Development of on-chip proximity ligation assay with in situ single

34
Development of on-chip proximity ligation assay with in situ single molecule sequencing readout Xiao He Degree project in applied biotechnology, Master of Science (2 years), 2011 Examensarbete i tillämpad bioteknik 45 hp till masterexamen, 2011 Biology Education Centre and Department of Immunology, Genetics and Pathology, Uppsala University Supervisors: Rachel Yuan Nong and Masood Kamali-Moghaddam

Transcript of Development of on-chip proximity ligation assay with in situ single

Page 1: Development of on-chip proximity ligation assay with in situ single

Development of on-chip proximity ligationassay with in situ single moleculesequencing readout

Xiao He

Degree project in applied biotechnology, Master of Science (2 years), 2011Examensarbete i tillämpad bioteknik 45 hp till masterexamen, 2011Biology Education Centre and Department of Immunology, Genetics and Pathology, UppsalaUniversitySupervisors: Rachel Yuan Nong and Masood Kamali-Moghaddam

Page 2: Development of on-chip proximity ligation assay with in situ single

1  

Contents Summary ........................................................................................................................................... 2 1.  Introduction ............................................................................................................................... 3 

1.1 Different protein detection methods .................................................................................... 3 1.2 Proximity Ligation Assay .................................................................................................... 4 1.3 Readout formats of PLA ..................................................................................................... 6 1.4 Aim ...................................................................................................................................... 8 1.5 Method description ............................................................................................................. 9 

2. Materials and methods ................................................................................................................ 12 2.1 Proteins, oligonucleotides and conjugates ........................................................................ 12 2.2 Antibody immobilization on microparticles ...................................................................... 12 2.3 Antibody immobilization on glass slides .......................................................................... 12 2.4 PLA on microparticles ...................................................................................................... 13 2.5 PLA on glass slides ........................................................................................................... 14 

2.5.1 Readout by hybridization of detection oligo .......................................................... 14 2.5.2 Readout by sequencing .......................................................................................... 15 

2.6 Data Analysis .................................................................................................................... 15 3. Results ......................................................................................................................................... 16 

3.1 Validation of oligo system ................................................................................................. 16 3.1.1 Benchmark new oligo system to existing oligo systems ........................................ 16 3.1.2 Formation of circular DNA .................................................................................... 18 

3.2 Immobilization of capture antibody via DNA ligation ...................................................... 19 3.2.1 Comparison of new immobilization method with existed one ............................... 19 3.2.2 Optimization of immobilization method ................................................................ 20 

3.3 Optimization of protein detection strategy ........................................................................ 22 3.4 Singleplex protein microarray ........................................................................................... 23 3.5 Duplex protein microarray ................................................................................................ 24 

4. Discussion ................................................................................................................................... 26 Acknowledgement .......................................................................................................................... 29 References ....................................................................................................................................... 30 Supplementary Table 1 .................................................................................................................... 33   

Page 3: Development of on-chip proximity ligation assay with in situ single

2  

Summary

During the last few years, protein biomarkers have played a critical role in the early

diagnostic of diseases. This calls for the development of bioanalytical technologies

with high sensitivity, specificity and precision. Proximity ligation assay (PLA) is a

recently developed technology for protein analysis. In PLA, two or more proximity

probes, normally oligonucleotide-conjugated antibodies are used to target the protein

of interest. The free ends of the attached oligonucleotides are joined by enzymatic

ligation if they are brought in proximity. Then the ligation products can be amplified

and detected. The multiple binding events and DNA amplification of PLA provide a

specific and sensitive assay for protein biomarkers at trace levels.

In this project, we aimed to develop a chip-based PLA with single molecule resolution

and sequencing readout. A new oligo system was constructed to enable multiplex

detection and three recognition events were recruited to enhance specificity. Rolling

circle amplification (RCA) was employed as localized amplification which allowed

the visualization and enumeration of individual molecules. Readout by

next-generation sequencing increased the precision of the assay.

Page 4: Development of on-chip proximity ligation assay with in situ single

3  

1. Introduction

Protein biomarkers are becoming increasingly important due to their potentials in

early disease diagnostics, personalized treatments and therapeutic monitoring.

Proteins related to a variety of cancers [1-3], heart [4], renal [5] and Alzheimer’s [6]

have been reported. This has motivated researchers to further develop and apply new

technologies for biomarker discovery and quantitative detection. Most biomarkers

reside in biological fluids such as serum, plasma or urine and they exhibit a large

difference in abundance-over 12 orders of magnitude [7]. Therefore, protein

detection offering high sensitivity, specificity and precision over a broad dynamic

range is required.

1.1 Different protein detection methods

Mass spectrometry (MS) is one of the most commonly used techniques for profiling

protein biomarkers. The principle of MS for protein identification is through

measuring the mass-to-charge ratio of charged particles. Proteins are digested with

proteases, normally trypsin, into peptides prior to MS analysis. Then the peptides are

introduced to a mass analyzer and identified. The merit of MS is its high throughput

property which permits the analysis of thousands of proteins per run [8]. However, the

instrumental dynamic range of 104 to 105 limits the application on profiling proteins

with wide dynamic range [9] and the variations from sample collection and

preparation hinder the reproducibility [10].

Immunoassays like enzyme-linked immunosorbent assay (ELISA) was introduced in

1971. [11]. In ELISA, the proteins are immobilized to a surface and specific

antibodies are applied to the surface to bind the proteins. An enzyme linked to the

antibody is converted to some detectable signals after reacting with substrates. ELISA

and its variants such as sandwich ELISA have been widely employed in the detection

of biomarkers [12]. Nevertheless, the sensitivity of labels and detection equipment

Page 5: Development of on-chip proximity ligation assay with in situ single

4  

restricts the performance of ELISA. Also, the unspecific bindings of probe to wrong

irrelevant targets give rise to background which results in the loss of sensitivity [13]

Protein microarray provides a multiplex approach to analyze proteins. Normally,

antibodies are affixed at separate locations in order on a glass slide. After proteins are

captured, detector antibodies labeled with fluorescences are added and detected upon

their binding to targets. Although protein microarrays open a way to measure different

proteins simultaneous in biological samples, it’s difficult to establish specific and

sensitive assays because of the technical challenges. The diverse properties of proteins,

the interferences from other reagents and the cross-reactivities of antibodies all

contribute to the challenges [14].

1.2 Proximity Ligation Assay

The proximity ligation assay (PLA) was first described by Fredriksson et al. in 2002

as a highly sensitive method for detecting cytokine platelet-derived growth factor

(PDGF). The method depends on a simultaneous binding of two affinity probes to the

target protein. The affinity probes used were DNA aptamers which can be extended at

either the 5’ or 3’end by additional sequence elements. When the two probes bind the

same target, the 5’ and 3’ free ends are brought to close proximity, then a connector

oligonucleotide which has complementary sequence to both probes is added and it

templates the enzymatic ligation of the two free ends. After that the ligation products

can be amplified and detected by quantitative real time PCR (q-PCR). The first

version of PLA was carried out in solution and there were no washing or separation

steps. It showed a 1000 fold higher sensitivity in detecting PDGF B-chain comparing

to sandwich ELISA [15].

The introduction of antibodies to prepare affinity probes increased the utility of PLA.

Polyclonal antibodies and monoclonal antibodies were used to conjugate to

oligonucleotides. The conjugation was through either covalent coupling or

Page 6: Development of on-chip proximity ligation assay with in situ single

5  

biotin-streptavidin interactions. Protein detection with high sensitivity and wide

dynamic range was achieved by this antibody-based PLA [16].

Solid phase PLA (SP-PLA) combined the advantages of PLA and sandwich

immunoassays. Capture antibodies are first immobilized on the solid supports. After

target proteins are captured, pairs of proximity probes are added and amplifiable

reporter DNA molecules are generated by enzymatic ligation. Different solid supports

have been used such as the surface of modified tubes and microparticles. SP-PLA

increases the specificity of the assay by requiring three recognition events, enabling

more precisely multiplex protein analysis. In addition, the microparticle-based PLA

permits the concentration of target molecules by magnetic forces and depletion of

excess probes as well as interfering agents by washes. Therefore, SP-PLA is suitable

for detecting minute amount of proteins in complex biological samples such as plasma

or serum or whole blood. According to Darmanis et al. SP-PLA showed a similar limit

of detection (LOD) comparing to solution-phase PLA whereas presenting a broader

dynamic range, and the protein detection based on microparticles was superior in a

multiplex setting [17].

Multiplexed solid-phase PLA (MultiPLAy) is a newly developed approach for parallel

detection of proteins. The recruit of microparticle-based PLA ensured the specificity

and sensitivity of the assay while the use of next-generation sequencing readout

improved precision and throughput. The ability of simultaneous detection of 35

proteins with low cross-reactivity and improved sensitivity comparing to traditional

sandwich ELISA has been demonstrated [18].

PLA has also been developed to monitor proteins and protein-protein interactions in

individual cells and tissues at single molecule resolution which is known as in situ

PLA. In in situ PLA, cells and tissues are fixed to a slide and then two

oligonucleotide-conjugated proximity probes are added. When the two proximity

probes are brought close to each other by binding to the same protein or interacting

Page 7: Development of on-chip proximity ligation assay with in situ single

6  

proteins, they can guide the hybridization and enzymatic ligation of two subsequently

added connector oligonucleotides. After ligation, a circular DNA molecule is

generated that can serve as an endless template for rolling circle amplification (RCA).

RCA is an amplification method carried out by DNA polymerase such as phi29 by

employing circularized DNA as template. An hour RCA reaction can generate a

single-stranded DNA constituted of about 1000 complementary copies of the DNA

circle in length of around 100 nt. In in situ PLA, RCA is primed from one of the

oligonucleotides conjugated to proximity probes, resulting in a randomly coiled

single-stranded DNA molecule linking to the protein-probe complex. Such RCA

product (RCP) remain localized at the same place where the protein stays, enabling

the study of proteins or protein-protein interactions in cells and tissues. RCPs can be

detected by hybridization of complementary fluorescence-labeled oligonucleotides

since the product has hundreds of repeated sequences. After hybridization, RCP

appears as a sub-micrometer sized bright spot which can be easily visualized through

a fluorescence microscope [19, 20].

The strength of PLA is that it converts the detection of proteins and their molecular

events such as interactions into an easier way of detecting nucleic acids. The use of

DNA molecules as surrogate markers allows the amplification of signal which gives

rise to the high sensitivity of the assay. Furthermore, PLA recruits multiple

recognition events to create one amplifiable DNA reporter. In this way, the

background from unspecific adsorption is dramatically reduced, in the meantime, the

specificity of the assay is increased.

1.3 Readout formats of PLA

The readout format plays an important role in the immunoassay in a way that the

precision and sensitivity of it will influence the performance of the assay. Various

readout formats are used for PLA such as q-PCR [15-17], sequencing [18] and

microscopy [19].

Page 8: Development of on-chip proximity ligation assay with in situ single

7  

Quantitative polymerase chain reaction (q-PCR), also called real time polymerase

chain reaction (RT-PCR) is the most commonly used readout for solution-phase and

solid-phase PLA. The procedure of q-PCR is similar to PCR which is usually

employed to amplify target DNA molecules. The real-time monitoring feature of

q-PCR makes it possible to both detect and quantify specific sequences. At the end of

each amplification cycle, the amplified DNA products are detected and quantified

through measurement of fluorescence intensity. Two kinds of fluorescent labels for

DNA products are nonspecific fluorescent dyes which bind to any double-stranded

DNA (such as SYBR Green) and sequence-specific DNA probe containing a

fluorescent reporter which only hybridizes to its complementary target (such as

TaqMan probe).

The parameter used to indicate fluorescence intensity is cycle threshold (Ct). Ct is

defined as the number of cycles needed for the fluorescent signal to reach the

threshold. So the higher the Ct level is, the fewer the amount of starting DNA

molecules in the sample. When a new detection method is established, a standard

curve is made to determine the LOD, dynamic range and efficiency of the detection.

In PLA, the standard curve is made by plotting Ct values against a serial of protein

concentrations on a logarithmic scale. The Y axis is usually inverted since the Ct

values are inversely proportional to the amount of target proteins.

Fluorescence microscopy has been applied as the readout for in situ PLA. In situ PLA

employs RCA as amplification and the RCPs produced can be visualized under

microscope by hybridization of fluorescent labels. Each protein or protein-protein

interaction represented by one RCP can be enumerated, allowing quantification of

proteins or protein-protein interactions in cells and tissues. BlobFinder developed by

Uppsala University is designed in particular for enumerating RCPs [21]. A later

version of BlobFinder can identify RCPs based on their 3D subcellular locations [22].

Next-generation sequencing has greatly transformed today’s biological research. [23].

Page 9: Development of on-chip proximity ligation assay with in situ single

8  

It also provides a new readout format for PLA taking advantage of its high-throughput

property. The ability of massively parallel sequencing permits the investigation of

large number of sequences in one reaction which is suitable for multiplex PLA.

To date, three commercially available platforms for next-generation sequencing are

454 (Roche), Genome Analyzer (Illumina) and ABI-SOLiD (Applied Biosystems)

[24]. The sequencing principle employed by Illumina is sequencing by synthesis

(SBS). Sequencing templates are immobilized on a flow cell surface and undergone

bridge amplification, resulting in millions of double-stranded DNA clusters on the

surface. After that, a single fluorescently labeled deoxynucleoside triphosphate (dNTP)

is incorporated to each nucleic acid chain and an image is taken for identification. The

four nucleotides are distinguished from each other by their different fluorescent labels.

After the first cycle of sequencing, the fluorescent dye is cleaved and the next

incorporation could take place. [25].

1.4 Aim

The aim of this project was to establish a general method for detecting multiplex

proteins with single molecule resolution. Different proteins could be encoded by

proximity ligation and decoded by single molecule sequencing. Single molecule

detection is the ultimate level of quantification and it enables digitally recording of

individual molecules with high precision. To achieve that, we designed an oligo

system based on the in situ PLA system described by Soderberg et al., but with a

novel hairpin structure that decreased oligo complexity when multiplexed. Flow cell

surface was used as the solid support where protein capture and detection events were

taking place in situ. Three recognition events were recruited to assure specificity.

RCA was employed as localized amplification method to permit single molecule

resolution, and the Illumina sequencing platform was used as readout.

Page 10: Development of on-chip proximity ligation assay with in situ single

9  

1.5 Method description

In this method, short oligonucleotides are first immobilized on a chip (Figure 1a).

Next, capture conjugates are hybridized to those short oligonucleotides (Figure 1b)

and linked to the short oligonucleotides by enzymatic ligation (Figure 1c). Target

protein and a pair of proximity probes conjugated with

hairpin-structure-oligonucleotides are added in sequence (Figure 1d). After the

formation of protein-probe complex (Figure 1e), a nicking enzyme which recognizes

the specific sequence in the hairpin structure is applied. The cleaved oligonucleotides,

if in proximity, can be guided to form a circular DNA by adding two splints which can

be digested by UNG later. The circularized DNA molecule serves as the endless

template for RCA. The oligonucleotide linking to the solid support can pop up after

cleaving and serve as the RCA primer. The oligonucleotides of the other two

proximity probes have 5’ ends, preventing them from serving as primers (Figure 1f).

The circle generated can hybridize to the primer on the support even when the

complex dissociates (Figure 1g), so the RCP stays on solid support (Figure 1h).

Illumina sequencing platform is employed for detecting RCPs. The approximate 1000

times amplification after one hour RCA satisfies the requirement of Illumina imaging.

Page 11: Development of on-chip proximity ligation assay with in situ single

10  

Figure 1. Schematic description of on-chip PLA procedure. (a) The oligonucleotides are immobilized

on the solid support by biotin-streptavidin interactions. (b) Capture conjugates are added and

hybridized to the oligonucleotides. (c) After enzymatic ligation, the capture conjugates are linked to the

solid support. (d) The target protein and two proximity probes with hairpin structure oligonucleotides

are added in turn. Unique barcodes for different proteins are designed in the red part of the probe to

enable multiplex detection. The blue and green parts for the other proteins are the same which allows

the use of universal splints. (e) A protein-probe complex is formed on the solid support. (f) Nicking

enzyme nicks the specific sequence between stem and loop in the probes while the cleaved

oligonucleotides remain hybridize to the probes. Those oligonucleotides are guided to form a circle by

addition of two splint oligonucleotides. The oligonucleotide on the solid support can pop up after

cleavage and serve as the primer for RCA later. (g) Enzymatic ligation happens at the junctions of two

connector oligonucleotides. The circular oligonucleotide formed hybridizes to the primer. (h) The

immobilized oligonucleotide primes on-chip RCA and the RCP can be detected.

Comparing to the oligo system used in SP-PLA, the amplified products by PCR

would diffuse away which doesn’t allow a localized detection. However, in this oligo

system, the generated RCA template can hybridize to the primer on the solid support,

Page 12: Development of on-chip proximity ligation assay with in situ single

11  

so the RCP will be retained at the site where the proteins are bound. Another

advantage of employing the oligonucleotide on the solid support as primer is that the

RCP will always be linked to the solid support no matter the protein-probe complex

dissociates or not. The signal in this system is generated only when three binding

events, two for circularization of RCA template and one for priming, taking place on

the same protein, therefore the detection efficiency is assured.

Comparing to the oligo system used in in situ PLA, this new oligo system simplifies

the procedure of multiplex detection. In a multiplex detection, different proteins are

encoded by a short DNA sequence in the proximity probe which can be decoded by

sequencing later. In the oligo system of in situ PLA, different proteins have their own

proximity probes and the number of different connector oligonucleotides added is

dependent on the number of proteins detected. In this new system, different protein

barcodes are included in the stem part of the hairpin structure so that different

proximity probes are carrying their own connector oligonucleotides. The other

sequence except the barcode is the same for all the proteins. In this way, only

universal splints are needed which simplifies the whole process.

Page 13: Development of on-chip proximity ligation assay with in situ single

12  

2. Materials and methods

2.1 Proteins, oligonucleotides and conjugates

Antigens, affinity-purified polyclonal antibodies, and their biotinylated forms for

VEGF and Mouse IgG were purchased from R&D Systems. The oligonucleotides

used in the experiments are summarized in Supplementary Table 1.

Antibody-oligonucleotide conjugates were prepared as described by Soderberg et al.

[19]

2.2 Antibody immobilization on microparticles

To immobilize antibodies on Dynabeads Myone Streptavidin T1 (Invitrogen) via

DNA ligation, 50 μl (0.5 mg) beads were washed twice with 500 μl washing buffer

(0.05% Tween-20 (Sigma-Aldrich), 1 × PBS). Biotinylated oligonucleotides (Biomers)

of 100 μl with corresponding concentrations (500 nM, 100 nM, 20 nM, 8 nM) were

added and incubated for 1 h at RT under rotation. After that, the beads were washed

twice with 500 μl washing buffer and 100 μl capture conjugates with corresponding

concentrations (250 nM, 50 nM, 10 nM, 2 nM) were added and incubated for 1 h at

RT under rotation. The beads were washed twice with 500 μl washing buffer prior to

the addition of 100 μl ligation mix (1×PCR buffer (Invitrogen), 2.5 mM MgCl2

(Invitrogen), 1 unit of T4 DNA ligase (Fermentas), 0.08 mM ATP (Fermentas)) and

incubated at 37 oC for 10 min. Next, the beads were washed with 500 μl washing

buffer and resuspended with 100 μl storage buffer (0.1% purified BSA (New England

Biolabs) and 1 × PBS). The microparticles immobilized with antibodies can be stored

at 4 oC for at least two months. The immobilization of biotinylated antibodies to

microparticles was performed as described by Darmanis et al. [18]

2.3 Antibody immobilization on glass slides

Biotinylated oligonucleotides (Biomers) were printed on TRIDIA™ BA streptavidin

Page 14: Development of on-chip proximity ligation assay with in situ single

13  

coated slide (SurModics). Each slide was printed with 14 sub-arrays of 20 × 20

features. The array printing and blocking protocols were described by Ericsson et al.

Secure-Seal 16 (Grace Bio-labs) was attached to the slide prior to use. Forty μl of 50

nM capture conjugates were added to each reaction chamber and incubated over night

at 4oC. Next, each well was gently flushed with 1ml 1 × PBS and dried. 40 μl ligation

mix (as described previously) was added and incubated for 10 min at 37oC followed

by washing and drying. After that the glass slide was stored at 4oC and ready for PLA.

2.4 PLA on microparticles

For every PLA reaction, 1 μl (5 μg) microparticles with immobilized antibodies were

used. The storage buffer was removed and the microparticles were resuspended with 5

μl PLA buffer. Samples were prepared by diluting antigens in serial with PLA buffer

(1 mM D-biotin (Invitrogen), 0.1% purified BSA (Bovine Serum Albumin, New

England Biolabs), 0.05% Tween-20 (Sigma-Aldrich), 100 nM goat IgG

(Sigma-Aldrich), 0.1 μg/μl salmon sperm DNA (Invitrogen), 5 mM EDTA and 1 ×

PBS). In all dilution series, a negative control with only PLA buffer was included to

determine background. Five μl microparticles were mixed with 45 μl sample in a well

of a 96-well PCR plate and incubated for 1.5 h at RT under rotation. Next, the 96-well

PCR plate was placed on a 96-well plate magnet (Invitrogen) and washed twice with

100 μl washing buffer. Fifty μl PLA probe mix containing 1 nM of each probe was

added and incubated for another 1.5 h at RT under rotation. Afterwards, the

microparticles were washed twice and 30 μl nicking mix (1 × NEB buffer (New

England Biolabs), 0.1 μg/μl purified BSA (New England Biolabs), 0.5 unit of Nt.BtsI

(New England Biolabs)) was added. The PCR plate was incubated at 37oC for 30min

and the microparticles were washed thereafter. Fifty μl ligation mix containing 1 ×

PCR buffer (Invitrogen), 2.5 mM MgCl2 (Invitrogen), 1 unit of T4 DNA ligase

(Fermentas), 100 nM of two splints and 0.08 mM ATP (Fermentas) was added and

incubated at 37oC for 10 min. After washing, an UNG mix of 30 μl containing 1×PCR

buffer (Invitrogen), 2.5 mM MgCl2 (Invitrogen), 1 unit of UNG (Fermentas) was

Page 15: Development of on-chip proximity ligation assay with in situ single

14  

added and incubated for 5 min at 37oC. Next, the microparticles were washed again,

30 μl RCA mix with 1 × phi29 buffer (Fermentas), 0.5 μg/μl purified BSA (New

England Biolabs), 0.2 mM dNTPs (Fermentas), 6 units of phi29 (Fermentas) was

added. Upon the addition of RCA mix, the whole plate was incubated at 37oC for

30min followed by washing. Finally, 50 μl PCR mix containing 1 × PCR buffer

(Invitrogen), 2.5 mM MgCl2 (Invitrogen), 100 nM of forward and reverse primers

(Integrated DNA Technology), 0.2 mM dNTPs (with dUTP, Fermentas), 0.5 × SYBR

Gold and 1.5 units of Taq polymerase (Fermentas) was added. The thermocycling

program was denaturation at 95oC for 10 min followed by 40 cycles of 15 s at 95oC

and 1 min at 60oC.

2.5 PLA on glass slides

Samples were prepared by diluting antigens to proper concentrations with PLA buffer.

Forty μl were added to each reaction chamber and incubated at 37oC for 1.5h. Each

well was flushed with 1ml 1 × PBS and dried. Next, 40 μl of PLA probe mix

containing 5 nM of each probe was added and incubated at 37oC for another 1.5 h.

After that the wells were washed and 40 μl of the nicking mix was added and

incubated for 30 min at 37oC. After washing, 40 μl ligation mix was added and

incubated at 37oC for 10 min. Each well was washed before the addition of 40 μl

UNG mix. After incubating at 37oC for 5 min, each well was washed and

reconstituted with 40 μl RCA mix. The nicking mix, ligation mix, UNG mix and RCA

mix were the same as used on microparticles.

2.5.1 Readout by hybridization of detection oligo

Upon 1 h RCA reaction at 37oC, the wells were washed and 40 μl of hybridization

mix containing 100 nM detection oligo and 1 × hybridization buffer (100 mM

Tris-HCl (pH 7.6), 300 mM NaCl, 500 nM EDTA and 0.05% Tween-20) was added.

The glass slide was incubated at 37oC for 30 min. Next, each well was washed and

dried. Mounting media was added and the glass slide was covered with coverslip and

Page 16: Development of on-chip proximity ligation assay with in situ single

15  

ready to observe under microscope.

2.5.2 Readout by sequencing

Upon 1h RCA reaction at 37oC, the wells were washed and 40 μl of hybridization mix

containing 100 nM sequence primer and 1 × hybridization buffer was added. The

glass slide was incubated at 37oC for 30 min. Afterwards each well was equilibrated

with 1 × incorporation buffer (Illumina). The wells were dried and 40 μl incorporation

mix (Illumina) was added and incubated at 55oC for 10min. Lastly, each well was

washed and dried again. Mounting media was added and the glass slide was covered

with coverslip and ready to observe under microscope.

2.6 Data Analysis

The q-PCR results were analyzed with MxPro software (Stratagene). Ct values were

exported and analyzed with Microsoft Excel. The microscopic images were analyzed

with BlobFinder and the blobs were enumerated.

Page 17: Development of on-chip proximity ligation assay with in situ single

16  

3. Results

3.1 Validation of oligo system

3.1.1 Benchmark new oligo system to existing oligo systems

The new oligo system, namely Nick system was compared with existing oligo

systems namely Biovic and S3 systems, for SP-PLA and in situ PLA, respectively. All

the assays were performed by detecting vascular endothelial growth factor (VEGF) on

microparticles with q-PCR readout, which is the standard SP-PLA method. Figure 2

shows the detection of a serial dilution of VEGF in PLA buffer. The LOD of the three

assays was in the low fM range and they all exhibited a broad dynamic range of up to

6 orders of magnitude.

A comparison of detection efficiency between different oligo systems for protein at 10

pM is summarized in Table 1. The number of molecules detected is calculated by Ct

vaules of q-PCR results. Detection efficiency is the number of molecules detected

divided by the number of molecules spiked in theoretically. The result shows that the

detection efficiency of Nick system is close to that of Biovic system while the S3

system is lower.

From above, we conclude that the Nick oligo system perform equally or even better

than the existing oligo systems for detecting proteins in buffer.

Page 18: Development of on-chip proximity ligation assay with in situ single

17  

Figure 2. Comparison of performance between different oligo systems. (a) Biovic system for SP-PLA.

(b) S3 system for in situ PLA. (c) Nick system. All measurements were performed on microparticles by

detecting a serial dilution of VEGF in PLA buffer. The y axes display Ct values of q-PCR, the x axes

display concentrations of the target proteins in pM. The 0.001 pM on x axis is a negative control where

no protein is included. Error bars along the y axes indicate the standard deviation from duplicates.

Page 19: Development of on-chip proximity ligation assay with in situ single

18  

Table 1. Comparison of detecting efficiency between different oligo systems

Oligo systems Ct

No. of

molecules

detecteda

Theoretically

spiked in

No. of

molecules

spiked in

Detection

efficiencyb

Biovic system 24 32768 10 pM*45 μl 2.70E+08 1/8267

S3 system 18 2097 10 pM*45 μl 2.70E+08 1/129175

Nick system 16 16777 10 pM*45 μl 2.70E+08 1/16146 a calculated by Ct values b No. of molecules detected divided by No. of molecules spiked in

3.1.2 Formation of circular DNA

To further validate the oligo system we investigated if the signal in q-PCR is from the

circular DNA. T7 exonuclease and phi29 polymerase were used after the enzymatic

ligation. T7 exonuclease has 5’-3’ exonuclease activity while phi29 polymerase has

3’-5’ exonuclease activity. Therefore, all the un-ligated products will be degraded

when exposed to both of them. Figure 3 shows that the signal from the reactions with

addition of both T7 exonoclease and phi29 polymerase is comparable to reactions

with addition of only phi29 polymerase. This result indicates that the un-ligated DNA

is neglectable and the signal detected in real-time PCR is derived from the DNA

circle.

Page 20: Development of on-chip proximity ligation assay with in situ single

 

Figu

detec

enzy

stand

3.2

3.2.

The

anti

desc

satu

exp

strep

anti

a se

high

the

high

ure 3. Detect

cting 10 pM

ymatic ligation

dard deviation

Immobiliza

.1 Compari

e immobiliz

ibody and

cribed in S

urated with

ected to oc

ptavidin-co

ibodies. Tho

erial dilution

her backgro

curve redu

her to lower

0

2

4

6

8

10

12

14

16

18

Ct

tion of circu

VEGF in P

n. The y axis

n from duplica

ation of cap

ison of new

zation of cap

oligonucleo

SP-PLA by

h biotinylat

cupy 10%

oated beads

ose two kin

n of VEGF

ound while l

uces gradua

r protein con

+T7 e

ular DNA. Th

PLA buffer. T

displays Ct v

ates.

pture antib

w immobiliz

pture antibo

otides affix

Darmanis

ted oligonu

of the oligo

were used t

nds of antibo

in PLA buf

lower signa

ally indicati

ncentration

exo +phi29

19 

he measurem

T7 exonucleas

values of q-P

body via DN

zation meth

ody by liga

xed on sup

et al. [18].

ucleotides a

os, were ad

to immobili

ody-immob

ffer (Figure

al comparing

ing that de

.

ments were pe

se and phi29

PCR. Error ba

NA ligation

hod with ex

ation of olig

pports was

The strept

and capture

dded and lig

ize compara

ilized beads

e 4). The ne

g to the orig

etection effi

+ phi29

erformed on

9 polymerase

ars along the y

n

xisted one

gonucleotide

compared

tavidin-coat

e conjugate

gated. In pa

able amount

s were used

ew immobil

ginal metho

ficiency is d

10 

microparticle

were added

y axis indicat

es linked to

d with the

ted beads w

es which w

arallel, the s

t of biotinyl

d in detectio

ization pres

od. The slop

decreased f

pM VEGF

es by

after

te the

o the

one

were

were

same

lated

on of

sents

pe of

from

Page 21: Development of on-chip proximity ligation assay with in situ single

20  

Figure 4. Comparison of new immobilization strategy (diamonds) with the original one (squares). All

measurements were performed on microparticles by detecting a serial dilution of VEGF in PLA buffer.

The y axis displays Ct values of q-PCR, the x axis displays concentrations of the target proteins in pM.

The 0.001 pM on x axis is a negative control where no protein is included. Error bars along the y axis

indicate the standard deviation from duplicates.

3.2.2 Optimization of immobilization method

In order to increase the detection efficiency, the amount of capture conjugates and

affixed oligos on the solid supports were titrated. Twenty-five percent of the beads

surface was covered with oligonucleotides and the amount of capture conjugates was

reduced five times for each experiment (Figure 5). We observed that the signal

increased when the amount of capture conjugates was decreased. As the concentration

of capture conjugates decreased to 4 nM, the signal was close to the control, however

the amount of capture antibodies was not enough for high protein concentrations as

could be seen in 1000 pM data point. The titration of oligonucleotides on the solid

supports showed a similar trend (Figure 6). A reduction of the oligonucleotides on the

surface resulted in the growth of signal. However, signals started to decrease when

only 0.2% of the surface was covered with oligos. The reason for this phenomenon

might be the competition of polyclonal antibodies. When the capture antibodies were

locating in close proximity on the solid support, the epitopes of each target protein

15

20

25

30

35

40

0.001 0.01 0.1 1 10 100 1000

Ct

VEGF (pM)

oligo saturated beads

control

Page 22: Development of on-chip proximity ligation assay with in situ single

21  

could be occupied by more than one antibody and the other two proximity probes

added later on will be blocked.

Figure 5. Titration of the capture conjugates. All measurements were performed on microparticles by

detecting a serial dilution of VEGF in PLA buffer. Twenty-five percent of the microparticles were

covered by oligos and the amount of capture conjugates was decreased five times for each experiment.

Control indicates the original SP-PLA immobilization method. The y axis displays Ct values of q-PCR,

the x axis displays concentrations of the target proteins in pM. The 0.001 pM on x axis is a negative

control where no protein is included. Error bars along the y axis indicate the standard deviation from

duplicates.

15

20

25

30

35

40

0.001 0.01 0.1 1 10 100 1000

Ct

VEGF (pM)

25% coverage 100nM

25% coverage 20nM

25% coverage 4nM

control

Page 23: Development of on-chip proximity ligation assay with in situ single

22  

Figure 6. Titration of the oligos on solid support. All measurements were performed on microparticles

by detecting a serial dilution of VEGF in PLA buffer. The amount of oligos on microparticles was

reduced five times for each experiment while the number of capture conjugates was constant. Control

indicates the original SP-PLA immobilization method. The y axis displays Ct values of q-PCR, the x

axis displays concentrations of the target proteins in pM. The 0.001 pM on x axis is a negative control

where no protein is included. Error bars along the y axis indicate the standard deviation from

duplicates.

3.3 Optimization of protein detection strategy

The original protocol, in which target proteins are incubated with capture antibodies

prior to the addition of the other two proximity probes, might encounter

antibody-competition problems. In order to avoid such problems, two strategies were

compared; 1) Three probes were incubated with target protein in solution, then the

protein-probe complex was fished out by ligating oligo on the probe to oligos

immobilized on magnetic beads (Figure 7, capture before ligation). 2) Two proximity

probes and the magnetic beads immobilized with capture antibodies via DNA ligation

were incubated with target protein in solution (Figure 7, capture after ligation). The

results indicate that the ‘capture after ligation’ method has comparable signal with

control yet higher background, the ‘capture before ligation’ method has comparable

signal-to-noise with the control.

15

20

25

30

35

40

0.001 0.01 0.1 1 10 100 1000

Ct

VEGF (pM)

5 % coverage 100nM

1 % coverage 100nM

0.2 % coverage 100nM

control

Page 24: Development of on-chip proximity ligation assay with in situ single

23  

Figure 7. Comparison of different protein detection strategy. All measurements were performed on

microparticles by detecting a serial dilution of VEGF in PLA buffer. Control indicates the original

SP-PLA immobilization method. The y axis displays Ct values of q-PCR, the x axis displays

concentrations of the target proteins in pM. The 0.001 pM on x axis is a negative control where no

protein is included. Error bars along the y axis indicate the standard deviation from duplicates.

3.4 Singleplex protein microarray

To investigate assay performance on planar flow cell surfaces, we used a glass slide as

solid support for the detection of VEGF. Capture conjugates were ligated to the

arrayed oligonucleotides and the procedure is the same as we described before. Cy3

labeled detection oligonucleotides were used for the detection of RCA products.

Epifluorescence microscopy was used as readout. RCA products within individual

sub-array were enumerated by BlobFinder. Representative microarray features for

different protein concentrations are shown in Figure 8. When the protein

concentration is 0.1 nM, the signal is too low to be proportional to others. The

characteristics of this assay are summarized in Table 2. The number of molecules

detected is calculated by the number of molecules in each sub-array multiplied by the

number of sub-arrays. The detection efficiency is the number of molecules detected

divided by the number of molecules theoretically spiked in. The detection efficiency

15

20

25

30

35

40

0.001 0.01 0.1 1 10 100 1000

Ct

VEGF (pM)

capture before ligation

capture after ligation

control

Page 25: Development of on-chip proximity ligation assay with in situ single

24  

is lower than that of the q-PCR readout.

Figure 8. Single molecule counting in singleplex protein microarray features. The image indicates a

microarray feature with RCA products detected by fluorescent labeled oligonucleotides.

Table 2. Summary of singleplex protein microarray performance

Protein conc. No. of RCA

products/sub-array

No. of

molecules

detected

Theoretically

spiked in

No. of

molecules

spiked in

Detection

efficiency

10nM 10253 4.00E+06 10 nM*40 μl 2.40E+11 1/58714

1nM 1189 5.00E+05 1 nM*40 μl 2.40E+10 1/50630

3.5 Duplex protein microarray

To further exploit the assay performance in a multiplex setting, a duplex protein

microarray was performed on glass slide. VEGF and Mouse IgG were employed as

target proteins and the ratio between them was 1:1. Specific protein barcode for each

Page 26: Development of on-chip proximity ligation assay with in situ single

25  

protein was included in the proximity probes. After on chip proximity ligation and

RCA, the amplified products were visualized by calling the first base using Illumina

sequencing reagents (Figure 9). Due to the similar emission spectra of the ‘G’ and ‘T’

and limitation of the filters used, ‘T’ (red) also showed up in the filter of ‘G’ (green)

as can be seen in a magnified subregion in Figure 9. The result of this assay is

summarized in Table 3. The count ratio of VEGF to Mouse IgG is 1:3 which will be

discussed later.

Figure 9. Single molecule counting in duplex protein microarray features. The image indicates a

microarray feature with RCA products detected by calling the first base with Illumina sequencing

reagents. Individual RCA products are featured in a magnified subregion.

Table 3. Summary of duplex protein microarray performance

Protein Spiked in

ratio

No. of RCA

products/sub-array Count ratio

VEGF 1 1140 1

Mouse

IgG 1 3603 3

Page 27: Development of on-chip proximity ligation assay with in situ single

26  

4. Discussion

The study of protein function in biological systems will improve our understanding of

human diseases. The ultimate goal of proteomics is to link and identify individual

protein with disease which requires a method for discovery and quantitative analysis

of biomarkers. Protein microarray offers a way of simultaneously studying a large

number of proteins in a minute amount of sample within single experiment. However,

the specificity and sensitivity of traditional protein microarrays could not meet the

demand of proteomic research. A highly specific and sensitive method for multiplex

detecting proteins is needed.

This project aimed to construct a highly specific multiplex protein detection method

with single molecule resolution. The method was based on PLA which was pioneered

in this group. Chip was recruited as solid support and multiple recognition events

were required for assurance of specificity. Sequencing of locally amplified RCPs

enables detection with single molecule resolution.

In this method, Nick oligo systems were designed to simplify the procedure of

multiplex detection. Two proximity probes were carrying their own barcodes and

corresponding connector oligonucleotides for different proteins and two universal

splints were needed to close the gap. The performance of this oligo system was

comparable to the existing oligo systems in terms of dynamic range, LOD and

efficiency. However, the system can be further optimized to increase efficiency, for

example, by reducing the two ligation events to one but maintaining the three

recognitions. One probe can have barcode and carrying the connector oligonucleotide,

the other probe can serve as the splint to close the gap and a third probe initiates the

RCA. In this way, the procedure can be further simplified and the decrease of ligation

events may increase the efficiency of the detection, since the ligation efficiency

contributes to the efficiency of the whole detection.

Page 28: Development of on-chip proximity ligation assay with in situ single

27  

The immobilization of this method was through linking oligonucleotide-conjugated

antibodies to existed oligonucleotides on the solid support. In this immobilization, the

capture antibodies are more flexible than the one in SP-PLA described by Darmanis et

al. So the chance that one protein being captured by different antibodies is higher.

This may block the binding of the other two probes which results in a decrease of

signal. The titration of capture conjugates and the employment of different incubation

methods help to increase the signal to some extent. Another way to solve this problem

is by using monoclonal antibodies instead of polyclonal ones. Those monoclonal

antibodies will bind to a defined epitope of the target protein so there is no

competition between probes and this approach could further improve the performance

of the assay.

Compared with microparticles, the detection efficiency on glass slide was lower. This

could be attributed to many reasons. First, the protocol of this experiment on glass

slides hadn’t been optimized. We could achieve better results by adjusting the

incubation time, the reaction temperature, the probe concentration, etc. In addition,

the surface kinetics of microarrays is not as good as microparticles, which means the

reaction on planar surface is not as efficient as the one on particles. One way to

improve is by decreasing the height of the microarray chamber. Last but not least,

surface chemistry of the array can be changed to achieve better result. The

immobilization of oligonucleotides now is through biotin streptavidin interactions.

However, there are many commercially available chips with different surface

chemistries which may improve the efficiency of the assay.

In duplex protein microarray, the detected ratio of VEGF and Mouse IgG was

different from the ratio of input. The reason may be due to the different concentration

of capture antibodies for VEGF and Mouse IgG in the initial immobilization steps,

resulting in different amount of capture probes were affixed on the slide. Another

explanation for this may be the varies of detection efficiency between different

proteins. Furthermore, the readout for this detection could be improved by using

Page 29: Development of on-chip proximity ligation assay with in situ single

28  

microscope with proper filters or the Illumina sequencer.

Page 30: Development of on-chip proximity ligation assay with in situ single

29  

Acknowledgement

I would like to thank Ulf Landegren and Masood Kamali-Moghaddam for giving me

the opportunity to work in this group. Also, I would give my thanks to my supervisor

Rachel Nong for all the inspiring ideas, patience in experiments and critical comments

on the thesis. Last but not least, I would like to thank all the people in the group of

Molecular Tools for all the help they gave me and this wonderful research experience.

Page 31: Development of on-chip proximity ligation assay with in situ single

30  

References

[1] Diamandis E.P. and. Yousef G.M. (2002) Human Tissue Kallikreins: A Family of New Cancer

Biomarkers. Clinical Chemisry 48,1198-1205

[2] Ye B., Cramer D.W., Skates S.J., Gygi S.P., Pratomo V. Fu L., Horick N.K., Licklider L.J., Schorge

J.O., Berkowitz R.S. and Mok S.C. (2003) Haptoglobin-α Subunit As Potential Serum Biomarker in

Ovarian Cancer Identification and Characterization Using Proteomic Profiling and Mass Spectrometry.

Clinical Cancer Research 9, 2904-2911

[3] Zangar, R.C.; Daly, D.S.; White, A.M. (2006) ELISA microarray technology as a high-throughput

system for cancer biomarker validation. Expert Review of Proteomics 3, 37-44

[4] Kucher N., Goldhaber S.Z. (2003) Cardiac Biomarkers for Risk Stratification of Patients With

Acute Pulmonary Embolism. Circulation 108, 2191-2194

[5] Sieber M., Hoffmann D., Adler M., Vaidya V.S., Clement M., Bonventre J.V., Zidek N., Rached E.,

Amberg A., Callanan J.J., Dekant W. and Mally A. (2009) Comparative Analysis of Novel Noninvasive

Renal Biomarkers and Metabonomic Changes in a Rat Model of Gentamicin Nephrotoxicity.

Toxicological Sciences 109 (2), 336-349

[6] Ray S., Britschgi M., Herbert C., Takeda-Uchimura Y., Boxer A., Blennow K., Friedman L.F.,

Galasko D.R., Jutel M., Karydas A., Kaye J.A., Leszek J., Miller B.L., Minthon L., Quinn J.F.,

Rabinovici G.D., Robinso W.H., Sabbagh M.N., So Y.T., Sparks D.L., Tabaton M., Tinklenberg J.,

Yesavage J.A., Tibshirani R. and Wyss-Coray T. (2007) Classification and prediction of clinical

Alzheimer's diagnosis based on plasma signaling proteins. Nature Medicine 13, 1359-1362

[7] Anderson, N. L., and Anderson, N. G. (2002) The human plasma proteome: history, character, and

diagnostic prospects. Molecular & Cellular Proteomics 1,845–867

[8] Groseclose M.R., Massion P.P., Chaurand P., Caprioli R.M. (2008) High-throughput proteomic

analysis of formalin-fixed paraffin-embedded tissue microarrays using MALDI imaging mass

spectrometry. Proteomics 8, 3715-3724

[9] Domon B. and Aebersold R. (2010) Options and considerations when selecting a quantitative

proteomics strategy. Nature biotechnology 28, 710-721

[10] Prakash A., Piening B., Whiteaker J., Zhang H., Shaffer S.A., Martin D., Hohmann L., Cooke K.,

Page 32: Development of on-chip proximity ligation assay with in situ single

31  

Olson J.M., Hansen S., Flory M.R., Lee H., Watts J., Goodlett D.R., Aebersold R., Paulovich A. and

Schwikowski B. (2007) Assessing Bias in Experiment Design for Large Scale Mass

Spectrometry-based Quantitative Proteomics. Molecular & cellular proteomics 6, 1741-1748

[11] Engvall E, Perlman P (1971) Enzyme-linked immunosorbent assay (ELISA). Quantitative assay of

immunoglobulin G. Immunochemistry 8 (9), 871–874

[12] Nishi K., Chikae M., Hatano Y., Mizukami H., Yamashita M., Sakakibara R. and Tamiya E.(2002)

Development and application of a monoclonal antibody-based sandwich ELISA for quantification of

Japanese medaka (Oryzias latipes) vitellogenin. Comparative Biochemistry and Physiology Part C:

Toxicology & Pharmacology 132 (2), 161-169

[13] Ronald A. and Stimson W. H. (1998). The evolution of immunoassay technology. Parasitology 117,

S13-27

[14] Kingsmore S.F. (2006) Multiplexed protein measurement: technologies and applications of protein

and antibody arrays. Nature Reviews Drug Discovery 5, 310-321

[15] Fredriksson S, Gullberg M, Jarvius J, Olsson C, Pietras K, Gústafsdóttir S.M, Ostman A,

Landegren U (2002) Protein detection using proximity-dependent DNA ligation assays. Nature

Biotechnology 20 (5), 473-7

[16] Gullberg M., Gustafsdottir S.M., Schallmeiner E., Jarvius J., Bjarnegard M., Betsholtz C.,

Landegren U., and Fredriksson S. (2004) Cytokine detection by antibody-based proximity ligation.

PNAS 101 (22), 8420-8424

[17] Darmanis S., Nong R.Y., Hammond M., Gu J., Alderborn A., Vanelid J., Siegbahn A.,

Gustafsdottir S., Ericsson O., Landegren U. and Kamali-Moghaddam M. (2010) Sensitive Plasma

Protein Analysis by Microparticle-based Proximity Ligation Assays. Molecular & Cellular Proteomics

9, 327-335

[18] Darmanis S, Nong R.Y., Vanelid J., Birgisson H., Siegbahn A., Isaksson M., Ericsson O.,

Fredriksson S., Glimelius B.,Wallentin L., Gustafsson M. G., Kamali-Moghaddam M., and Landegren

U. (2010) Multiplexed solid-phase proximity ligation assays: Highly specific, parallel protein

measurements with DNA readout. Manuscript.

[19] Soderberg O., Gullberg M., Jarvius M., Ridderstrale K., Leuchowius K.J., Jarvius J., Wester K.,

Hydbring P., Bahram F., Larsson L.G, Landegren U. (2006) Direct observation of individual

endogenous protein complexes in situ by proximity ligation. Nature Methods 3 (12), 995-1000

Page 33: Development of on-chip proximity ligation assay with in situ single

32  

[20] Weibrecht I., Leuchowius K.J., Clausson C.M., Conze T., Jarvius M., Howell W.M.,

Kamali-Moghaddam M. and Söderberg O. (2010) Proximity ligation assays: a recent addition to the

proteomics toolbox. Expert Review of Proteomics. 7(3), 401-9

[21] Allalou A, Wahlby C. (2009) BlobFinder, a tool for fluorescence microscopy image cytometry.

Comput. Methods Programs Biomed. 94(1), 58–65

[22] Pinidiyaarachchi A, Zieba A, Allalou A, Pardali K, Wahlby C. (2009) A detailed analysis of 3D

subcellular signal localization. Cytometry A 75(4), 319-328.

[23] Schuster S.C. (2008) Next-generation sequencing transforms today’s biology. Nature Methods

5(1), 16-18

[24] Marguerat S., Wilhelm B.T. and Bahler J. (2008) Next-generation sequencing: applications beyond

genomes. Biochemical Society Transactions. 36, 1091-6

[25] Technology Spotlight: Illumina Sequencing

Page 34: Development of on-chip proximity ligation assay with in situ single

33  

Supplementary Table 1

Name Sequence SLC1 5'-Thiol-CGCATCGCCCTTGGACTACGACTGACGAACCGCTTTGCCTGACTGA

TCGCTAAATCGTG-3' SLC2 5'-TCGTGTCTAAAGTCCGTTACCTTGATTCCCCTAACCCTCTTGAAAAATTCG

GCATCGGTGA-Thiol-3' BioFwd 5'-CATCGCCCTTGGACTACGA-3' BioRev 5'-GGGAATCAAGGTAACGGACTTTAG-3' BioSplint 5'-TACTTAGACACGACACGATTTAGTTT-3' S3 arm 1 5'-Thiol-AAAAAAAAAAGACGCTAATAGTTAAGACGCTTUUU-3' S3 arm 2 5'-Thiol-AAAAAAAAAATATGACAGAACTAGACACTCTT-3' S3 Backbone 5'-CTATTAGCGTCCAGTGAATGCGAGTCCGTCTAAGAGAGTAGTACAGCAGC

CGTCAAGAGTGTCTA-3' S3 Splint 5'-GTTCTGTCATATTTAAGCGTCTTAA-3' S3 Fwd 5'-GTAGTACAGCAGCCGTCA-3' S3 Rev 5'-TTAGACGGACTCGCATTC-3' Nick1 arm1 5'-Thiol-TTTTTTCGCGTGATGGAGTCTCCCACGATTTAGGTGGGAGACTCCAT

CACGGCAATGATACGG-3' Nick1 arm2 5'-Thiol-TTTTTTCGTCAACAGGAGTCTCCCGACCACCGATCGGGAGACTCCT

GTTGAGCTTAGACACGA-3' Nick1 Splint1

5'-CUAAAUCGUGUCGUGUCUAA-3'

Nick1 Splint2

5'-UCGGUGGUCGCCGUAUCAUU-3'

Nick1 Fwd 5'-AATGATACGGCGACCACCGA-3' Nick1 Rev 5'-CTAAATCGTGTCGTGTCTAA-3' Nick2 arm1 5'-CACGATTTAGATCACGTCGCAGTGTGACACTCTTTCCCTACACACTGCGA

CGTGATTTTTTT-Thiol-3' Nick2 arm2 5'-CGACGCTCTTCCGATCTTGTTGATCGGCAGTGTCTTAGACACGACACTGC

CGATCAACACGTTTTTT-Thiol-3' Nick2 arm3 5'-CACGATTTAGATCACGTCGCAGTGTGACACTCTTTCCCTACACACTGCGA

CGTGATTTTTTT-Thiol-3' Nick2 arm4 5'-CGACGCTCTTCCGATCTTGTTGATCGGCAGTGTCTTAGACACGACACTGC

CGATCAACACGTTTTTT-Thiol-3' Nick2 Fwd 5'-ACACTCTTTCCCTACACGACGCTCTTCCGATCT-3' Nick2 Rev 5'-CTAAATCGTGTCGTGTCTAA-3' Nick2 Bio-capture

5'-Biotin-TTTTTTTTTTCAAGCAGAAGACGGCATACGA-3'

Nick2 capture

5'-GCAGTGTTAGATCGTGTCGTGTCTAACACTGCTCGTATGCCGTCTTCTTTTTTTTTT-3'