strep;
Gibco, Grand Island, NY), 2 mM L-glutamine (Gibco, Grand
Island, NY) and 1 μg/mL puromycin. Store at 4 C.
3. 48-well tissue culture plate (Cat. # 353078, BD Falcon, San
Jose, CA).
4. Phosphate buffered saline, 1 (PBS; Invitrogen, Carlsbad, CA).
5. Fix solution: 1% formaldehyde, 0.2% glutaraldehyde in PBS.
Can be made in advance and stored in the dark at 4 C for
1–2 months.
6. X-gal stain solution made fresh: For each 1.0 mL of solution,
combine 949 μL PBS, 20 μL 0.2 M potassium ferrocyanide,
20 μL 0.2 M potassium ferricyanide, 1.0 μL 2.0 M Mg2CL, and
10 μL 40 mg/mL X-gal (5-bromo-4-chloro-3-indolyl-β-D-
galactopyranoside). Stock solutions of potassium ferrocyanide,
potassium ferricyanide, Mg2Cl, and X-gal can be prepared and
374 John R. Day
3 Methods
3.1 Setup of Imaging 1. Place the copy stand on a table and the light box (or light
Apparatus source of choice) on the base of the stand.
2. Assemble the camera by screwing the lens onto the camera.
3. Mount the camera to the stand using the 1/400 -20 threaded
mount. The most straightforward way to mount the camera is
illustrated in Fig. 1a. An alternative method to mount the
camera requires a custom-built adapter (see Note 6). The alter-
native method is illustrated in Fig. 1b and a photograph of
the setup is shown in Fig. 2. Screw the mount adapter onto the
copy stand, then attach the camera to the adapter using the
tripod mount screw (Fig. 3).
4. The height of the camera above the light box is adjustable and
depends on the zoom setting of the lens. In our setup, the
distance between the surface of the light box and the mounting
screw hole of the camera is 14.25 in. (36.2 cm). The tip of the
lens is 6.25 in. (15.9 cm) above the light box. Level the light
box and camera using a standard bubble level.
CCD Camera Detection of HIV Infection 375
Fig. 1 Schematic diagrams of the CCD camera setup. (a) The simplest setup using direct attachment of the
camera to the copy stand. The FireWire ports on the camera are on the backside in the perspective shown. (b)
Use of an aluminum mount adapter and tripod screw to mount the camera rotated at 180 (see Notes 4 and 6)
Fig. 2. Photograph of the actual camera setup showing the light box, copy stand,
CCD camera, macro lens, and computer
376 John R. Day
Fig. 3. (a) Photograph of the camera mounted to the copy stand. (b) Close-up of the mounted camera with
mount adapter
3.2 Capture and 1. These instructions assume the use of the Micropublisher 5.0
Analysis of Images RTV camera, Optem 18–108 mm macro video zoom lens, and
QCapture Pro software. Other cameras, capture software, and
3.2.1 Capturing Images
analysis software may be used. Please follow the directions
included with your specific components.
2. Follow the setup instructions from QImaging to load the
camera drivers and QCapture Pro software. You will connect
the FireWire cable to the computer during this process.
3. To define the capture settings, launch QCapture Pro and click
the camera icon on the toolbar (or select Acquire/Video-Digital
from the menu) to open the acquisition dialog box. Use the
Basic Dialog (rather than the Advanced Dialog) by clicking the
“Basic Dialog” button. If the button in the lower left says
“Advanced” then you are already looking at the Basic Dialog.
4. Check the settings by clicking the “More >>” button at the
bottom of the window. This expands the window. Using ver-
sion 5.0.0.16 of QCapture Pro and with the specific light
source we used, the settings are as follows (Fig. 4) (see Note
7 for possible variations):
l Exp Acq: 00.300.00, Adjust Exp for Binning (checked).
l Binning: Pvw (2 2 for focusing, 4 4 for moving the
plate); Acq (1 1).
l Capture area dimensions: Left (320), Top (0), Right (2239),
Bottom (1919) [final resolution, 1920 1920 pixels].
CCD Camera Detection of HIV Infection 377
Fig. 4. Screen shot of the capture settings within QCapture Pro capture software
initial height adjustment of the camera above the light box may
be necessary as well, to achieve the desired result. Use 2 2
Preview (Pvw) binning to focus, then switch to 4 4 for
moving the plate. As long as Acq binning remains at 1 1
the full resolution image will be acquired. The binning status of
Pvw does not affect the acquired image, but only the image
seen in the preview window. Binning at 4 4 allows for faster
on-screen response of plate movement (see Note 9).
9. Capture images by clicking the “Snap” button. Move the 48-
well plate to view the next well, click “Snap,” and repeat for the
entire plate.
10. After acquiring all the images, close the live preview window by
clicking the “Stop” button, then the “X” in the upper right
corner of the acquisition window.
11. Save the images to a new folder. A quick way to save all the
images is to select the menu Window/Close All, then click “Yes
All.” When prompted, type a name for each file. Save the files as
TIFF for analysis by the Romanizer software.
3.2.2 Analyzing Images 1. The Romanizer program will automatically count blue-stained
HIV-infected cells in 1920 1920 pixel, 24-bit color images.
Note that in addition to counting blue-stained cells, the soft-
ware may also count overly clumped cells, dust, or fibers that
appear dark in the image (see Note 10).
2. Open the file folder where the Romanizer program is stored.
Copy 5MGL.exe and 5MGL.PAR into the folder where you
saved your images. The program must reside in the same folder
as your TIFF images, and it will analyze all images within that
folder.
3. Double-click 5MGL.exe to launch the counting program. You
should see one of your images with a red circle superimposed
upon it (Fig. 5). The circle defines the region to be analyzed.
Using the computer mouse, click and drag the circle to the
center of your well to check its size. The circle should be a little
smaller than the size of the well to prevent analyzing the edge
of the well. If you need to adjust the size follow these steps:
(a) Close the program by clicking Exit on the menu.
(b) Open the file 5MGL.PAR file (see Note 11) using a simple
text editor such as the Notepad.
(c) Increase or decrease the number next to “circle radius in
tiff pixels” depending on the desired change in the circle
radius.
(d) Save your changes and close the file.
CCD Camera Detection of HIV Infection 379
Fig. 5. Screen shot of the Romanizer program. The red circle indicates the area
that will be analyzed by the program. The position of the analysis circle was
defined for each image because of potential variations in the placement of the
well within the digital image
Fig. 6. Image-analysis of HIV-infected P4R5 cells. (a) Digital photograph of a monolayer of P4R5 cells infected
with HIV and stained with X-gal within one well of a 48-well tissue culture plate using a MicroPublisher 5.0
camera and macro video zoom lens. (b) The same well after processing by the analysis software. The boxed
region is magnified in Fig. 7
3.3 HIV Infectivity 1. These instructions describe an assay to quantify the infectivity
Assay of live HIV virus. Sterile cell culture technique and safe labora-
tory practices for handling live HIV are beyond the scope of
this description. We performed these assays in a Biosafety Level
3 (BL3) facility approved by the Environmental Health and
Safety department of our institution.
2. The P4R5 cell line is an adherent cell line derived from HeLa
cells. The cells are engineered to express β-galactosidase after
infection with HIV. Maintain them according to standard cell
culture procedures for adherent cell lines.
3. Plate cells at a density of 2 104 cells per well in a 48-well
tissue culture plate and place them in a humidified, 37 C, 5%
CO2 incubator overnight.
4. The next day, carefully aspirate the media, avoiding contact
with the cell monolayer. Infect the cells in duplicate wells
with 100 μL of serial dilutions of virus in complete media.
After a 2 h incubation at 37 C, add 400 μL of complete
media. Incubate for 2 days at 37 C.
5. Carefully aspirate the media from the wells and add 1 mL of fix
solution to each well. Incubate for 5 min at room temperature.
The fix solution renders the virus non-infectious.
6. Aspirate the fix solution and wash the wells twice with PBS.
CCD Camera Detection of HIV Infection 381
Fig. 7 Magnified view of HIV-infected P4R5 cells. Magnified images of the boxed region of Fig. 6 depicting (a)
the original digital photograph, (b) the same region viewed through the 20 objective of a microscope, (c) the
processed image including small red dots as indicators of the objects counted and (d) outlines of infected cells
surrounding red dot markers. The arrows indicate cell groups counted singly using Simple count and as two
cells using Smart count
382 John R. Day
small red dot to allow the user to examine more closely how the
software is performing and exactly what objects in the image
are counted (Fig. 7c). To illustrate this more clearly, the out-
lines of infected foci are shown encircling each red dot
(Fig. 7d). Two or more red dots within a group of cells that
come in contact in the image are counted as one infection,
using “Simple” count and as multiple foci using “Smart” count
(see Note 12).
4 Notes
Fig. 8 (a) Photograph of the mount adapter with tripod mounting screw. (b) Photograph of mount adapter
holes. One hole was drilled wide enough for the tripod mounting screw to pass through easily. The second hole
was drilled with a 1/4”-20 thread for mounting onto the copy stand
384 John R. Day
Acknowledgments
References
1. Harada S, Koyanagi Y, Yamamoto N (1985) of primary strains of human and simian immu-
Infection of HTLV-III/LAV in HTLV-I-car- nodeficiency viruses. Virology 233:193–198
rying cells MT-2 and MT-4 and application in a 11. Wei X, Decker JM, Wang S, Hui H, Kappes JC,
plaque assay. Science 229:563–566 Wu X, Salazar-Gonzalez JF, Salazar MG, Kilby
2. McKeating JA, McKnight A, McIntosh K, JM, Saag MS, Komarova NL, Nowak MA,
Clapham PR, Mulder C, Weiss RA (1989) Hahn BH, Kwong PD, Shaw GM (2003) Anti-
Evaluation of human and simian immunodefi- body neutralization and escape by HIV-1.
ciency virus plaque and neutralization assays. J Nature 422:307–312
Gen Virol 70:3327–3333 12. Montefiori DC (2009) Measuring HIV neu-
3. Nara PL, Fischinger PJ (1988) Quantitative tralization in a luciferase reporter gene assay.
infectivity assay for HIV-1 and -2. Nature Methods Mol Biol 485:395–405
332:469–470 13. Tilton CA, Tabler CO, Lucera MB, Marek SL,
4. Chesebro B, Wehrly K (1988) Development of Haqqani AA, Tilton JC (2014) A combination
a sensitive quantitative focal assay for human HIV reporter virus system for measuring post-
immunodeficiency virus infectivity. J Virol entry event efficiency and viral outcome in pri-
62:3779–3788 mary CD4+ T cell subsets. J Virol Methods
5. Akrigg A, Wilkinson GW, Angliss S, Greenaway 195:164–169
PJ (1991) HIV-1 indicator cell lines. AIDS 14. Xu R, El-Hage N, Dever SM (2015)
5:153–158 Fluorescently-labeled RNA packaging into
6. Chackerian B, Long EM, Luciw PA, Over- HIV-1 particles: direct examination of infectiv-
baugh J (1997) Human immunodeficiency ity across central nervous system cell types.
virus type 1 coreceptors participate in posten- J Virol Methods 224:20–29
try stages in the virus replication cycle and 15. Day JR, Martı́nez LE, Šášik R, Hitchin DL,
function in simian immunodeficiency virus Dueck ME, Richman DD, Guatelli JC (2006)
infection. J Virol 71:3932–3939 A computer-based, image-analysis method to
7. Charneau P, Mirambeau G, Roux P, Paulous S, quantify HIV-1 infection in a single-cycle infec-
Buc H, Clavel F (1994) HIV-1 reverse tran- tious center assay. J Virol Methods 137:125–133
scription. A termination step at the center of 16. Beyon JDE, Lamb DR (eds) (1980) Charge-
the genome. J Mol Biol 241:651–662 coupled devices and their applications.
8. Deng H, Liu R, Ellmeier W, Choe S, Unutmaz McGraw-Hill, UK
D, Burkhart M, Di Marzio P, Marmon S, Sut- 17. Holst GC (1998) CCD arrays, cameras, and
ton RE, Hill CM, Davis CB, Peiper SC, Schall displays, 2nd edn. JCD Publishing/SPIE Opti-
TJ, Littman DR, Landau NR (1996) Identifi- cal Engineering Press, Winter Park, FL/Bel-
cation of a major co-receptor for primary iso- lingham, WA
lates of HIV-1. Nature 381:661–666 18. Howes MJ, Morgan DV (eds) (1979) Charge-
9. Kimpton J, Emerman M (1992) Detection of couple devices and systems. John Wiley &
replication-competent and pseudo-typed Sons, New York, NY
human immunodeficiency virus with a sensitive 19. NIH. (2005) NIH Image/ImageJ, a public
cell line on the basis of activation of an domain program developed at the US.
integrated beta-galactosidase gene. J Virol National Institutes of Health. Available at
66:2232–2239 http://rsb.info.nih.gov/nih-image/
10. Vodicka MA, Goh WC, Wu LI, Rogel ME, 20. Soille P (2003) Morphological image analysis:
Bartz SR, Schweickart VL, Raport CJ, Emer- principles and applications. Springer-Verlag,
man M (1997) Indicator cell lines for detection New York, NY
Chapter 24
Abstract
Real-time, on-site detection and quantification of different trace analytes is a challenge that requires both
searching a general class of molecules to recognize a broad range of contaminants and translating this
recognition to easily detectable signals. Functional nucleic acids, which include DNAzymes (DNA with
catalytic activity) and aptamers (nucleic acids that bind an analyte), are ideal candidates for the target
recognition. These nucleic acids can be selected by a combinatorial biology method called in vitro selection
to interact with a particular analyte with high specificity and sensitivity. Furthermore, they can be
incorporated into sensors by attaching signaling molecules. Due to the high extinction coefficients and
distance-dependent optical properties, metallic nanoparticles such as the commonly used gold nanoparti-
cles have been shown to be very attractive in converting analyte-specific functional DNA into colorimetric
sensors. DNAzyme directed assembly of gold nanoparticles has been used to make colorimetric sensors for
metal ions such as lead, uranium, and copper. To make the operation even easier and less vulnerable to
operator’s errors, dipstick tests have been constructed. Here, we describe protocols for the preparation of
DNAzyme-linked gold nanoparticles (AuNP) that are then immobilized on to lateral flow devices to make
easy-to-use dipstick tests for metal ions.
Key words Dipstick, Nanoparticle, Sensor, Colorimetric, Lateral flow, DNAzyme, Metal ions
1 Introduction
1.1 DNAzymes or Nucleic acids have recently emerged as an important platform for
Deoxyribozymes as selective molecular recognition, one major requirement for sensors.
Sensors for Metal Ions Long considered as passive molecules for the storage of genetic
information, RNA and DNA molecules with catalytic function
similar to protein enzymes were discovered in the early 1980s and
1990s, respectively [1–3]. These enzymes are called ribozymes
(catalytic RNA) and deoxyribozymes or DNAzymes (catalytic
DNA). The nucleic acid enzymes usually require a metal ion co-
factor to perform their catalytic function and can be tailored to be
specific for a particular metal ion. In addition, nucleic acids that
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_24,
© Springer Science+Business Media LLC 2017
389
390 Debapriya Mazumdar et al.
the nucleic acid substrate. All these DNAzymes (except the Cu2+
dependent DNAzyme) are RNA cleaving enzymes that catalyze the
cleavage of a single phosphodiester bond within the rA (RNA)
embedded in the DNA substrate. They share a similar secondary
structure containing a substrate strand and an enzyme strand hybri-
dizing to each other through the two arms on each side of the
cleavage site (Fig. 1c). It has been determined that, unlike the DNA
sequences in the single stranded loop, the sequence identity of two
arms do not contribute significantly to either metal binding or
activity, as long as they can form double stranded DNA to provide
stability [12]. Therefore the sequence length and GC content can
be designed so that, in the absence of the target metal ion, the
enzyme strand and substrate strand have a melting temperature
above the ambient temperature, making them hybridize to each
other. In the presence of target metal ion, however, the cleavage of
the substrate strand changed the melting temperature between the
two halves of the cleaved products and enzyme strand to below the
ambient temperature so that they can be released from the enzyme
strand. Some of the fastest DNAzymes have a catalytic efficiency
(kcat/Km) of 109 M1 min1 [19], rivaling that of protein enzymes
and thus they are ideal for fast sensing.
As a major component for sensors, nucleic acids possess many
advantages. First, DNA/RNA targeting essentially any molecule of
choice can be obtained through combinatorial selections [4–6, 10],
which provide a unique opportunity to construct a general sensing
platform for a broad range of analytes. Second, nucleic acids, par-
ticularly DNA, are very stable and can be denatured and renatured
many times without losing their binding abilities, allowing a long
shelf life. Third, nucleic acids have predictable base pairing interac-
tions, which have been proven to be very useful for rational sensor
design that are difficult to attain when developing protein or
organic molecule-based sensors. Finally, a broad range of DNA
modifications can be chemically synthesized with relatively low
cost, allowing convenient and precise conjugation. These proper-
ties make DNA and RNA ideal candidates to create sensors.
Since natural nucleic acids do not possess functional groups
that can generate absorption in the visible region, external signaling
labels need to be applied to convert them into sensors. To achieve
this goal, many inorganic nanoparticles have been employed. The
next few sections describe the properties of these nanoparticles and
preparation of a DNAzyme based lateral flow device using gold
nanoparticles.
1.2 Physical Depending on the composition, size, and shape of inorganic nano-
Properties of particles, a wide range of properties can be obtained. Inorganic
Nanoparticles metallic [20, 21], semiconductor [22], and magnetic nanoparticles
[23] can all be used to make functional sensors with different
detection modes. In this protocol, metallic nanoparticles are used.
Dipstick Metal ion Detection 393
1.3 Nanoparticle Many metal specific DNAzymes have been successfully converted
Conjugated DNAzymes into fluorescent sensors using a catalytic beacon technology [30]
for Sensing where the DNA enzyme and substrate are tagged with a combina-
Application tion of organic fluorophore and quencher. Sensors for metal ions
such as Pb2+ [13], UO22+ [17], Hg2+ [31], Cu2+ [32] are reported
in literature and these have been commercialized by ANDalyze Inc.
using a platform that consists of consumable fluorescent sensor
cartridges and a handheld fluorimeter.
Although fluorescent sensors are very sensitive and provide a
method of precise quantitative detection, colorimetric sensors have
been developed to make the detection possible by the naked eye.
In 1996, Mirkin and coworkers utilized the DNA induced assembly
of AuNPs to make a colorimetric sensor for nucleic acids [25].
Lu and coworkers expanded the scope of sensing to analytes
beyond nucleic acids, by combining AuNPs with DNAzymes [27,
28, 33, 34]. The sensing method is depicted using the Pb2+ depen-
dent 17E DNAzyme as a representative example (Fig. 2). AuNPs
functionalized with short thiol modified DNA are assembled on the
arms of the extended substrate which is in turn hybridized to the
enzyme. Since each AuNP is functionalized with many DNA
strands, blue aggregates are formed. In the presence of Pb2+, the
enzyme catalyzed cleavage of the substrate will disassemble the
aggregate producing red color. The color can be spotted on a
TLC plate and one such representative test is shown in the inset
of Fig. 2. Red color of increasing intensity is seen with Pb2+,
whereas the sensors containing other metals are blue. The reaction
is fast with color change occurring within 10 min under optimized
conditions and the detection limit is 100 nM.
Although not described in detail in this chapter, AuNP-based
aptamer sensors for various analytes, including several small organic
molecules are reported in literature [20, 33, 35–37]. The AuNPs
can also be replaced by other metallic nanoparticles, so that differ-
ent color changes can be achieved in the presence of different
394 Debapriya Mazumdar et al.
Fig. 2 DNAzyme based colorimetric sensor for Pb2+ detection. In the absence of Pb2+, the oligonucleotide
functionalized AuNPs are assembled on the substrate to form blue aggregates. When Pb2+ is present, the
substrate is cleaved and the aggregate is disassembled to yield red colored dispersed AuNPs. Inset: Picture of
sensor incubated with increasing concentrations of Pb2+ and with other metal ions (that are all 5 μM in
concentration)
1.4 Dipstick Tests While these colorimetric biosensors are an important step toward
Based on real-time sensing as the signal is detectable by the naked eye,
Immobilization of without the need for expensive instrumentation, they still require
DNAzyme-Gold laboratory type operations, such as precise transfer and mixing of
Nanoparticles onto a multiple solutions. In addition, although the sensitivity is high
Lateral Flow Device when the absorbance is recorded using a UV–Vis spectrophotome-
ter, it is often difficult to distinguish the red color of dispersed
nanoparticles against a blue background from the aggregates, par-
ticularly at low metal-ion concentrations. Furthermore, AuNPs are
not very stable in solution state and are vulnerable to aggregation
under a variety of conditions thereby making it difficult to store the
sensors for a long period of time.
Lateral flow devices are an ideal platform for making dipstick
type tests to further improve the performance of DNAzyme-AuNP
colorimetric sensors. In addition to eliminating precise solution
transfer and allowing separation of AuNPs to make it easier to
distinguish colors, the reagents can be prepared in a dry or nearly
dry state, making the device stable at ambient conditions for a long
period of time. The home pregnancy test is the most commonly
used lateral flow devices and several other antibody-based tests are
also well-known; however, DNA-based dipstick tests are not com-
mon. Glynou et al. reported a lateral flow device for the detection
of DNA [38]. To expand on the range of analytes detected, we
previously reported dipstick tests for the detection of adenosine and
cocaine using AuNPs conjugated to aptamers [39]. The protcol to
Dipstick Metal ion Detection 395
Fig. 3 (a) 8–17 DNAzyme reaction: In the presence of Pb2+, the 17E enzyme catalyzes the cleavage of the
chimeric substrate, 17S at the single ribo-linkage (rA). (b) Modified 8–17 construct conjugated to AuNPs
(called Enz-SubAuNP) used for the dipstick tests. B inside the purple box denotes biotin. (c) Assembled lateral
flow device. (d) Negative control: In the absence of Pb2+, AuNP-uncleaved substrate is captured at the control
zone via streptavidin-biotin interaction, producing a single red line. (e) Positive test: Substrate is cleaved in the
presence of Pb2+ and the AuNP-cleaved product migrates beyond the control zone to be captured at the test
zone by hybridization to complementary DNA. Two lines are produced
2 Materials
3 Methods
Fig. 4 Results of dipstick test for lead: (a) performance when the Enz-SubAuNP was pre-immobilized on the
conjugate pad and the Pb2+ reaction occurred on the surface of the device; (b) performance when Enz-
SubAuNP was allowed to react with Pb2+ in solution and then placed on the conjugate pad
4 Notes
Fig. 5 Constructs used for the optimization of dipstick test. Construct (c) had the best performance in the
presence of Pb2+ and was used for all other tests reported in this chapter. Construct (a) and (b) have different
arm lengths and worse performance comparing to construct (c)
Acknowledgment
References
1. Kruger K, Grabowski PJ, Zaug AJ, Sands J, selection of catalytic DNA. Comb Chem High
Gottschling DE, Cech TR (1982) Self-splicing Throughput Screen 5:327–335
RNA: autoexcision and autocyclization of the 16. Carmi N, Shultz LA, Breaker RR (1996) In
ribosomal RNA intervening sequence of Tetra- vitro selection of self-cleaving DNAs. Chem
hymena. Cell 31:147–157 Biol 3:1039–1046
2. Guerrier-Takada C, Gardiner K, Marsh T, Pace 17. Liu J, Brown AK, Meng X, Cropek DM, Istok
N, Altman S (1983) The RNA moiety of ribo- JD, Watson DB, Lu Y (2007) A catalytic bea-
nuclease P is the catalytic subunit of the con sensor for uranium with parts-per-trillion
enzyme. Cell 35:849–857 sensitivity and millionfold selectivity. Proc Natl
3. Breaker RR, Joyce GF (1994) A DNA enzyme Acad Sci U S A 104:2056
that cleaves RNA. Chem Biol 1:223–229 18. Vannela R, Adriaens P (2007) In vitro selection
4. Tuerk C, Gold L (1990) Systematic evolution of Hg (II) and As (V)-dependent RNA-cleaving
of ligands by exponential enrichment: RNA DNAzymes. Environ Eng Sci 24:73–84
ligands to bacteriophage T4 DNA polymerase. 19. Santoro SW, Joyce GF (1997) A general pur-
Science 249:505–510 pose RNA-cleaving DNA enzyme. Proc Natl
5. Ellington AD, Szostak JW (1990) In vitro Acad Sci U S A 94:4262–4266
selection of RNA molecules that bind specific 20. Liu J, Lu Y (2006) Fast colorimetric sensing of
ligands. Nature 346:818–822 adenosine and cocaine based on a general sen-
6. Wilson DS, Szostak JW (1999) In vitro selec- sor design involving aptamers and nanoparti-
tion of functional nucleic acids. Annu Rev Bio- cles. Angew Chem Int Ed Engl 45:90–94
chem 68:611–647 21. Liu J, Lu Y (2006) Preparation of aptamer-
7. Gold L, Polisky B, Uhlenbeck O, Yarus M linked gold nanoparticle purple aggregates for
(1995) Diversity of oligonucleotide functions. colorimetric sensing of analytes. Nat Protoc
Annu Rev Biochem 64:763–797 1:246–252
8. Breaker RR (2002) Engineered allosteric ribo- 22. Liu J, Lee JH, Lu Y (2007) Quantum dot
zymes as biosensor components. Curr Opin encoding of aptamer-linked nanostructures
Biotechnol 13:31–39 for one pot simultaneous detection of multiple
9. Hesselberth J, Robertson MP, Jhaveri S, analytes. Anal Chem 79:4120–4125
Ellington AD (2000) In vitro selection of 23. Yigit MV, Mazumdar D, Kim H-K, Lee JH,
nucleic acids for diagnostic applications. Rev Odintsov B, Lu Y (2007) Smart “turn-on”
Mol Biotechnol 74:15–25 magnetic resonance contrast agents based on
10. Famulok M (1999) Oligonucleotide aptamers aptamer-functionalized superparamagnetic
that recognize small molecules. Curr Opin iron oxide nanoparticles. Chembiochem
Struct Biol 9:324–329 8:1675–1678
11. Ihms HE, Lu Y (2012) In vitro selection of 24. Mirkin CA, Letsinger RL, Mucic RC, Storhoff
metal ion-selective DNAzymes. Methods Mol JJ (1996) A DNA-based method for rationally
Biol 848:297–316 assembling nanoparticles into macroscopic
12. Brown AK, Li J, Pavot CMB, Lu Y (2003) A materials. Nature 382:607–609
lead-dependent DNAzyme with a two-step 25. Storhoff JJ, Elghanian R, Mucic RC, Mirkin
mechanism. Biochemistry 42:7152–7161 CA, Letsinger RL (1998) One-pot colorimet-
13. Li J, Lu Y (2000) A highly sensitive and selec- ric differentiation of polynucleotides with sin-
tive catalytic DNA biosensor for lead ions. J gle base imperfections using gold nanoparticle
Am Chem Soc 122:10466–10467 probes. J Am Chem Soc 120:1959–1964
14. Li J, Zheng W, Kwon AH, Lu Y (2000) In vitro 26. Liu J, Lu Y (2006) Smart nanomaterials
selection and characterization of a highly effi- responsive to multiple chemical stimuli with
cient Zn(II)-dependent RNA-cleaving deoxyr- controllable cooperativity. Adv Mater
ibozyme. Nucleic Acids Res 28:481–488 18:1667–1671
15. Bruesehoff PJ, Li J, Augustine AJ, Lu Y (2002) 27. Liu J, Lu Y (2003) A colorimetric lead biosen-
Improving metal ion specificity during in vitro sor using DNAzyme-directed assembly of gold
406 Debapriya Mazumdar et al.
Abstract
Clinical and environmental analyses frequently necessitate rapid, simple, and inexpensive point-of-care or
field tests. These semiquantitative tests may be later followed up by confirmatory laboratory-based assays,
but provide an initial scenario assessment important for resource mobilization and threat confinement.
Lateral-flow assays (LFAs) and dip-stick assays, which are typically antibody-based and yield a visually
detectable signal, provide an assay format suiting these applications extremely well. Signal generation is
commonly obtained through the use of colloidal gold or latex beads, which yield a colored band either
directly proportional or inversely proportional to the concentration of the analyte of interest. Here, dye-
encapsulating liposomes as a highly visible alternative are discussed. The semiquantitative LFA biosensor
described in this chapter relies on a sandwich immunoassay for the detection of myoglobin in whole blood.
After an acute myocardial infarction (AMI) event, several cardiac markers are released into the blood, the
most common of which are troponin, creatine kinase MB, C-reactive protein, and myoglobin. Due to its
early release, myoglobin has value as an indicator of a recent heart attack amongst conditions which present
with similar symptoms and its lack of elevation can effectively rule out a heart attack (Brogan et al., Ann
Emerg Med 24:665–671, 1994). The assay described within relies on sandwich complex formation
between a membrane immobilized capture monoclonal antibody against myoglobin, a detector biotiny-
lated monoclonal antibody against a different epitope on myoglobin, and streptavidin-conjugated visible
dye (sulforhodamine B)-encapsulating liposomes to allow for signal generation.
1 Introduction
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_25,
© Springer Science+Business Media LLC 2017
407
408 Katie A. Edwards et al.
0.1
Fig. 1 Temporal release of myoglobin, CK-MB, and cTnT and cTnI. Reprinted with
permission from Christenson RH, Azzazy HME. Biomarkers of Myocardial Necro-
sis: Past, Present and Future. In Morrow DA, ed. Cardiovascular Biomarkers:
Pathophysiology and Clinical Management. Totowa, NJ: Humana Press, 2006
1.1 Sandwich While simple for the end-user to operate, from an engineering
Immunoassay Format standpoint, the sandwich lateral flow immunoassay format is a
relatively complicated multicomponent design encompassing a
sample pad, conjugate pad, analytical membrane, absorbent pad,
signaling species, blocking constituents, and multiple antibodies.
When these components are assembled appropriately, the end-user
simply needs to add a liquid sample and wait typically 5–15 min
before a visible signal is assessed. The flow of the fluid in such an
assay is detailed in Fig. 2.
A liquid sample ideally with minimal preparation requirements
is applied to the sample pad via an opening in the housing. Depend-
ing on the sample matrix, the sample pad can serve as a particulate
filter, site to retain interfering substances, and provide pH buffering
capacity with the overall aim to prepare the fluid for subsequent
analysis and dispense it evenly. Once the fluid passes through the
sample pad, it then passes through a conjugate pad where it rehy-
drates a previously dehydrated signaling species, which is typically
410 Katie A. Edwards et al.
Fig. 2 Lateral flow assay fluid path. (Top) A liquid sample is applied to a sample pad through a port in the
plastic housing (not shown). The liquid sample then passes through the conjugate pad where the signaling
species (most commonly colloidal gold) with an attached detection antibody becomes rehydrated. The solution
then passes onto the analytical membrane were capture and control antibodies are immobilized, then lastly is
wicked by the absorbent pad at the opposite end of the assembly. (Middle) If the analyte is present, it can form
sandwich complexes with the detection antibody on the colloidal gold and the capture antibody immobilized
on the analytical membrane. A visible signal where the capture antibody is immobilized is observed as well as
a visible signal where the control antibody is immobilized to indicate successful fluid flow and conjugate
release. (Bottom) If the analyte is not present, a signal at only the control line is observed
colloidal gold or visibly dyed latex beads. The surface of the signal-
ing species is functionalized with “detection” antibodies against the
analyte of interest and can form antibody-antigen complexes with
analyte in the sample solution if it is present. The fluid then passes
onto the analytical membrane where a second antibody is immobi-
lized at the test line. This “capture” antibody, most commonly a
monoclonal antibody (mAb), can recognize a different epitope on
the target analyte than that recognized by the detection antibody
and serves to retain the passing analyte-signaling particle complexes.
The sandwich complex formed between the immobilized capture
antibody, analyte, and detection antibody-tagged particles allows a
visible signal to be observed in the presence of the target analyte due
to the attached signaling species. When the analyte is not present, no
sandwich complexes are formed and thus no signal is generated. The
intensity of the signal is proportional to the analyte concentration,
which can be used for semiquantitative determinations. At a location
downstream of the test line is a control line. This control line
typically consists of a secondary antibody, which can bind to the
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 411
1.1.1 Membrane Types Most commonly, nitrocellulose membranes are used as analytical
membranes for LFAs due to their high binding capacity, low cost,
and wide availability [35]. The mechanism behind protein binding
to nitrocellulose is not fully understood, but is believed to be non-
covalent through hydrophobic, hydrogen bonding, and electro-
static interactions [36, 37]. The physical adsorption of nucleic
acid probes through drying to nitrocellulose is believed to result
in attachment through hydrophobic interactions, an effect which
may be enhanced through modification with a poly-T tail [38].
We have also had success using polyethersulfone membranes for
nucleic acid sandwich hybridization assays [22, 23, 39].
More recently, membranes based on electrospun nanofibers
have been applied for LFAs [40]. Electrospun nanofibers are
prepared by using a simple set-up consisting of a power voltage
supply, a syringe with metal needle, a syringe pump and a ground
plate. A polymer solution (dope) is filled into the syringe and
pumped out at a steady flow rate. A high voltage (7–15 kV) is
applied between the tip of the needle and the ground plate. A taylor
cone is formed and a nanofiber in the size range of a few tens of
nanometers to a few micrometers are collected on the grounded
plate, depending on the spinning dope and electrospinning condi-
tions chosen. Nitrocellulose and nylon via electrospinning have been
shown for protein slot blot and Western blot applications [41].
By doping the composite with poly-L-lysine (PLL) or polylactic
acid (PLA), specific functional groups including amino and carbox-
ylic acid groups, respectively, may be incorporated [42]. Additionally,
through control over polymer dope composition, voltage, distance
between needle tip and ground plate control over the mat thickness
and porosity can be achieved and offers significant surface area for
immobilization.
1.1.2 Membrane The ideal properties of the membrane include particle and pore size
Properties consistency, hydrophilicity, high protein binding, and durability.
As nitrocellulose membranes are a fibrous polymeric matrix rather
than uniform particles, the membranes are characterized by their
capillary rise rate (or capillary flow time), rather than by a specific
particle or pore size. This rate is the amount of time required for fluid
to flow 4 cm, and based on a survey of current commercial offerings
from major manufacturers, is generally between 60 and 280 s. Since
fluid flow speed in lateral flow membranes decreases with the
distance traveled, the capillary rise rate is a more consistent measure
of the speed of flow [43]. This parameter is important to consider
during assay development since a fast wicking rate allows for more
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 413
rapid results and lower background, but may adversely affect assay
sensitivity since less time for interaction between the target and the
antibodies immobilized at the test line is available [44].
Commercially available nitrocellulose membranes vary in phys-
ical properties based on the effective “pore size,” the amount of
trapped air, and the thickness of their nitrocellulose layer. These
properties affect the surface area available for immobilization of
capture reagents. The binding capacity of nitrocellulose itself
is inherently high, with estimates ranging from 50 to 200 μg
IgG/cm2 [45]. The thickness of commercially available membranes
typically ranges from 100 to 150 μm. Thicker membranes can allow
more sample fluid to be accommodated by the membrane during the
assay and more capture reagent to be applied. However, the latter
does not necessarily yield greater sensitivity in lateral flow assays with
optical detection since due to the opacity of nitrocellulose, only the
top 10 μm of membrane is visible to the user [45]. One advantage
to thicker membranes is improved tensile strength, however, for ease
of handling, durability, and avoidance of adhesive interactions, a
supported membrane should be chosen. These membranes are man-
ufactured by directly casting the nitrocellulose membrane material
onto a plastic (polyester or cellulose acetate) backing. The backing
thickness typically ranges from 50 to 225 μm [43]. Alternatively,
non-supported membranes can be laminated manually prior to use.
LFAs may be further made more durable and user-friendly by encas-
ing them in an injected molded plastic housing such as that shown in
Fig. 3.
1.2 Detection LFAs typically utilize antibody-labeled colloidal gold [5, 19, 50] as
Mechanisms a visualization method. Colloidal gold ranges in size 2–250 nm,
though 30–80 nm particles are preferred for LFAs [24]. A diameter
of 40 nm has been reported to be optimal, allowing for clear
visualization, dense packing of these small particles at the capture
zone, and reduced steric hindrance for protein binding [51]. These
particles have a characteristic red color, allowing for visual detection
or semiquantitative measurements with portable reflectometers or
scanners [52–54]. Proteins are associated with gold particles ioni-
cally through the particle’s negative charge with positively charged
amino acids such as lysine; through hydrophobic interactions of
amino acids such as tyrosine and tryptophan; and through sharing
of electrons between gold and sulfur atoms of cysteine [51]. When
labeled with anti-biotin or streptavidin, such species allow for rec-
ognition of biotinylated biorecognition elements, such as DNA
oligonucleotides or antibodies. The sensitivity of LFAs using such
particles can be increased with silver enhancement [55]. Alterna-
tively, antibody-tagged latex particles [56, 57], up-converting
phosphors [58], superparamagnetic particles [59], or visible dye-
encapsulating liposomes [60] have been used as a signal enhance-
ment means. The latter species are the focus of this chapter.
Liposomes are vesicles formed through the association of phos-
pholipid molecules in an aqueous environment, yielding a structure
with the hydrocarbon tails forming a lipid bilayer and hydrophilic
headgroups directed at both the aqueous core and external aqueous
medium. One of the common methods for liposome formation is
known as the reverse-phase evaporation method [61, 62]. Here,
phospholipids are dissolved in organic solvent; the mixture is intro-
duced to an aqueous medium containing high concentrations of
visible dye; the solvent is removed under vacuum; the resulting
liposomes are passed through defined pore-size membranes to
improve homogeneity and reduce the number of lipid bilayers
(lamellarity); then the unencapsulated material is removed through
size-exclusion chromatography and dialysis. We commonly use
sulforhodamine B (SRB) dye, which is relatively inexpensive, highly
416 Katie A. Edwards et al.
Fig. 5 Liposome structure. (a) Biorecognition elements can be covalently conjugated to or inserted into lipid
bilayers through hydrophobic interactions (not to scale). (b) The large internal volume of unilamellar vesicles
can encapsulate hundreds of thousands of hydrophilic signaling molecules and provide for their stability. (c)
Surfactant introduction can provide for instantaneous signal enhancement through release of encapsulants.
Fluorophores encapsulated within liposomes at high concentrations undergo self-quenching with is overcome
upon release into the surrounding medium
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 417
1.3 Universal Assays Both immunoassays and nucleic acid based lateral flow assays can
also be made in a universal format, obviating the need for the
generation of specifically labeled membranes and detection
elements. This can be done through membrane immobilization of
streptavidin [23] or anti-fluorescein antibodies [22] and
subsequent addition of biotinylated or fluorescein-labeled capture
antibodies, respectively, with the remaining assay components.
Protein A/G [67], streptavidin/avidin [22, 68], or generic oligo-
nucleotide [23] labels can be conjugated to the liposomes to form
generic species capable of facile recognition of the Fc0 region of
antibodies, biotinylated biorecognition elements, or complemen-
tary generic oligonucleotides, respectively. Anti-fluorescein, anti-
biotin, or anti-digoxigenin are also options for common universal
liposome tags [69]. These assays are of particular interest in
research laboratories where a variety of antibodies or probes need
to be screened or where different analytes need to be tested. From a
commercial and manufacturing standpoint, the universal format is
of interest since only one type of membrane and liposome need to
be prepared which simplifies packaging of tests for any analyte.
2 Materials
Materials listed here are those that have been used successfully in
our laboratory, though substitutions may be made. Unless other-
wise specified, reagents are molecular biology grade and are pur-
chased from VWR (Bridgeport, NJ). Table 1 lists the biomolecules
used for the detection of myoglobin as an example.
2.1 Liposome 1. Bath sonicator (Aquasonic Model 150D, VWR, Bridgeport, NJ).
Preparation 2. Rotary evaporator (Model R-114, Buchi, New Castle, DE).
3. 50-mL round bottom flask (Catalog # 80068-756, VWR).
4. Mini-extruder (Catalog # 610000, Avanti Polar Lipids, Alabas-
ter, AL), including two 1 mL syringes, Teflon supports and o-
rings, and extruder holder/heating block. When purchasing
initially, the membranes and supports listed in #5 of this section
are included with this catalog number as of the date that this
chapter was prepared.
5. Extrusion membranes (0.4 and 1.0 μm polycarbonate mem-
branes*, 19 mm) and filter supports (Catalog # 610007,
610010, and 610014, respectively, Avanti Polar Lipids).
6. 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), choles-
terol, 1,2-dipalmitoyl-sn-glycero-3-[phospho-rac-(1-glycerol)],
sodium salt (DPPG), 1,2-dipalmitoyl-sn-glycero-3-phos-
phoethanolamine-N-glutaryl, sodium salt (N-glutaryl DPPE)
(Catalog #: 850355, 700000, 840455, and 870245, respec-
tively, Avanti Polar Lipids).
418 Katie A. Edwards et al.
Table 1
Reagents needed for the development of a LFA for human myoglobin
3 Methods
3.1 Liposome A flowchart of this process is shown in Scheme 1. Note that the
Preparation times listed in this flowchart, and others in this chapter, reflect
overall times for each step, including setup and incubations. For
specific times for each step, please refer to the text.
Scheme 2 Flow chart for the preparation of lateral flow membranes with immobilized capture antibodies
3.2.1 Automated The following instructions are for operation of the CAMAG Lino-
Application of Capture mat IV, but should be adaptable for similar applicators using the
Antibodies Using a Linomat respective manufacturer’s directions.
IV or Similar TLC Plate
5. Set the Limonat IV parameters as follows:
Applicator
6. Fill the syringe slowly with the capture antibody solution mix-
ture to avoid air bubbles, then insert syringe fully into syringe
holder.
424 Katie A. Edwards et al.
3.2.2 Manual Application If a TLC or similar applicator is not available, the capture antibody
of Capture Antibodies Using solution may be applied manually to the lateral flow membranes.
a Pipettor Here, a round spot for the capture and control zones would result,
versus the line seen in Fig. 3 generated using the Linomat for
antibody application. This procedure is useful for optimization
purposes, but is more laborious than the TLC applicator procedure
due to the need to apply antibodies and tape individual membranes.
1. Cut the membrane sections into 4.5 mm pieces. We’ve found it
beneficial to tape a laminated piece of paper to the paper cutter
with lines every 4.5 mm for a guide. Then, align one end of one
section with this paper and advance the section, making per-
pendicular cuts every 4.5 mm.
2. Using tape with the sticky side exposed placed onto a piece of
cardboard, lightly tape the top of each membrane to a piece of
clean cardboard using tweezers.
3. Apply 1 μL of the capture antibody solution to each membrane
3.0 cm from the bottom of the strip using a microvolume
pipettor. The more consistent the antibody application is, the
more consistent the LFA results will be. The antibody solution
will rapidly seep into the membrane.
4. Deposit the control zone 3.5 cm from the base of the mem-
brane card following the same procedure.
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 425
3.2.3 Membrane Drying 5. Place the membranes or membrane cards in the vacuum oven at
and Blocking Steps 40 C and set vacuum to 1500 Hg for 1.5 h. Alternatively,
membranes or membrane cards may be stored overnight in a
tightly sealed Tupperware container with the bottom filled to
0.500 with Drierite and covered with layers of large Kimwipes.
6. Remove membranes from vacuum oven or desiccated storage
container.
7. Pour blocking reagent into a plastic Tupperware container so
that it is <50% full.
8. Using tweezers, gently remove each membrane or membrane
card and place in the blocking reagent laminated side down. Be
sure to allow each membrane to gradually sink into your block-
ing reagent, otherwise blocking will be uneven. You can gently
help them along by pressing the ends under the solution.
9. Place container on shaker using slow agitation for 30 min. The
speed should be set such that the membranes are being covered
by solution and moving freely, but not so much that they
become bunched up and solution comes over the sides.
10. Using tweezers, remove each membrane or membrane section
and lie flat on three layers of large Kimwipes. Place 2–3 layers of
Kimwipes on top of membranes, then add a heavy flat object on
top of the membranes, such as a heavy lab supplies catalog.
11. Retape membranes back onto the cardboard.
12. Return the taped membranes to the vacuum oven at 25–30 C
and set vacuum to 1500 Hg for 3–4 h.
13. Remove membrane strips or sheets from cardboard and store in
vacuum sealed bags at 4 C or as above in step 17 at in sealed
Tupperware container at ambient temperature.
LFA assembly steps
14. Cut the absorbent pad into 1.8 20.0 cm sections
corresponding to the number of membrane cards prepared in
step 2.
15. Gently peel away the film covering the top section of the
HF090 card.
16. Apply the absorbent pad such that it overlaps the nitrocellulose
by 2 mm (Fig. 6).
3.0 cm 0.5 cm
4.5 mm
1.9 cm 2 mm 2 mm 1.8 cm
Fig. 6 (Left) Layout of assembled LFA membrane showing overlaps and positions of sample and absorbent
pads and antibody deposition zones. (Right) Image of assembled HF090 membrane with VFE unbound glass
sample pad and cellulose fiber absorbent pad before insertion into cassette
426 Katie A. Edwards et al.
17. Apply gentle, but even, pressure along the length of the absor-
bent pad to secure it to the underlying adhesive.
18. If the membranes are to be used for assay optimization pur-
poses only in the absence of whole blood, the bottoms may be
cut off. To cut off the bottoms, align the paper cutter such that
the lowest 0.5 mm of nitrocellulose will be removed when the
cut is made (see Note 5).
19. The bottom of the HF090 membrane cards can accommodate
both a conjugate and a sample pad. For the assays described
within, only a sample pad is used. Prior to assembly, this sample
pad is cut into 1.9 20 cm sections and blocked with 0.05%
(w/v) casein, 0.25% (w/v) sucrose for 30 min and dried in a
vacuum oven at 21 C and 1500 Hg overnight.
20. Gently peel away the film covering the bottom sections of the
HF090 card.
21. Apply the sample pad such that it overlaps the nitrocellulose by
2 mm.
22. Apply gentle, but even, pressure along the length of the sample
pad to secure it to the underlying adhesive. Excessive pressure
may crush the delicate sample pads and impede flow of the
samples.
23. The assembled membrane sheets may be cut into strips of
desired widths prior to storage as described in step 12. Our
membranes are typically cut into 4.5 mm widths.
3.3.1 Bartlett Assays for 1. Liposome samples (20 μL) in triplicate are dehydrated at
Phospholipid Content 180 C for 10 min, then mixed and heated with 1.5 mL of
3.33 N H2SO4 for 2 h at the same temperature. Standards
prepared from potassium phosphate dibasic in deionized
water (16, 32, 64, 128, and 256 nmol phosphate per tube in
triplicate) are subjected concurrently to the same procedure.
Note that inorganic phosphates will interfere with the Bartlett
assay and need to be considered if these are used in alternate
buffers or diluents used for liposome preparation.
2. 100 μL of 30% hydrogen peroxide is added and the mixture is
returned to the oven for 1.5 h. The tubes are permitted to cool
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 427
3.4 Liposome The liposome conjugation procedure assumes that the phospho-
Conjugation lipid concentration, determined as described in Subheading 3.3 or
by other means, is known. The procedure described within is has
been optimized for the conjugation of streptavidin to liposomes
and may be modified to accommodate other proteins by varying the
EDC and protein concentrations. It utilizes EDC to form a zero-
length linkage between carboxylic acid groups on the liposomes
and protein amine groups. Before beginning this procedure, ensure
that the protein to be conjugated is purified and does not contain
any stabilizing proteins or buffer components with amine groups.
1. Remove EDC from 20 C storage and allow to warm to
ambient temperature while proceeding with the following
steps.
2. Add desired volume of liposomes prepared in Subheading 3.1
to a 1.5-mL microcentrifuge tube.
3. Using the known phospholipid concentration, calculate the
nmol of total lipid present. Be sure to account for non-
phospholipid constituents such as cholesterol in your
calculation.
4. Determine the amount of protein needed for conjugation.
Typically, 0.05 mol% of the total lipid content is a reasonable
amount.
428 Katie A. Edwards et al.
3.5 Assay The steps within this section are helpful for determining the best
Optimization Using conditions for antibody/antigen interaction and signaling. Here,
Recombinant the buffer composition, biotinylated antibody, and liposome con-
Myoglobin centrations may be varied to attain the desired level of detection.
This information can be utilized to streamline the development of
the assay in whole blood. If needed, the capture antibody concen-
tration can also be optimized during membrane preparation which
can be done easily if the manual application approach is used.
1. Add 10 μL myoglobin solutions to the bottom of wells of a flat-
bottom, non-binding microtiter plate (see Note 7).
2. Insert a 4.5 mm membrane strip with immobilized capture and
control antibodies with the base cut off and allow volume to
wick up membrane.
3. Once the solution has completely wicked, the membrane strips
are transferred to wells containing 10 μL of MESS with 0.1%
(w/v) BSA.
4. Once this solution has wicked, membranes are transferred to
wells containing a solution with 6 μg/mL biotinylated anti-
myoglobin antibodies and liposomes diluted to 250 μM
phospholipid.
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 429
3.6 Assay Format In our studies, 16 mL of human venous blood was drawn into EDTA
for the Analysis vacutainer tubes by venipuncture at the Gannett Health Center at
of Myoglobin in Whole Cornell University. Participants provided informed consent for this
Blood study, which was approved by the institutional review board at Cornell
University. No participant identifier was associated with each sample.
Important: Human blood should be treated as it is potentially
infectious and proper precautions for handling and disposal should
be followed, in accordance with your organization’s protocols.
Obtaining human blood samples also requires informed consent
of your subjects, in accordance with your organization’s institu-
tional review board.
1. Fully assembled membrane strips with blocked sample pads,
immobilized capture and control antibodies, and absorbent
pads are inserted into a commercially available cassette. The
dimensions of the absorbent pad may be adjusted to ensure
that the pressure points of the cassette line up with the sample
and absorbent pad overlaps; the sample hole lines up with the
sample pad; and the test and control lines are visible.
2. Whole blood is spiked with 0–350 ng/mL myoglobin.
3. 50 μL of the spiked blood is added to 50 μL of an antibody/
liposome solution and is gently vortexed for 5 min.
4. 100 μL of the blood mixture is transferred to the sample pad.
5. Allow assay to run for 15 min. A signal proportional to the
myoglobin concentration should result (Fig. 7).
3.7 Analysis The membranes may be read by eye for qualitative results, however,
of Results semiquantitative results may be obtained if a scanner, digital cam-
era, or reflectometer is available. After the membranes have fully
dried (typically 20–30 min), scan an image of the membranes using
a computer and flatbed scanner, then use any available densitome-
try software to quantify band intensity. Alternatively, if a reflectom-
eter is available, the readings should be taken at this time following
the instrument manufacturer’s instructions. For optimal reproduc-
ibility and sensitivity, the intensity of the bands should be read as
soon as the membranes dry as it tends to decrease over time (days to
weeks) and exposure to light. While membranes may also be
scanned or read using a reflectometer when wet, care must be
taken to avoid leaving impressions in the nitrocellulose. Whether
read wet or dry, consistency in the timing of measurement is key for
optimal reproducibility.
430 Katie A. Edwards et al.
Fig. 7 Whole-blood lateral flow assay at spiked myoglobin concentrations ranging from 350 ng/mL (left) to
0 ng/mL. 50 μL blood samples were spiked with various myoglobin concentrations and added to 50 μL of an
antibody/liposome solution. This mixture was gently vortexed, and after 5 min, all 100 μL were added to the
sample pad. Results were observed after 15 min. A visible signal at the test line could be seen at a spiked
myoglobin concentration of 75 ng/mL (see Notes 8 and 9)
4 Notes
Acknowledgments
References
1. Helbig JH, Luck PC, Kunz B, Bubert A (2006) membrane strip tests compared. Urology 53
Evaluation of the Duopath Legionella lateral (1):155–160
flow assay for identification of Legionella pneu- 14. http://www.brittneylimited.com/PDF%20Files/
mophila and Legionella species culture isolates. PSA-Prostrate-strip.pdf
Appl Environ Microbiol 72(6):4489–4491 15. http://www.nhdiag.com/botulism.shtml
2. Koide M, Haranaga S, Higa F, Tateyama M, 16. Sharma SK, Eblen BS, Bull RL, Burr DH,
Yamane N, Fujita J (2007) Comparative evalu- Whiting RC (2005) Evaluation of lateral-flow
ation of Duopath Legionella lateral flow assay clostridium botulinum neurotoxin detection
against the conventional culture method using kits for food analysis. Appl Environ Microbiol
Legionella pneumophila and Legionella anisa 71(7):3935–3941
strains. Jpn J Infect Dis 60(4):214–216
17. Park CH, Kim HJ, Hixon DL, Bubert A
3. http://www.kit.nl/biomedical_research/assets/ (2003) Evaluation of the duopath verotoxin
images/Lepto_lateral_flow_protocol.doc test for detection of shiga toxins in cultures of
4. Johnston SP, Ballard MM, Beach MJ, Causer human stools. J Clin Microbiol 41
L, Wilkins PP (2003) Evaluation of three com- (6):2650–2653
mercial assays for detection of Giardia and 18. Capps KL, McLaughlin EM, Murray AWA et al
Cryptosporidium organisms in fecal specimens. (2004) Validation of three rapid screening
J Clin Microbiol 41(2):623–626 methods for detection of verotoxin-producing
5. Ketema F, Zeh C, Edelman DC, Saville R, Escherichia coli in foods: interlaboratory study.
Constantine NT (2001) Assessment of the per- J AOAC Int 87(1):68–77
formance of a rapid, lateral flow assay for the 19. Smits HL, Eapen CK, Sugathan S et al (2001)
detection of antibodies to HIV. J Acquir Lateral-flow assay for rapid serodiagnosis of
Immune Defic Syndr 27(1):63–70 human leptospirosis. Clin Diagn Lab Immunol
6. Cazacu AC, Demmler GJ, Neuman MA et al 8(1):166–169
(2004) Comparison of a new lateral-flow chro- 20. Kassler WJ, Dillon BA, Haley C, Jones WK,
matographic membrane immunoassay to viral Goldman A (1997) On-site, rapid HIV testing
culture for rapid detection and differentiation with same-day results and counseling. AIDS 11
of influenza A and B viruses in respiratory spe- (8):1045–1051
cimens. J Clin Microbiol 42(8):3661–3664
21. Prevention USCfDCa (1996) Provisional pub-
7. Mokkapati VK, Sam Niedbala R, Kardos K et al lic health service recommendations for chemo-
(2007) Evaluation of UPlink-RSV: prototype prophylaxis after occupational exposure to
rapid antigen test for detection of respiratory HIV. MMWR Morb Mortal Wkly Rep
syncytial virus infection. Ann N Y Acad Sci 45:468–480
1098:476–485
22. Baeumner AJ, Jones C, Wong CY, Price A
8. Slinger R, Milk R, Gaboury I, Diaz-Mitoma F (2004) A generic sandwich-type biosensor
(2004) Evaluation of the QuickLab RSV test, a with nanomolar detection limits. Anal Bioanal
new rapid lateral-flow immunoassay for detec- Chem 378(6):1587–1593
tion of respiratory syncytial virus antigen. J
Clin Microbiol 42(8):3731–3733 23. Baeumner AJ, Pretz J, Fang S (2004) A univer-
sal nucleic acid sequence biosensor with nano-
9. van Hengel AJ, Capelletti C, Brohee M, molar detection limits. Anal Chem 76
Anklam E (2006) Validation of two commer- (4):888–894
cial lateral flow devices for the detection of
peanut proteins in cookies: interlaboratory 24. Seal J, Braven H, Wallace P (2006) Point-of-
study. J AOAC Int 89(2):462–468 care nucleic acid lateral-flow tests. IVD Tech-
nol 8:41–54
10. http://www.craigmedical.com/salivascreen6_
drug_tests.htm 25. Tomoda H, Aoki N (2000) Prognostic value of
C-reactive protein levels within six hours after
11. http://www.brittneylimited.com/PDF%20 the onset of acute myocardial infarction. Am
Files/FDA_Files/Pregnancy-strip.pdf Heart J 140(2):324–328
12. Tiplady S (2013) Lateral flow and consumer 26. Qureshi A, Gurbuz Y, Niazi JH (2012) Bio-
diagnostics. In: Wild D (ed) The immunoassay sensors for cardiac biomarkers detection: a
handbook, 4th edn. Elsevier, Oxford, U.K. review. Sens Actuators B Chem
13. Jung K, Zachow J, Lein M et al (1999) Rapid 171–172:62–76
detection of elevated prostate-specific antigen 27. Anderson JL, Adams CD, Antman EM et al
levels in blood: performance of various (2011) 2011 ACCF/AHA Focused Update
Liposome-Enhanced Lateral-Flow Assays for Clinical Analyses 433
Incorporated Into the ACC/AHA 2007 40. Reinholt SJ, Sonnenfeldt A, Naik A, Frey MW,
Guidelines for the Management of Patients Baeumner AJ (2014) Developing new materi-
With Unstable Angina/Non-ST-Elevation als for paper-based diagnostics using electro-
Myocardial Infarction: A Report of the Ameri- spun nanofibers. Anal Bioanal Chem 406
can College of Cardiology Foundation/Amer- (14):3297–3304
ican Heart Association Task Force on Practice 41. Manis AE, Bowman JR, Bowlin GL, Simpson
Guidelines. Circulation 123(18):e426–e579 DG (2007) Electrospun nitrocellulose and
28. Brogan GX Jr, Friedman S, McCuskey C et al nylon: design and fabrication of novel high
(1994) Evaluation of a new rapid quantitative performance platforms for protein blotting
immunoassay for serum myoglobin versus CK- applications. J Biol Eng 1:2–2
MB for ruling out acute myocardial infarction 42. Matlock-Colangelo L, Baeumner AJ (2012)
in the emergency department. Ann Emerg Recent progress in the design of nanofiber-
Med 24(4):665–671 based biosensing devices. Lab Chip 12
29. McCord J, Nowak RM, McCullough PA et al (15):2612–2620
(2001) Ninety-minute exclusion of acute myo- 43. Mansfield M (2009) Nitrocellulose membranes
cardial infarction by use of quantitative point- for lateral flow immunoassays: a technical trea-
of-care testing of myoglobin and troponin I. tise. In: Wong RC, Tse HY (eds) Lateral flow
Circulation 104(13):1483–1488 immunoassay. Humana Press, New York, pp
30. Alexander LL (2016) Early prediction of acute 95–113
myocardial infarction from clinical history, 44. Bangs LI (1999) Lateral flow tests. Vol Tech
examination, and electrocardiogram in the Note 303:1–6
emergency room. Ann Emerg Med 20 45. Millipore. Rapid lateral flow test strips—
(11):1270 considerations for product development.
31. Schiff HB, MacSearraigh ET, Kallmeyer JC http://www.emdmillipore.com/US/en/
(1978) Myoglobinuria, rhabdomyolysis and products/ivd-oem-materials-reagents/lateral-
marathon running. Q J Med 47(188):463–472 flow-membranes/n6mb.qB.L0YAAAE_gut3.
32. C-Peptide ELISA Kit ab178641. www.abcam. Lxi,nav?bd¼1. Accessed 18 Mar 2016
com/C-Peptide-ELISA-Kit-ab178641.pdf. 46. Batteiger B, Newhall WJ, Jones RB (1982) The
Accessed 23 Mar 2016 use of Tween 20 as a blocking agent in the
33. Brown M (2008) Antibodies: Key to a robust immunological detection of proteins trans-
lateral flow immunoassay. In: Wong RC, Tse ferred to nitrocellulose membranes. J Immunol
HY (eds) Lateral flow immunoassay. Humana Methods 55(3):297–307
Press, New York 47. Jones KD (1999) Troubleshooting protein
34. O’Farrell B (2013) Lateral flow immunoasay binding in nitrocellulose membranes. Part I:
systems: Evolution from the current state of Principles. IVD Technol 5:32–41
the art to the next generation of highly sensi- 48. Van Dam AP, Van den Brink HG, Smeenk RJ
tive, quantitative rapid assays. In: Wild D (ed) (1990) Technical problems concerning the use
The immunoassay handbook, 4th edn. Else- of immunoblots for the detection of antinu-
vier, Oxford clear antibodies. J Immunol Methods 129
35. O’Farrell B, Bauer J (2006) Developing highly (1):63–70
sensitive, more-reproducible lateral-flow 49. Zampieri S, Ghirardello A, Doria A et al (2000)
assays. Part 1: New approaches to old pro- The use of Tween 20 in immunoblotting assays
blems. IVD Technol 10(6):11–15 for the detection of autoantibodies in connec-
36. Hans H, Beer EJ, Pflanz K, Klewitz TM (2002) tive tissue diseases. J Immunol Methods 239
Qualification of cellulose nitrate membranes (1–2):1–11
for lateral-flow assays. IVD Technol 8:35–42 50. Smits HL, Chee HD, Eapen CK et al (2001)
37. Harlow E, Lane D (1988) Antibodies: a labo- Latex based, rapid and easy assay for human
ratory manual. Cold Spring Harbor, Cold leptospirosis in a single test format. Trop Med
Spring Harbor Laboratories Int Health 6(2):114–118
38. Jones K (2001) Membrane immobilization of 51. Carney J, Braven H, Seal J, Whitworth E
nucleic acids. Part 2: Probe attachment techni- (2006) Present and future applications of gold
ques. IVD Technol pp 1–7 in rapid assays. IVD Technol 12:41–49
39. Edwards KA, Baeumner AJ (2006) Optimiza- 52. Chan CP, Sum KW, Cheung KY et al (2003)
tion of DNA-tagged dye-encapsulating lipo- Development of a quantitative lateral-flow
somes for lateral-flow assays based on assay for rapid detection of fatty acid-binding
sandwich hybridization. Anal Bioanal Chem protein. J Immunol Methods 279
386:1335–1343 (1–2):91–100
434 Katie A. Edwards et al.
53. Wang S, Zhang C, Wang J, Zhang Y (2005) sandwich-hybridization assays. Anal Biochem
Development of colloidal gold-based flow- 244(2):260–269
through and lateral-flow immunoassays for 65. Rongen HAH, Bult A, vanBennekom WP
the rapid detection of the insecticide carbaryl. (1997) Liposomes and immunoassays. J
Anal Chim Acta 546(2):161–166 Immunol Methods 204(2):105–133
54. Klewitz T, Gessler F, Beer H, Pflanz K, Scheper 66. Edwards KA, Baeumner AJ (2013) Periplasmic
T (2006) Immunochromatographic assay for binding protein-based detection of maltose
determination of botulinum neurotoxin type using liposomes: a new class of biorecognition
D. Sens Actuat Chem 113(2):582–589 elements in competitive assays. Anal Chem 85
55. Shyu RH, Shyu HF, Liu HW, Tang SS (2002) (5):2770–2778
Colloidal gold-based immunochromato- 67. Chen CS, Baeumner AJ, Durst RA (2005) Pro-
graphic assay for detection of ricin. Toxicon tein G-liposomal nanovesicles as universal
40(3):255–258 reagents for immunoassays. Talanta 67
56. Greenwald R, Esfandiari J, Lesellier S et al (1):205–211
(2003) Improved serodetection of Mycobacte- 68. Plant AL, Brizgys MV, Locasio-Brown L,
rium bovis infection in badgers (Meles meles) Durst RA (1989) Generic liposome reagent
using multiantigen test formats. Diagn Micro- for immunoassays. Anal Biochem 176
biol Infect Dis 46(3):197–203 (2):420–426
57. Birnbaum S, Uden C, Magnusson CG, Nilsson 69. Edwards KA, Baeumner AJ (2007) DNA-
S (1992) Latex-based thin-layer immunoaffi- oligonucleotide encapsulating liposomes as a
nity chromatography for quantitation of pro- secondary signal amplification means. Anal
tein analytes. Anal Biochem 206(1):168–171 Chem 79(5):1806–1815
58. Corstjens P, Zuiderwijk M, Brink A et al 70. Koppel DE (1972) Analysis of macromolecular
(2001) Use of up-converting phosphor repor- polydispersity in intensity correlation
ters in lateral-flow assays to detect specific spectroscopy—method of Cumulants. J Chem
nucleic acid sequences: a rapid, sensitive DNA Phys 57(11):4814–4820
test to identify human papillomavirus type 16 71. Frisken BJ (2001) Revisiting the method of
infection. Clin Chem 47(10):1885–1893 cumulants for the analysis of dynamic light-
59. LaBorde R, O’Farrell B (2002) Paramagnetic- scattering data. Appl Opt 40(24):4087–4091
particle detection in lateral-flow assays. IVD 72. Fiske CH, Subbarow Y (1925) The colorimet-
Technol 8(3):36–41 ric determination of phosphorus. J Biol Chem
60. Rule GS, Montagna RA, Durst RA (1996) 66(2):375–400
Rapid method for visual identification of spe- 73. Edwards KA, Curtis KL, Sailor J, Baeumner A
cific DNA sequences based on DNA-tagged (2008) Universal liposomes: preparation and
liposomes. Clin Chem 42(8):1206–1209 usage for the detection of mRNA. Anal Bioanal
61. Szoka F, Olson F, Heath T, Vail W, Mayhew E, Chem 391(5):1689–1702
Papahadjopoulos D (1980) Preparation of uni- 74. Edwards KA, Baeumner AJ (2009) Liposome-
lamellar liposomes of intermediate size enhanced lateral-flow assays for the sandwich-
(0.1–0.2 mumol) by a combination of reverse hybridization detection of RNA. Methods Mol
phase evaporation and extrusion through poly- Biol 504:185–215
carbonate membranes. Biochim Biophys Acta
601(3):559–571 75. Bartlett GR (1959) Phosphorus assay in col-
umn chromatography. J Biol Chem 234
62. Szoka F Jr, Papahadjopoulos D (1978) Proce- (3):466–468
dure for preparation of liposomes with large
internal aqueous space and high capture by 76. Fenzl C, Hirsch T, Baeumner AJ Liposomes
reverse-phase evaporation. Proc Natl Acad Sci with high refractive index encapsulants as tun-
U S A 75(9):4194–4198 able signal amplification tools in surface Plas-
mon resonance spectroscopy. Anal Chem 87
63. Edwards KA, Baeumner AJ (2006) Sequential (21):11157–11163
injection analysis system for the sandwich
hybridization-based detection of nucleic acids. 77. Sallach SM, Nowak R, Hudson MP et al
Anal Chem 78(6):1958–1966 (2004) A change in serum myoglobin to detect
acute myocardial infarction in patients with
64. Rule GS, Montagna RA, Durst RA (1997) normal troponin I levels. Am J Cardiol 94
Characteristics of DNA-tagged liposomes (7):864–867
allowing their use in capillary-migration,
Chapter 26
Abstract
Lateral flow immunoassays have been widely used in recent years for detection of toxins, heavy metals, and
biomarkers. To improve the efficiency of individual lateral flow immunoassays, multiplex analytical strips
play an important role in the detection of several important analytes. In this chapter, development of a dual
lateral flow immunoassay is presented for detection of a variety of low molecular weight molecules. Various
buffers, additives, and materials are introduced and evaluated. Depending on the analyte to be tested, the
technique allows for selection of optimum buffers, additives, and other materials.
Key words Lateral flow, Gold nanoparticles, Fumonisin B1, Deoxynivalenol, Aflatoxin B1
1 Introduction
With the advantages of speed, low cost, and ease of use, lateral flow
immunoassays can be developed to detect toxins, biomarkers,
drugs, etc., and are especially useful for point-of-care applications
[1]. Like other immunoassays, the principle of lateral flow immu-
noassays includes antigen–antibody and antibody–antibody (such
as mouse antibody and goat anti-mouse antibody) binding. More-
over, capillary action is employed to induce fluid flow through the
device without need of further mechanical or human intervention.
Signal detection is provided via gold nanoparticles or quantum dot
fluorescence. For most lateral flow immunoassays minimal training
is necessary and results can be observed in 20 min. These assays will
help address future needs in area as diverse as environmental moni-
toring, biomarker assays, and identification of toxins and drugs.
However, qualitative lateral flow immunoassays still lack ade-
quate sensitivity for many applications. Current detection limits
range from 1–10 ng/mL. Moreover, this immunoassay provides
only “yes or no” qualitative results. The future challenge of
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0_26,
© Springer Science+Business Media LLC 2017
435
436 Yuan-Kai Wang et al.
Table 1
Comparison between competitive and sandwich lateral flow immunoassay
Fig. 2 Results judgment and reactions in test line of indirect competitive and sandwich lateral flow
immunoassay
bind with secondary antibody in the control line, and red color will
be observed on the control line (see Fig. 2).
The sandwich lateral flow immunoassays are mainly used to
detect large molecule analytes, such as proteins, bacteria, and bio-
markers. For each analyte, the monoclonal antibodies 1# and 2# are
the antibodies against the different epitopes of the analyte. Nor-
mally, the monoclonal antibody conjugated with colloidal gold
nanoparticles named 1#, and the monoclonal antibody is immobi-
lized in a line on the NC membrane as the test line named 2#. The
anaylte in the extract of samples will be reacted with colloidal
gold–monoclonal antibody 1# conjugate firstly, and continuously
reacted with the monoclonal antibody 2# on the test line. Thus a
clear red line can be observed, which indicate the positive result (see
Fig. 2).
To our knowledge, the uses of inappropriate buffers for lateral
flow immunoassay will induce instability of the gold nanoparticle-
labeled antibodies and affect sensitivity (see Note 1). The intensities
of the test lines, stability and release rate of the mixtures (gold
nanoparticle-labeled antibodies and sample extraction), and detec-
tion sensitivity are markedly affected by the types of buffer and
concentrations of their components. Moreover, in different lateral
flow immunoassays (different analytes, antibodies, or materials),
the optimum buffers may be different. To optimize the immunoas-
say, different buffers, additives, surfactants (see Note 2) would be
evaluated. The optimum buffers and concentrations of components
are needed to determine for lower detection limits, balance of
Development of Dual Quantitative Lateral Flow Immunoassay. . . 439
intensities of the test and control lines, and stability of the gold
nanoparticle-labeled antibodies. The buffers in lateral flow immu-
noassay are mainly PBS and borate buffer (from 2 mM to 100 mM),
and the pH values are ranges from 6.5 to 8.2. The surfactant is also
an essential component for the conjugation/sample pad pretreat-
ing buffer, and it will affect the color of lines and the sensitivity.
Different types of surfactants (Tween 20, Triton X-100, Tetronic
1307, etc.) can be evaluated, and selected in the certain lateral flow
immunoassay.
2 Materials
3 Methods
3.1 Preparation The flow chart of methods in this study is shown on Fig. 3. Firstly,
of Colloidal Gold 100 mL tetrachloroauric acid solution (0.01%, w/v) is stirred and
Nanoparticles (25 nm) heated until boiling (see Note 3) in conical flask. A volume of
0.7 mL 1% sodium citrate (w/v) is then added to the solution
quickly [6] (see Note 4). In the next 2 min, the color of mixture
is changed from transparent to dark, then clear wine red. Decreas-
ing the power to gentle heating for 5 min, then the mixture is
cooled down to room temperature and stored at 4 C. The diame-
ter of nanoparticles is determined by the wavelength scanning and
transmission electron microscope (see Fig. 4). The maximum
absorbing wavelength is 525 nm for the 25 nm colloidal gold
nanoparticles, based on the equation relating the gold nanoparticle
size X (nm) and the maximum absorption wavelength Y (nm):
Y ¼ 0.4271X + 514.56 [7]. In transmission electron microscopy
the mean diameter of gold nanoparticles is 24.8 3.2 nm as
measured by averaging a random selection of 100 particles.
Concentration of antibodies
Preparation of colloidal gold-labeled monoclonal antibodies
Optimization pH of colloidal gold solution
Sample preparation
Fig. 3 Flow chart of quantitative lateral flow immunoassay in this study. The optimization steps are needed for
each lateral flow immunoassay
Development of Dual Quantitative Lateral Flow Immunoassay. . . 441
a b
0.5
0.4
Absorbance
0.3
0.2
0.1
0
400 450 500 550 600
100 nm
Wavelength (nm)
Fig. 4 Measurement of 25 nm colloidal gold nanoparticles by wavelength scanning (a) and transmission
electron microscope (b)
Fig. 5 Different concentrations of coating ZEN-BSA and FB1-OVA conjugates and colloidal gold labeled
antibody in lateral flow immunoassay for ZEN (a) and FB1 (b)
The fluid will pass through the sample pad, conjugation pad, NC
membrane, and absorption pad under capillary action. Clear red
lines (test lines) can be observed if the concentration of coated
antigen and colloidal gold-labeled monoclonal antibody are
enough, and selected as the optimum coating concentration.
Based on the optimization of individual test strip, dual lateral
flow strip is developed subsequently. ZEN-BSA (0.05 mg/mL) and
FB1-OVA (0.2 mg/mL) are dispensed onto the NC membrane as
two test lines (see Fig. 1). The goat anti-mouse antibody (0.07 mg/
mL) is also dispensed onto the NC membrane (1.0 μL/cm) to form
the control line, positioned at 0.5 cm above the Test 2 line. Mix-
tures of the two colloidal gold-labeled antibodies (mAb-ZEN and
mAb-FB1) are prepared at different ratios (1:9, 1.5:8.5, 2:8,
2.5:8.5, and 3:7, v/v) to acquire similar product intensities in the
Test 2 and Test 1 lines. A low concentration of colloidal gold-
labeled antibodies results in higher sensitivity, but narrows the
detection range. Thus, the use of an optimum dilution ratio
could balance the sensitivity and detection range in the lateral
flow immunoassay. The optimum ratio of the gold nanoparticles-
mAb-ZEN and gold nanoparticles-mAb-FB1 is 1.5:8.5 (v/v). Fur-
thermore, different dilution ratios (1:1.5, 1:2, 1:2.5, 1:3, 1:3.5,
and 1:4, v/v) of these mixtures are assessed using the 20 mM
borate buffer (pH 8.2) containing 6% trehalose (w/v), 1% BSA
(w/v), and 0.05% sodium azide (w/v). The optimum dilution ratio
of the colloidal gold-labeled antibodies mixture is 1:2.5 (v/v) in
the colloidal gold–monoclonal antibody dilution buffer. Then
10 μL mixtue is added on the pretreated conjugation pad and
444 Yuan-Kai Wang et al.
3.4 Test Procedure Twofold dilution series of ZEN (from 25 ng/mL) or FB1 (from
500 ng/mL) are prepared in 50 mM PBST. 250 μL standard
solutions are tested to determine the detection limit and ranges of
the strip. During reaction in the test strip, ZEN or FB1 in the
mixture would compete for binding with the specific antibodies
gold nanoparticles–monoclonal antibody for ZEN or gold nano-
particles–monoclonal antibody for FB1 against the coating antigen
conjugates, thus altering the reaction intensity on the test lines
(Test 1 or Test 2 lines, respectively, see Fig. 6).
The intensity of the test line and control lines are captured with
the strip reader after 20 min of reaction. Concentrations of ZEN or
FB1 in the samples are quantified from the dose–response curves
(band intensity vs concentrations of ZEN or FB1 in the standard
solutions) which are run simultaneously in triplicate. The strip
reader results from different concentrations of ZEN and FB1 are
used to determine the calibration curves of ZEN and FB1 (see
Fig. 7, by Origin 6.0, from OriginLab, Northampton, MA,
USA). Relative standard deviations for different samples are deter-
mined and normally need to be lower than 15%. After extraction
Fig. 6 Lateral flow dual immunoassay strips for fumonisin B1 (top) and zearalenone (bottom). The concentra-
tions of FB1 from 1 to 9 (top): 500, 250, 125, 62.5, 31.3, 15.6, 7.81, 3.91 and 0 ng/mL, and those of ZEN from
1 to 9 (bottom): 25, 12.5, 6.25, 3.13, 1.56, 0.781, 0.391, 0.195 and 0 ng/mL
Development of Dual Quantitative Lateral Flow Immunoassay. . . 445
from the spiked and natural samples, the dilution ratios (1:2.5, 1:5,
1:10, 1:15, and 1:20, v/v) of the sample extracts are also optimized
in order to decrease the methanol content in the extraction buffer
and the influence of proteins or other components in the sample
matrixes. The spiked and natural samples are determined for ZEN
or FB1 by quantitative lateral flow immunoassay and LC-MS/MS.
The recovery rates of the spiked samples are compared and the
446 Yuan-Kai Wang et al.
4 Notes
References
1. Quesada-González D, Merkoçi A (2015) paper microfluidics. Proc Natl Acad Sci U S A
Nanoparticle-based lateral flow biosensors. Bio- 111(10):3671–3676
sens Bioelectron 73:47–63 5. Wang YK, Wang J, Wang YC et al (2011) Prepa-
2. Feng S, Caire R, Cortazar B et al (2014) Immu- ration of anti-zearalenone monoclonal antibo-
nochromatographic diagnostic test analysis dies and development of an indirect
using Google Glass. ACS Nano 8(3): competitive ELISA for zearalenone. Microbiol-
3069–3079 ogy China 38(12):1793–1800
3. Kolosova AY, Sibanda L, Dumoulin F et al 6. Frens G (1973) Preparation of gold dispersions
(2008) Lateral-flow colloidal gold-based immu- of varying particle size: controlled nucleation for
noassay for the rapid detection of deoxynivalenol the regulation of the particle size in monodis-
with two indicator ranges. Anal Chim Acta 616 perse gold suspensions. Nat Phys Sci 241:20–22
(2):235–244 7. Chen X, Liu S (2004) Colloidal gold labeling
4. Warren AD, Kwong GA, Wood DK et al (2014) immunoassay and its application in rapid detec-
Point-of-care diagnostics for noncommunicable tion of small molecule. Pharm Biotechnol
diseases using synthetic urinary biomarkers and 11:278–280
INDEX
Avraham Rasooly and Ben Prickril (eds.), Biosensors and Biodetection: Methods and Protocols Volume 1:
Optical-Based Detectors, Methods in Molecular Biology, vol. 1571, DOI 10.1007/978-1-4939-6848-0,
© Springer Science+Business Media LLC 2017
449
450 B IOSENSORS
Index
AND BIODETECTION
Charged-coupled devices (CCDs) (cont.) thermal bonding of PMMA .......................... 292, 294
plate assay material .................................................. 239 Conversion process ....................................................... 335
screens, portable devices ......................................... 237 CorelDraw™ ................................................................. 332
Shiga toxin activity ........................................ 237–238, Cryptosporidium parvum .............................................. 408
240, 243, 245
system sensitivity, factors ............................... 245, 247 D
technical cost and scientific-grade.......................... 233 Deoxynivalenol.............................................................. 441
Chemoprophylaxis ........................................................ 408 Deoxyribozymes.......................................... 389, 390, 392
CMOSs. See Complementary
DeskJet multifunctional printer ................................... 335
metal–oxide–semiconductors (CMOSs) 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine
CO2 laser system ........................................................... 332 (DPPC) ........................................................ 416
Colocalization, 3D plasmonic nanogap arrays
Dipstick tests ................................................................. 401
biomolecular interaction........................................... 16 DMR. See Dynamic mass redistribution (DMR)
circular and triangular patterns ................................ 23 DNA hybridization ....................................................... 395
colocalized biomolecular interaction ....................... 17
DNAzymes ................................................. 358, 360, 362,
DNA hybridization 368, 389, 390, 392
molecular binding capacity .................... 25, 26, 28 3D spheroid cell culture ...................................... 148–151
preparation .................................................... 23–24
2D-SPRi. See Two-dimensional surface plasmon
sensitivity enhancement ......................... 24, 25, 27 resonance imaging (2D-SPRi)
DNA molecules ......................................................... 22 Dulbecco’s Modified Eagle Medium
2D nanogap production ........................................... 21 (DMEM).................................... 35, 37, 38, 44
DNA preparation ...................................................... 19
Dynamic mass redistribution (DMR) .........................145,
electromagnetic field distribution ............................ 22 147, 150, 152–154, 157, 158
evaporation ..........................................................16, 28
fluorescence-based sensing ....................................... 15 E
nanogap fabrication ..................................... 17, 19, 20
nanogap reducuion ................................................... 22 ECM. See Extracellular matrix (ECM)
near-field intensity distribution ................................ 22 EGFR. See Epithelial growth factor receptor (EGFR)
optical set-up ..........................................17–18, 20, 21 Electroluminescence (EL) illumination .............. 236, 237
surface plasmon (SP) ................................................ 15 Electrospun nanofibers ................................................. 410
Colorectal cancer cell .................................................... 145 Enz-SubAuNP...................................................... 396, 401
Colorimetric biosensors ................................................ 329 Enzymatic tests.............................................................. 338
Colorimetric detection system ............................ 330, 335 Enzyme-linked immunosorbent assays
Colorimetric PCR ................................................ 360, 362 (ELISAs) ............................................. 408, 411
Colorimetric sensors ................................... 393, 394, 402 Enzyme–substrate complex .......................................... 396
Complementary metal–oxide–semiconductors Epithelial growth factor receptor
(CMOSs)................... 233–236, 245, 269, 281 (EGFR) ............................................. 64, 67, 68
Contamination detection device Evanescent field biosensor ................................... 163, 174
biopharmaceuticals.................................................. 288 Extracellular matrix
bioreactors ............................................................... 288 (ECM).......................143, 144, 146, 150, 156
cell viability assays ................................................... 288
F
colony-forming units (CFU)......................... 296–298
control and visualization......................................... 298 Fixed-angle surface plasmon resonance ......................... 76
E. coli .............................................................. 290, 295 Fluorescent sensors ....................................................... 393
fluorescene intensity................................................ 297 Fluorometer.......................................................... 236, 242
growth phase, cells .................................................. 290 Free prostate-specific antigen
materials.......................................................... 290–291 (f-PSA) ................................................ 4, 12, 13
methods ................................................................... 288 Fumonisin B1 ....................................................... 439, 444
microfluidic cassettes...................................... 292, 298
optics and optoelectronics ...................................... 293 G
pathogens and biologics ......................................... 287
β-Galactosidase.............................................................. 372
photodiodes............................................................. 293
GFP. See Green fluorescent protein (GFP)
portable kinetics fluorometer ........................ 291–294
Giardia lamblia ............................................................ 408
resazurin .................................................289, 295–297
Glucose assay ................................................................. 334
BIOSENSORS AND BIODETECTION
Index 451
Glutamate assay ............................................................. 334 electromagnetic radiation ....................................... 130
GNRs. See Gold nanorods (GNRs) functionalization and immobilization.................... 137
Gold-coated tilted fiber Bragg gratings (TFBGs) growth conditions ................................................... 135
architectures .............................................................. 49 instruments.............................................................. 134
biochemical reactions................................................ 49 ITO-coated glass ..................................................... 137
bioreceptors ............................................................... 51 local electromagnetic field
Bragg wavelength...................................................... 53 simulation ........................................... 139, 140
configurations............................................................ 49 multiplexed bioprobes ............................................ 132
cytokeratins 7 (CK7) .......................................... 63–65 multiplexed biosensing, human and rabbit IgG
definition, biosensors ................................................ 47 samples ................................................ 137, 139
differential diagnosis ................................................. 51 nanoplasmonic biosensing
EGFR ......................................................................... 64 antigen detection ..................................... 135, 136
glass optical fiber ....................................................... 52 Beckman-Coulter UV-NIR
gold deposition, optical fiber surface .................51, 56 spectrophotometer ...................................... 136
high-resolution sensing............................................. 61 fabrication, nanoplasmonic biochip ................. 135
Kretschmann-Raether approach .........................48, 62 preparation ............................................... 134, 135
light coupling mechanism ........................................ 53 shapes ....................................................................... 130
optical fiber devices ................................................... 62 Sigma-Aldrich.......................................................... 137
optical system, production ....................................... 51 SPR band intensity and wavelength....................... 130
phase mask technique .........................................53, 56 UV–V spectra ................................................. 130, 131
photo-inscription ................................................53, 56 working solutions.................................................... 134
plasmonic generation ................................................ 50 Gp120............................................................................ 222
polyclonal antibodies ................................................ 67 G-quadruplex .............................................. 358, 360, 362
proteins and cells.......................................... 58–63, 67 Graphene nanoplatelets (GNPs) ......................... 344, 347
sensitivities ................................................................. 50 Graphic software ........................................................... 329
single-mode optical fiber .......................................... 52 Green fluorescent protein (GFP) ....................... 238, 240,
spectrum of................................................................ 51 242, 243, 246
SPR optical fiber sensors........................................... 50
SPR signature, EH and TM modes ...................58, 59 H
SRI sensitivity ......................................................60, 61 Hanks’ balanced salts (HBBS) .................................36–38
surface chemistry....................................................... 52 Hepatitis B virus (HBV)
surface functionalization.....................................56, 57
amplification ............................................................ 359
temperature changes ................................................. 55 colorimetric detection................... 359–361, 363–365
transducers................................................................. 49 colorimetric PCR .................................................... 360
transmembrane receptors
DNA extraction....................................................... 361
biosensing platform.......................................68, 69 DNAzyme................................................................ 358
development ........................................................ 69 G-quadruplexes ....................................................... 358
EGFR ...................................................... 64, 67, 68
nucleic acid testing .................................................. 358
optical fiber-based biochemical sensors ............. 68 One-Step PCR ........................................................ 362
surface functionalization..................................... 64 32
P labeled probe .................................. 359, 362, 363
transmission spectrum .............................................. 54
probe and primers .......................................... 361, 362
transmitted amplitude spectrum .......................50, 57, serologic immunity ................................................. 357
58, 60 serum calibration curve........................................... 361
wavelength tracking .................................................. 55 serum sample extraction ......................................... 359
Gold nanoparticles (AuNPs) ............................... 393–395
serum samples ......................................................... 367
lateral flow device and detection ................... 401, 402 TaqMan assay ................................358, 360, 365, 366
preparation ..................................................... 398, 400 Uracil N-glycosylase (UNG) .................................. 368
thiol-modified DNA ...................................... 399, 400
HIV genetic tests .......................................................... 222
Gold nanorods (GNRs) HIV infectivity assay ..................................................... 373
antibodies ....................................................... 132, 133 HIV long terminal repeat (LTR) ................................. 372
biofunctionalization ................................................ 133
Human growth hormone (hGH) ...................... 168, 169,
biosensing, imaging and drug delivery .................. 129 175–177, 181, 182
chemicals.................................................................. 133 Hydrocarbon ................................................................. 413
452 B IOSENSORS
Index
AND BIODETECTION
Point-of-Care (POC) R
CMOS...................................................................... 269
development ............................................................ 268 Reflectometric interference spectroscopy (RIfS)
fluidics ...................................................................... 268 advanced set-up .............................................. 210, 213
fluorescence ............................................................. 268 advantage ................................................................. 208
hydrodynamic focusing.................................. 268, 269 affinity and kinetic constants ......................... 217, 218
smartphone cameras................................................ 268 antibody concentration........................................... 217
wide-field flow cytometer .............................. 273, 274 antigen-antibody interaction ......................... 216, 217
Polyacrylamide gel electrophoresis (PAGE) ................ 395 BiaCore .................................................................... 207
Polyethylene glycol (PEG) ........................................... 415 biomolecular interaction......................................... 207
Polylactic acid (PLA) .................................................... 410 biopolymer ..................................................... 212, 215
Poly-L-lysine (PLL) ....................................................... 410 biosensing ................................................................ 219
Polymerase chain reaction (PCR) cleaning procedure.................................................. 218
convective format ..........................252, 254, 256, 262 detection principle .................................................. 208
electrode dissolution experiments.......................... 254 glass substrates ........................................................ 210
experiments ............................................................. 254 immobilization
gold-standard nucleic acid-based detection amino-terminated ligands................................. 215
assay.............................................................. 252 biotin.................................................................. 216
kinetics ..................................................................... 253 carboxy terminated ligands............................... 216
medical diagnostic tools ......................................... 251 thiol terminated ligands.................................... 216
microscale convective flow states .................. 252, 253 1-lambda RIDe ....................................................... 219
optical analysis ......................................................... 251 label-free optical biosensor ............................ 207, 220
smartphones. see Smartphones, PCR liquid handling ........................................................ 210
thermal cycling protocols ....................................... 252 optical thickness ...................................................... 208
voice communication .............................................. 252 parallel setup................................................... 210, 214
Polyvinyl alcohol (PVA)................................................ 415 phospahete buffered saline ..................................... 219
Polyvinylpyrrolidone (PVP) ......................................... 415 set-up .............................................................. 211–213
PPE. See Personal protective equipment (PPE) silanization............................................................... 212
Prostate-specific antigen (f-PSA).................................... 13 single setup .............................................................. 210
Protein biomarker biosensors...................................12, 13 software.................................................................... 211
Protein kinase C (PKC) .......................................... 35, 39, surface chemistry............................................ 209, 210
40, 42, 44, 45 temperature ............................................................. 208
PTEN. See Phosphatase and tensin homolog (PTEN) transducers...................................................... 209, 218
Resonance energy transfer (RET)
Q attributes of paper substrates.................................. 304
calibration curve, target DNA................................ 316
QCapture Pro capture software ................................... 377 data acquisition .............................................. 316–317
Quantitative data analysis ............................................. 230 data analysis
Quantitative multivariate imaging (QMI) ................... 228 digital images............................................ 319, 320
Quantum dots (QDs) .......................................... 222, 226 gQD/Cy3 RET Pair ................................ 317, 319
bioconjugation microscope images, UCNP/QD RET
DTPA-terminated oligonucleotides................. 313 Pair ...................................................... 320, 321
hexahistidine-terminated decentralized diagnostic assays ............................... 304
oligonucleotides ................................. 312, 313 instrumentation and equipment.................... 308, 309
gQD/Cy3....................................................... 304, 307 oligonucleotide sequences,
imidazole modified paper substrates ...................... 315 hybridization....................................... 308, 309
neutral density (ND) filter...................................... 324 paper-based solid-phase nucleic acid
photoluminescence ................................................. 324 hybridization.............................. 304, 306, 307
properties................................................................. 302 pseudo-coloring of images ..................................... 324
salt aging.................................................................. 322 QDs. see Quantum dots (QDs)
structural types ........................................................ 302 reagents........................................................... 306, 308
surface area .............................................................. 302 SMN1 sequence ............................................. 318, 322
water soluble................................................... 311–312 solid-phase assays .......................... 303–305, 318, 319
BIOSENSORS AND BIODETECTION
Index 455
UCNPs. see Upconverting nanoparticles (UCNPs) BupH PBS ............................................................... 353
wax printing, papers CRP analysis ............................................................ 344
aldehyde and imidazole functionality ............. 314, DIA .......................................................................... 345
315 human CRP IAs ............................ 347–349, 351, 352
aldehyde functionality ....................................... 314 iPAD4 ...................................................................... 346
substrates .................................................. 313, 314 iPAD mini ................................................................ 346
Resonant waveguide grating (RGW) iPhone 5s ................................................................. 346
3D cell models ........................................................ 144 IVD .......................................................................... 344
3D spheroid cell culture ................................ 148–151 MTP......................................................................... 352
adhesion.......................................................... 157, 158 pixel intensity (PI)................................................... 352
biosensor microplate ...................................... 144, 146 POC ......................................................................... 343
cell invasion ............................................................. 143 PPE .......................................................................... 354
cell migration........................................................... 143 Smartphones, PCR
chemotaxis ............................................................... 143 advantages................................................................ 252
data analysis software .............................................. 148 amplification ............................................................ 255
extracellular matrix (ECM) .................................... 143 DNA replication ...................................................... 263
intra-sensor referencing .......................................... 158 electrochemical dissolution ...................259, 262–264
label-free single cell monitoring.................... 144, 145 enzyme..................................................................... 265
Matrigel ................................................. 146, 147, 150 fabrication................................................................ 259
microplates and instruments .................................. 148 fluorescence analysis....................................... 258–261
multicellular spheroid ............................................. 144 instrument portability ............................................. 255
optimal seeding density, spheroids......................... 157 label-free detection......................................... 262, 263
single cell monitoring ............................................. 146 molding processes ................................................... 262
spheroids, cancer cells ............................................. 146 photography ............................................................ 265
tissue culture medium and cell line............... 145, 148 temperature settings....................................... 255–258
transwell invasion assay .................................. 156, 157 voice communication .............................................. 252
tumor metastasis ..................................................... 143 SPPs. See Surface plasmon polaritons (SPPs)
wavelength sweeping technique ............................. 146 SPR. See Surface plasmon resonance (SPR)
wound healing assay.............................. 152, 155, 156 Streak imaging flow cytometry
Respiratory syncytial virus (RSV)................................. 408 adhesive layer........................................................... 284
RGW. See Resonant waveguide grating (RGW) background signal and noise
Ribozymes ..................................................................... 389 reduction............................................. 277–280
cell culture and fluorescence staining .................... 271
S cell culture and labeling ................................. 272, 273
cell image ........................................................ 269–271
SA-AuNP. See Sialic acid gold nanoparticle (SA-AuNP)
Sandwich immunoassay ................................................ 351 CMOS imaging sensors .......................................... 269
“capture” antibody ................................................. 410 color channel extraction ................................ 278–281
components ............................................................. 271
“detection” antibodies............................................ 410
flow of fluid ............................................................. 409 computer control and data analysis........................ 272
membrane properties ..................................... 410, 414 configuration ........................................................... 277
development ............................................................ 268
membrane types ...................................................... 410
quantification........................................................... 411 description ............................................................... 267
Sialic acid gold nanoparticle (SA-AuNP) flow cell fluid delivery and imaging ....................... 271
absorption spectra ................................................... 114 focal ratio ................................................................. 283
optimization ................................................... 282, 283
concentration ................................................. 112, 113
description ............................................................... 112 POC. see Point-of-care (POC)
hemagglutinin ......................................................... 114 rare cell counting ........................................... 281, 282
webcam performance ............................ 275, 277, 284
linear correlation ..................................................... 115
particle size .............................................................. 113 wide-field flow cell fabrication....................... 274–276
Signal amplification ....................................................... 120 Streptavidin.................................................................... 413
Sulforhodamine B (SRB) dye ....................................... 413
Signal transduction ........................................................... 5
Silica nanoparticles (SiO2NP) ............................. 333, 338 Surface biofunctionalization, BiMW
Smartphone-based colorimetric reader (SBCR) (3-aminopropyl)triethoxysilane
Ab-bound MTP .................................... 346, 350, 351 (APTES)..................................... 174, 177, 178
456 B IOSENSORS
Index
AND BIODETECTION