development of a microfluidic device for selective

113
DEVELOPMENT OF A MICROFLUIDIC DEVICE FOR SELECTIVE ELECTRICAL LYSIS OF PLASMA MEMBRANES OF SINGLE CELLS by Duoaud Fawz Shah A thesis submitted in conformity with the requirements for the degree of Master of Science Medical Biophysics University of Toronto © Copyright by Duoaud Fawz Shah 2010

Upload: others

Post on 10-Nov-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

DEVELOPMENT OF A MICROFLUIDIC DEVICE FOR SELECTIVE ELECTRICAL LYSIS OF PLASMA

MEMBRANES OF SINGLE CELLS

by

Duoaud Fawz Shah

A thesis submitted in conformity with the requirements for the degree of Master of Science

Medical Biophysics University of Toronto

© Copyright by Duoaud Fawz Shah 2010

ii

Development of a microfluidic device for the selective electrical

lysis of plasma membranes of single cells

Duoaud Fawz Shah

Master of Science

Medical Biophysics

University of Toronto

2010

Abstract

A primary objective of modern biology is to understand the molecular mechanisms which

underlie cellular functions and a crucial part of this task is the ability to manipulate and analyze

individual cells. As a result of interdisciplinary research, microfluidics may become the forefront

of analytical methods used by biologists. This technology can be used to gain unprecedented

opportunities for cell handling, lysis and investigation on a single cell basis. This thesis presents

the development of a microfluidic device capable of selecting individual cells and performing

selective electrical lysis of the plasma membrane, while verifying intactness of the nuclear

membrane. The device is fabricated by an improved photolithography method and integrates

molten solder as electrodes for lysis by a DC electric field. Quantification of lysis is

accomplished by video and image analysis, and measurement of the rate of ion diffusion from

the cell.

iii

Acknowledgments

The completion of this thesis has been the most significant academic challenge that I have faced;

however, overcoming the hurdles along the way would not have been possible without the

guidance, direction and support of my supervisor, Dr. Lothar Lilge. He allowed me to work

independently, and to learn from my mistakes, while always being available and accessible to

guide me through difficult times. Most importantly, I am grateful to him for helping me to

improve my thought process as a researcher. His leadership style, coupled with his overall

character has proven to me that I had the ideal supervisor for my graduate experience.

I would also like to thank the members of my supervisory committee, Dr. Jim Woodgett

and Dr. Alex Vitkin, for their support and the insight they provided to help steer my project

towards its objectives.

Sharing the challenges and victories of my work with members of the Lilge group has

made my graduate experience fulfilling and has helped me in overcoming frustration, and

sometimes coming up with inventive solutions, while keeping me grounded. For this, I would

like to thank this team of talented and extremely supportive students and staff. They provided an

environment which made it enjoyable to be at work daily, while enhancing my personal

experience as a graduate student and I wish them success in all their endeavours. Specifically, I

would like to thank Luc Charron for his advice and our many discussions which helped

overcome several fabrication challenges of the project, and Dr. Kumudesh Sritharan for helping

me with the fluorescent labelling and other in vitro work. The simulations in COMSOL would

not have been possible without the tutelage of Dr. Mohamed Abdelgawad, to whom I am

indebted. I am also grateful to Jane Walter, Benjamin Lai and Jennifer Street for their friendship

and support.

I owe the deepest gratitude to my mother for raising me to be well-rounded while

instilling the value of education. She has supported me through all my personal and academic

endeavours and has been an inspiration to me at all times. I would also like to thank my uncle

and aunt who have always encouraged me to remember my faith throughout my studies, and my

grandmother for her kindness and numerous meals which helped get me through graduate school.

Lastly, to my wonderful Daanish, I thank you for your patience, understanding and love

throughout this journey as your unwavering support has been my source of motivation.

iv

Table of Contents

Acknowledgments .......................................................................................................................... iii

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

List of Acronyms and Symbols ...................................................................................................... xi

Chapter 1 Introduction .................................................................................................................... 1

1 1

1.1 Overview of single cell analysis ......................................................................................... 1

1.2 Techniques for single cell analysis ..................................................................................... 6

1.2.1 Flow cytometry ....................................................................................................... 7

1.2.2 Fluorescence microscopy ........................................................................................ 8

1.2.3 Capillary electrophoresis......................................................................................... 8

1.2.4 Requirements for alternative analysis method ........................................................ 9

1.3 Single cell analysis on microfluidic platforms .................................................................... 9

1.3.1 Techniques for single cell analysis in microfluidics ............................................. 10

1.3.2 Microfluidics platform for multiple technique integration ................................... 11

1.4 Motivation for selective single cell lysis........................................................................... 12

1.5 Thesis organization ........................................................................................................... 13

Chapter 2 Electrical lysis of plasma membrane with intact nuclei ............................................... 14

2 14

2.1 Introduction ....................................................................................................................... 14

2.1.1 The plasma membrane and electroporation .......................................................... 14

2.1.2 Electrical lysis ....................................................................................................... 18

2.2 Materials and Methods ...................................................................................................... 22

2.2.1 Electric field simulations ...................................................................................... 22

2.3 Results and Discussion ..................................................................................................... 24

v

2.3.1 Electric field distribution dependency on geometry ............................................. 24

2.3.2 Electric field dependency on applied voltage ....................................................... 27

2.3.3 Determination of heat transfer to cell during lysis ................................................ 29

2.4 Conclusion ........................................................................................................................ 32

Chapter 3 Fabrication of microfluidics device with integrated electrodes for single cell lysis .... 33

3 33

3.1 Introduction ....................................................................................................................... 33

3.1.1 Design requirements of a single cell microfluidic device ..................................... 33

3.1.2 UV photolithography as a fabrication tool ............................................................ 34

3.2 Materials and Methods ...................................................................................................... 35

3.2.1 Photo mask production and optimization ............................................................. 35

3.2.2 Preparation of glass substrates .............................................................................. 38

3.2.3 SU-8 exposure and development .......................................................................... 41

3.2.4 Device fabrication by rapid prototyping of PDMS ............................................... 44

3.2.5 Material characterization of integrated electrodes ................................................ 47

3.3 Results and Discussion ..................................................................................................... 52

3.3.1 UV absorbance and micrograph resolution of photo masks ................................. 52

3.3.2 SU-8 spin coating and exposure ........................................................................... 54

3.3.3 PDMS device development ................................................................................... 57

3.3.4 Electrode material characterization ....................................................................... 60

3.3.5 Microfluidic device with integrated electrodes ..................................................... 64

3.4 Conclusion ........................................................................................................................ 65

Chapter 4 In vitro experimental verification of plasma membrane lysis ...................................... 66

4 66

4.1 Introduction ....................................................................................................................... 66

4.2 Materials and Methods ...................................................................................................... 67

vi

4.2.1 Experimental set-up .............................................................................................. 67

4.2.2 Visual monitoring of electric field induced cell lysis ........................................... 71

4.2.3 Data analysis ......................................................................................................... 72

4.3 Results and Discussion ..................................................................................................... 74

4.3.1 Flow control .......................................................................................................... 74

4.3.2 Hoechst and calcein staining and photobleaching ................................................ 74

4.3.3 Electrical lysis of 3T3 and 9L plasma membranes ............................................... 77

4.3.4 Variation of diffusion rate due to electric field ..................................................... 85

4.4 Conclusion ........................................................................................................................ 86

Chapter 5 Summary and Future Work .......................................................................................... 87

5 87

5.1 Summary ........................................................................................................................... 87

5.2 Contributions and perspectives ......................................................................................... 88

5.2.1 Microfluidic device fabrication for single cell electrical lysis .............................. 88

5.2.2 Selective electrical lysis of plasma membrane of single cells .............................. 89

5.3 Future Work ...................................................................................................................... 89

5.3.1 Engineering and fabrication aspects ..................................................................... 89

5.3.2 Integration of components .................................................................................... 90

5.3.3 Selective lysis by AC electric field ....................................................................... 92

5.3.4 Multiple fluorescent staining ................................................................................ 92

vii

List of Tables

1.1 Examples of single cell analysis techniques useful to investigate heterogeneity…….6

2.1 Dependency of simulated electric field range on applied voltage…………………....28

2.2 Calculated values of the capacitance of each dielectric layer………………………...31

3.1 Summary of printing resolution and photo films used by different suppliers………...37

3.2 Properties of electrically conductive pastes and epoxies……………………………..49

3.3 Thickness and OD measurements of 3 different photo masks…………......................53

3.4 Initial and final radiant energy per cm2 due to photo mask absorption…....................55

3.5 Variation of channel widths and gaps during fabrication process………....................60

3.6 Electrical properties of all conductive materials used as integrated electrodes……....63

4.1 Summary of diffusion rates and fluorescent intensity data for nine 3T3 cells….…….80

4.2 Summary of diffusion rates and fluorescent intensity data for nine 9L cells…............84

viii

List of Figures

1.1 Illustration of a eukaryotic cell………………………………………………………..2

1.2 The cell as a system with common input and output signals………………………….5

1.3 Integrated microfluidic device for single cell transport, lysis, capillary

electrophoresis and detection………………………………………………………....12

2.1 Illustration of phospholipid bilayer membrane……………………………………….14

2.2 Equivalent circuit representation of cell in suspension……………………………….15

2.3 Polarization of plasma membrane under influence of an electric field……………… 16

2.4 Modeled results for ΔVmem and ΔVorg, showing optimal frequency region……….......19

2.5 Illustration of various electrode configurations used in microfluidic research………..22

2.6 Geometric simulation of intended microfluidic chip including material properties......23

2.7 a) Simulation of electric field distribution between teeth-like electrodes;

b) Resistive heating between electrodes………………………………………………24

2.8 a) Simulation of electric field distribution between embedded 3-D electrodes,

with arrows shown in insulating region between electrodes and channel;

b) Resistive heating between electrodes………………………………………………26

2.9 Electric field strength as a function of distance in x-axis, showing regions of the

fluidic channel and insulating regions at an applied voltage of 4V…………………..27

2.10 Plot of electric field dependency on applied voltage as obtained from COMSOL

simulations…………………………………………………………………………….29

2.11 Parallel plate capacitor model of electrode – insulating region (C1 and C3) –

channel system (C2), showing insulating regions and channels as capacitors, with

corresponding thicknesses…………………………………………………………….30

3.1 Schematic AutoCAD drawing of integrated microfluidic device and injector

structure region………………………………………………………………………..36

3.2 Pre-printing illustration of photo mask and injector structure region………………...37

3.3 Spin speed vs. film thickness for various SU-8 photoresists…………………………40

3.4 Set-up of various layers during UV exposure of SU-8……………………………….42

ix

3.5 Illustration of cross sectional view of a microfluidic channel and method of

measuring side wall angle…………………………………………………………….44

3.6 Illustration of multi-step soft lithography processs…………………………………...45

3.7 Corona treater with PDMS chip and spin coated cover glass on a motorized

stage…………………………………………………………………………………...47

3.8 a) Tubing filled to measure resistance b) Cross sectional view and c) top view

of chip with electrode filled; d) Cross sectional view of chip showing resistance

measurement with inserted copper wire………………………………………………50

3.9 a) Absorbance as a function of wavelength for printed parts of photo mask,

b) Absorbance as a function of wavelength for transparent part of each photo mask...52

3.10 Micrograph images of mask features from 3 suppliers………………………………..54

3.11 Resolution improvement of SU-8 structures using glycerine for different

exposure times…………………………………………………………………………56

3.12 Micrograph images of PDMS channels showing side wall angle improvement

with glycerine at an exposure time of 12.5s…………………………………………...57

3.13 Side wall angle as a function of channel width for different exposure times…………57

3.14 (a) Central plane in the x-axis; (b) Central plane showing z-axis view (top),

and y-axis view through the fluidic channel (right); (c) Sample profiles through

various regions…………………………………………………………………………59

3.15 Physical properties of integrated electrodes made of different material, with

(a) carbon-PDMS; (b) nickel paste; (c) silver paste……………………………….…..61

3.16 Resistance as a function of tubing length for electrode materials………………….….62

3.17 Final microfluidic device with integrated solder electrodes……………………….….64

4.1 Image of flow control system……………………………………………………….....68

4.2 Calcein AM molecule (C46H46N2O23) and excitation/emission spectra…………….....69

4.3 Hoechst 333342 molecule (C27H37Cl3N6O4) and excitation/emission spectra…….…..70

4.4 Circuit diagram of function generator with external DC offset…………………….....71

4.5 Illustration of direction of fluorescent profile analysis………………………………..73

4.6 a) Hoechst staining of nuclei in cell reservoir; b) Single cell Hoechst stain

depicting line along which fluorescent intensity profile………………………………74

4.7 a) Hoechst fluorescent intensity profile as a function of position for various time

points; b) Area under each intensity peak as a function of time………………………75

x

4.8 Calcein staining of cytoplasm in cell reservoir; b) Single cell calcein stain

depicting line along which fluorescent intensity profile………………………………76

4.9 a) Calcein fluorescent intensity profile as a function of position for various time

points; b) Area under each intensity peak as a function of time……………………....76

4.10 a) Fluorescent image of cytoplasm, a) before lysis, 0s; b) after lysis, 4s……………..77

4.11 Fluorescent intensity profile as a function of position for 3 different 3T3 cells

on the same chip, a) cytoplasmic changes over time; b) nucleic changes between

t = 0s and t = 6s………………………………………………………………………..78

4.12 Plot of FWHM vs. Time, showing diffusion region and start of electric field

application……………………………………………………………………………..79

4.13 Fluorescent intensity profile as a function of position for 3 different 9L cells on

the same chip, a) cytoplasmic changes over time; b) nucleic changes between

t = 0s and t = 6s………………………………………………………………………..82

4.14 Plot of FWHM vs. Time, showing diffusion region and start of electric field

application……………………………………………………………………………..83

4.15 Dependency of diffusion rate of calcein on mean electric field strength for 3T3 cells..85

5.1 Parallelized system with multiple electrode filling ports……………………………...91

5.2 (a) Staining of nucleus with Hoechst and mitochondria with Mitotracker Red;

(b) Triple staining of cytoplasm (Calcein AM), nucleus (Hoechst) and plasma

membrane (R18)……………………………………………………………………….92

xi

List of Acronyms and Symbols

Acronyms

2-D 2 dimensional

3-D 3 dimensional

AC Alternating current

Bn Bismuth

CAD Computer-aided design

DC Direct current

DPI Dots per inch

fps Frames per second

FWHM Full width at half maximum

GFP Green fluorescent protein

GaIn Gallium-Indim

H2SO4 Sulphuric acid

H2O2 Hydrogen peroxide

In Indium

ITO Indium tin oxide

LIF Laser induced fluorescence

MEM Minimum essential medium

MEMS Microelectromechanical systems

OD Optical density

PDMS Polydimethylsiloxane

PMMA Polymethylmethacrylate

rad Radians

RPM Revolutions per minute

RTV Room temperature vulcanization

Sn Tin

USA United States of America

UV Ultraviolet

xii

V:V Volume to volume

VEGF Vascular endothelial growth factor

Symbols

ERAD Radiant energy [mJ]

P Lamp power [mW]

texp Exposure time [s]

ρ Electrical resistivity [Ω·cm]

σ Electrical conductivity [S·m-1

]

R Resistance [Ω]

A Surface area [m2]

l Length [m]

AOD Optical density [dimensionless]

I Radiant intensity [mW·cm-2

]

I0 Initial radiant intensity [mW·cm-2

]

u Fresnel number [dimensionless]

x Slit width [µm]

λ Wavelength [µm]

υ Side wall angle [0]

y Vertical distance between mask and substrate [µm]

RE Resistance of extracellular medium [Ω]

CE Capacitance of extracellular medium [F]

RC Resistance of cytoplasm [Ω]

CM Capacitance of cell membrane [F]

E Electric field [V·m-1

]

r Radius [m]

θ Polar angle [0]

t Time/pulse duration of the electric field [s]

f Frequency [Hz]

τc Charging time constant [s-1

]

xiii

Continued

εr Relative permittivity [dimensionless]

ε0 Permittivity of free space [F·m-1

]

Vapp Applied voltage [V]

d Distance of separation [m]

C Capacitance [F]

CT Total capacitance [F]

W Work [J]

m Mass [kg]

c Specific heat capacity [J·kg-1

·K-1

]

ΔT Change in temperature [K]

I Current [A]

Ec Dissipated energy [J]

1

Chapter 1 Introduction

1.1 Overview of single cell analysis

General characteristics of eukaryotic cells

Cells are the fundamental unit of life, controlling complex biochemical systems and hence

containing a variety of different molecules including proteins, DNA, RNA, phospholipids and

other smaller molecules existing within hydrophilic or lipophilic compartments of the cell. The

plasma membrane of cells is made up of phospholipids and proteins which regulate the exchange

of other molecules between the intracellular and extracellular environment of the cell and also

facilitates communication with the cell‟s exterior. Major organelles such as the nucleus,

mitochondria and lysozomes, also have membranes of similar composition to that of the plasma

membrane and isolate the intra-organelle compounds from the rest of the intracellular

environment.

Analysis of cellular contents varies in complexity depending on the specific focus of the

assay due to the available concentrations of molecules and ions within a cell, and may differ

between cell cycles. The type and concentration of proteins can vary significantly as a result of

oxidation, glycosylation and phosphorylation. A typical eukaryotic cell varies in size from 5 -

500µm and has a total volume of 0.1 – 0.8 pL, with low copy numbers of DNA, mRNA, and

proteins. For example, a normal cell has 1 – 2 copies of a specific DNA sequence, resulting in

nucleic acids representing ~7% of the cell‟s mass, while proteins account for ~15% of the mass,

ranging from 100fg - 10µg depending on the size of the cell [1].

2

Figure 1.1: Illustration of a eukaryotic cell (Adapted from [2])

Importance of single cell analysis

The ability to detect, identify, quantify and structurally analyze biomolecules of a particular type

from a large population of cells has led to a wealth of knowledge in molecular and cell biology,

which has culminated in the completion of the human genome sequence and that of hundreds of

other species. Well established protocols for gel electrophoresis, western, southern and northern

blotting, have enabled the separation of proteins, DNA and RNA respectively. In addition, X-ray

crystallography and NMR methods have solved the structures of complex biomolecules at atomic

resolution [3], mass spectrometry has identified and quantified a wide range of proteins, lipids

and metabolites [4], while microarrays probe large numbers of genes and proteins. These

methods and protocols collect a large volume of high-quality data based on population analysis.

While the techniques described above used by molecular biologists have yielded a wealth

of information on the properties of molecules, a systems biologist using those techniques would

be substantially data-starved due to a lack of information on the context in which molecules

operate and other biochemical processes which occur on short timescales or non-synchronously.

3

This includes, but is not limited to, enzyme – substrate relationships, physical interactions and

gene regulatory networks and their dynamic changes. A challenging, yet informative solution to

these dilemmas is comprehensive and quantitative analysis at the single cell level. The challenge

lies in detection, isolation, transport and in most instances, lysis of the cell of interest, while

highly sensitive analytical techniques are required since the amount of analyte present in or

extracted from a single cell is minute. Meanwhile, single cell measurements are informative

since a unique set of data is obtained for each cell and can be compared to or combined with data

from other individual cells to compute the distribution of the measured value over a population

of cells. Conversely, classical methods such as flow cytometry yield data that is averaged over a

large number of cells in each assay.

Cells can be classified at a significantly improved resolution by gaining knowledge of the

distribution and statistical significance of values over a population of individual cells, which

permits the detection of cellular variabilities, and for the discrimination between stochastic and

deterministic events within a cell. The ability to carry out extensive and robust measurements on

single cells offers the opportunity to further advance the knowledge and understanding of

cellular functions for the purpose of biology and medicine.

Heterogeneity of single cells and proteomics

Cellular heterogeneity is evident among individual cells that may be identical in appearance and

even those born from the same parent cell, as shown by Hu et al [5], as they differ in numerous

characteristics which include gene and protein expression, concentration of critical metabolite or

ion, and patterning cellular response to stimuli.

In proteomics, the spatial and temporal location of a protein is required to explain its

function; however, a cell responding to its environment, passing through cell cycles or recycling

its content results in synthesis, modification and degradation of proteins. Consequently, not all

proteins are present in all cells, with many having a fleeting existence. Di Carlo and Lee [6]

eloquently explain cellular heterogeneity by considering protein expressions from a bimodal

distribution of a specific cell line with high and low copy numbers of a specific protein. Classical

ensemble protocols would mask the bimodal distribution within the cell ensemble by detecting

only a mean value across all cells, while analysis of individual cells could resolve two distinct

subpopulations – one with high and another with low copy numbers – thus revealing two

different states of expression levels. The same is true, regardless of what is being assayed,

4

whether it be mRNA or other small molecules. While an assessment of expression levels do not

necessarily address function directly, the knowledge of when and where a gene is expressed can

provide information regarding the potential roles of these genes and possibly lead to gene

discovery in other species [7].

In the realm of stem cell research, recognizing heterogeneity and being able to monitor

the proteome on a cell by cell basis would yield greater understanding of the steps involved in

development. As pluripotent stem cells progress first to precursor cells then into differentiated

progeny cells, their protein expression level changes as is similarly observed when an embryo

changes from a zygote into a fully developed individual [1].

By hierarchical processes of proliferation and differentiation, stem cells are able to

generate large numbers of mature cells, while the stem cell pool itself is continuously self-

renewed. These developmental processes exist in many adult tissues, including colon, skin, blood

and brain. The hypothesis of the cancer stem cell suggests that the balance between

differentiation and self-renewal becomes deregulated while the basic hierarchical structure

remains [5, 8]. Evidence of cellular heterogeneity in populations at various stages of

differentiation exists in tumours and biopsies have shown that the majority of cells within the

tumour may be normal, while within the sub-population of abnormal cells significant

heterogeneity exists. It is further hypothesized that cell to cell heterogeneity in protein expression

and cellular composition of a tumour increases as the disease progresses and can thus be

correlated with prognosis [9].

Neuroscience offers another example of heterogeneity, where individual neurons in the

central nervous system reflect differences in their contents and architecture [10], making the

brain the most complex organ in all vertebrates. Classification of these differences offers the

opportunity to better understand the function of each cell and the biological neural network.

Single cells as individual systems

Cell biology seeks to identify how the collection of environmental stimuli to which a cell is

exposed may influence the behaviour of that cell. By considering the cell to be a system,

processing time-dependent input signals into output responses (Fig. 1.2), and being able to

predict this relationship, can result in an understanding of higher level organization of tissues,

organs and organisms, which may aid in determining therapeutic approaches to correct flaws

within the organization. An example of this is the cellular microenvironment or „niche‟, which

5

stem cell differentiation and self-renewal are dependent upon. Additionally, extracellular signals

influence programmed cell division and apoptosis on which normal form and structure of

organisms rely. These environmental factors include chemical signals, biological signals, and

physical signals inclusive of electrical, mechanical and thermal factors.

Figure 1.2: The cell as a system with common input and output signals (Adapted from [11])

It would be expected that with knowledge of the ensemble of environmental stimuli, a

prediction of the expected behaviour of a particular cell could be made; however, cells under

apparently identical environmental conditions have displayed heterogeneity [12], attributed

partly to probabilistic behaviour in the decision making process connecting input and output

signals. Because of the heterogeneity within a population, researchers seek tools that afford the

ability to analyze a quantity of single cells exposed to controlled input signals to determine the

variance in responses. Furthermore, this requires the analysis of as many analytes as possible

from each cell to understand the variety of internal processes that may have occurred, leading to

the observed responses, and the spatial and temporal location of the analytes.

6

1.2 Techniques for single cell analysis

A multitude of technologies now exist for the analysis of variations in chemical constituents in

single cells within a population. The tool chosen is typically dependent upon the analyte of

interest, the number of analytes to be detected, mechanism of detection and the influence of the

analytical tool by interference or modification of cellular constituents. Table 1.1 summarizes

some techniques used to investigate heterogeneity in single cells, and is followed by a brief

description of three of the more common methods.

Technique Single cell measurement Cell types investigated

Flow cytometry Noise in abundances of GFP- fusion

proteins

Yeast

Fluorescence

microscopy

Intracellular calcium release to

identify subpopulations differing in a

particular receptor

Human osteoblasts

CE – biomolecules Two dimensional separation of

proteins that are fluorescently

labelled on-line

MC3T3-E1 osteoprogenitor

and MCF-7 breast cancer cells

CE – organelles Separation and detection of

mitochondria labelled with DsRed2

143B osteosarcoma cells

Optical well arrays Kinetics of various gene expression

using GFP reporter

Escherichia coli

Electrochemical

detection

Time profile of bursts of insulin

secretion

Rat and human pancreatic beta

cells

Raman

microspectroscopy

Changes in Raman spectra indication

coexisting cell types

Clostridium beijerinckii in an

acetone-butanol fermentation

reactor

7

MALDI-MS Identification of neuropeptides Neuron cells isolated from

Aplysia californica

LCM and cDNA

microarray analysis

Gene expression profiling pointing

to two subpopulations

CA1 neurons from rat

hypocampus

Mulitplexed, real-time

RT-PCR

Quantification of 20 different

mRNAs

Human small intestine cells

GFP: Green fluorescent protein; CE: Capillary electrophoresis; MALDI-MS: matrix assisted laser

desorption/ionization mass spectrometry; LCM: laser capture microdissection; RT-PCR: reverse transcriptase

polymerase chain reaction

Table 1.1: Examples of single cell analysis techniques useful to investigate heterogeneity and the cell systems

investigated by various researchers (Adapted from [13])

1.2.1 Flow cytometry

Flow cytometry is a technique based on the principles of light scattering, light excitation and

emission of fluorescence. Cells or particles, ranging in sizes from 0.2 - 150µm, are passed in

single file by hydrodynamic focusing through a laser beam for sequential illumination. Scattering

parameters yield morphological information and can either be based on forward scatter, which is

dependent on cell volume, or side scatter, which correlates to inner complexities such as

membrane roughness, shape of the nucleus and cytoplasmic content. Meanwhile, depending on

the target, fluorescence provides functional and structural information, while affording the ability

to count, and select sub-populations of cells by fluorescence-activated cell sorting. Flow

cytometry analyses tens of thousands of cells per second and the information obtained allows a

statistical analysis of the population based on each cell or particle. Although flow cytometry

offers extremely high throughput and analysis of parameters such as total DNA or RNA content,

enzymatic activity, pH, cell viability and a long and constantly expanding list of other

parameters, it is limited due to its physical configuration, making it difficult to couple with the

subsequent steps for chemical cytometry [14]. Furthermore, cells remain in suspension for long

periods prior to detection, disrupting the biochemical balance between neighbouring cells, which

may alter cellular processes such as transcription and translation.

8

1.2.2 Fluorescence microscopy

Fluorescence microscopy is the quintessential imaging technique used in cellular and molecular

biology because of its intrinsic selectivity and specific detection of molecules at small

concentrations with good signal-to-background ratio. More than 3000 fluorescent probes exist to

label virtually any imaginable aspect of a biological system, while the large spectral range of

fluorophores allows simultaneous imaging of different cellular, subcellular and molecular

component [15]. This technique allows protein and gene expression to be measured as a function

of spatial position within a sample, yielding expression gradients, for example, for various types

of VEGF [16]. Additionally, the development of genetically encoded fluorophores, particularly

GFP and its variants, has allowed protein components of living systems to be genetically tagged,

making it possible to study protein-protein interactions and monitoring signalling events in living

cells [17]. The major caveat of fluorescence microscopy is its very low throughput and limitation

to a small number of fluorescence colours (N ~ 3), even when using advanced microscopy

techniques such as confocal and two-photon; however, this problem can be overcome with the

integration of hyperspectral imaging techniques [18]. Like flow cytometry, this technique is

constrained by the requirement of having apriori knowledge of the genes or proteins of interest,

and the availability of a suitable fluorophore.

1.2.3 Capillary electrophoresis

Capillary electrophoresis is the conventional tool used to perform high efficiency differential

transportation and separation of large and small molecules from single cells based on the

molecule‟s size, charge and hydrophobicity. The technique employs narrow bore capillaries (2-

200 µm inner diameter) in which separations are facilitated by the use of high voltages,

generating electroosmotic and electrophoretic flow of conductive buffer solutions and ionic

species respectively. Separation properties and the resultant electropherogram have characteristic

similarities to traditional polyacrylamide gel electrophoresis (PAGE) and modern high

performance liquid chromatography (HPLC) [19]. Capillary electrophoresis has the ability to

analyze the contents of single cells when capillaries of small inner diameters (2-10µm) are used,

allowing injection volumes in the pico and possibly femtoliter range. Furthermore, as first

demonstrated by Huang et al [20], the development of capillary array electrophoresis has given

rise to increased throughput by this technique while coupled capillaries with different separation

9

properties enable 2-D analysis of protein contents from single cells [21]. Unfortunately, run

times for high resolution separation can reach hours, thus reducing the throughput even if an

array of capillaries is used.

1.2.4 Requirements for alternative analysis method

The shortcomings identified above highlight the need for an analytical tool that has the ability to

address the following issues:

• selection of a priori identified cells from an ensemble,

• permit subcellular localization,

• increase the number of analytes from a single cell,

• enable high throughput to generate population statistics.

1.3 Single cell analysis on microfluidic platforms

The need for an ideal analytical tool capable of investigating a large number of parameters from

each cell within a population to identify variation, while concurrently having high throughput to

allow significant statistical analysis, has led to greater research efforts in the realm of

microfluidics.

Microfluidics, or lab-on-a-chip, is the technology of controlling and manipulating fluids

on the order of picolitres to microlitres using microchannels that are between 1-500µm in size.

Consequently, because of their small size and small volume, these devices are ideal for the

confinement and subsequent analysis of either whole or lysed single cells, making novel

experimentation a possibility while providing sophisticated and well controlled environments for

cellular investigation.

Devices are fabricated using MEMS techniques, mimicking well-defined processes in the

microelectronics industry and have typically been developed in glass and quartz, whose

transparent properties are ideal for optical analysis. However, polymers have become the

materials of choice for biological experimentation in the last 5 years [22]. Because a significant

number of devices are fabricated to meet a particular requirement, a plethora of additional

features have been integrated within the microfluidic environment to enable selection,

10

confinement, and lysis, of single cells. Examples of these include pumps and valves [23], optical

tweezers [24], dielectrophoretic traps [25], microdroplets [26], and electrodes [27, 28].

The success of this technology is based upon its enhanced analytical performance,

allowing fast, highly sensitive and reproducible analysis, while requiring low consumption of

chemicals and energy, thus making it less expensive at shorter processing times [22]. The ability

to perform parallel analysis or successive operations on the same device, sample manipulation,

reduced loss of analytes and contamination, and the possibility of high throughput is making

microfluidics a popular avenue to investigate cellular heterogeneity. Many of the traditional

techniques, discussed in Sections 1.2.1-1.2.3, have also been translated onto microfluidic devices

and will be described briefly.

1.3.1 Techniques for single cell analysis in microfluidics

Flow cytometry

The emergence of optofluidics, where microfluidics and photonics merge, has allowed the

integration of polymer waveguides and lenses for the purposes of excitation, focusing and

detection of light within the microchip, significantly alleviating the problems posed by micro-

macro interfacing as well as the interface with the user. As a result, numerous variations of

microchip based flow cytometers have been developed for sorting and analysis of single cells

and particles, ranging in size from 1-20µm [29, 30]. While the laminar flow conditions within

microfluidic systems reduces perturbations to cell physiology during sorting of viable cells,

microflow cytometers, with sorting speeds up to 500 particles·s-1

[30], have yet to match

conventional systems in performance.

Fluorescence microscopy

The same principles of fluorescence microscopy in macro devices for single cell analysis,

described in Section 1.2.2, apply within microfluidic systems. However, while microfluidic

systems have achieved high levels of integration with pumps and valves, electrodes and

waveguides, after sample preparation and processing within the microfluidic device, there is still

a need for fluorescence imaging and detection. Consequently, off-chip bulk optical elements

11

such as lenses and microscopes are common requirements for fluorescent microscopy in

microfluidics [24].

Capillary electrophoresis

Due to the narrow fluidic channels and confinement available in microfluidic devices, capillary

electrophoresis has been one of the most successfully translated and widely used macro

technologies on a microfluidic platform. The small chemical and energy footprints coupled with

the ability to fabricate parallel channels, allowing simultaneous separation and high throughput,

are desired features for researchers. Additionally, the electric field required to enable efficient

separation and electroosmotic flow can be achieved in microfluidic devices by using

significantly lower applied voltages as the length of the separation channels are shorter [31],

which also enable faster separation and shorter run times. The major limitation of capillary

electrophoresis in microfluidics is the detection of analytes, specifically when parallel separation

channels are used. Traditionally, a fluorescence microscope has been sufficient to detect analytes

in up to 8 parallel channels [24]; however more sensitive optical detection systems using

cylindrical optics have resulted in better sensitivity in detecting smaller molecules [32].

1.3.2 Microfluidics platform for multiple technique integration

Microfluidic technology offers an effective method of integrating multiple components to select,

manipulate, lyse and analyse single cells within a confined and well controlled fluidic

environment. Many of the macro techniques used for single cell analysis have been translated to

microfluidic platforms with comparable or better results. The overall aim of this program is to

develop a functional microfluidic device capable of selecting single cells from a population of

cells using optical tweezers [24], load individual cells into parallel channels, perform selective

lysis of the plasma membrane and other intracellular organelles, and use capillary electrophoresis

to separate the components of different fractions of the cell in sequential analytic steps while

using a high numerical aperture fiber-optic array for multiple single point detection (Fig. 1.3).

This thesis will be focused on developing a microfluidic device with integrated electrodes to

perform selective electrical lysis of the plasma membrane while demonstrating intactness of the

nuclear membrane, which is to be ruptured in a subsequent step.

12

Figure 1.3: Integrated microfluidic device for single cell transport, lysis, capillary electrophoresis and detection

(Courtesy of Luc Charron)

1.4 Motivation for selective single cell lysis

Currently, even on a microfluidic platform, single cell analytical techniques are unable to

separate the analysis of the various compartments of the cell, thus sequential lysis can be a low

resolution alternative to on chip microscopy. As a result of whole cell lysis, protein expressions

can only be measured for the entire single cell, whereas for signalling studies, the challenge is to

distinctly identify from what organelle within a cell a particular protein or molecule originated.

Gaining the ability to spatially and temporally determine the translocation of proteins and

molecules between cellular compartments can significantly increase the wealth of knowledge on

cellular functions, specifically understanding signalling pathways and the effects of

environmental factors on these pathways and cellular response. The main caveat of single cell

analysis is that it is impossible to repeat experiments on that particular cell, but with the ability to

perform analysis on different parts of the cell separately, the potential exists to increase the

information content from a single cell while using only a single fluorescent probe, quantifying a

protein of interest expressed at various sites or in various compartments.

Lysis regions

13

1.5 Thesis organization

This thesis presents an argument in support of single cell analysis to investigate heterogeneity

among cells and the use of microfluidics as a tool to perform analysis. In Chapter 2, a discussion

of the advantages of electrical lysis, the physical mechanisms involved in this process, and a

numerical model of the anticipated electric field is presented. The aim is to determine the applied

voltage required to induce lysis while quantifying the thermal effects of such a process on the

cell of interest. Chapter 3 details the fabrication and optimization of a microfluidic device and

the integration of electrodes. Chapter 4 describes the in vitro experiments performed to

demonstrate selective lysis of the plasma membrane. Finally, Chapter 5 concludes with a

summary of each chapter, highlighting the main findings in each, as well as outlining potential

future research.

14

Chapter 2 Electrical lysis of plasma membrane with intact nuclei

2.1 Introduction

2.1.1 The plasma membrane and electroporation

Plasma membrane

The phospholipid bilayer membrane, or plasma membrane, of a cell comprises an aqueous

solution, sandwiched by two fatty acid monolayers which are polar and hydrophilic (Fig. 2.1).

The inherently high electrical resistance (~104Ω [33]) of the membrane, results in it acting as a

dielectric barrier between the conductive intracellular, and extracellular aqueous environments,

which differ in osmolarity and ionic concentration. Consequently, a resting potential, typically

ranging from 60mV to 110mV [34, 35], exists across the plasma membrane and the system is

commonly modeled as a parallel plate capacitor [33, 36], with the membrane as a dielectric (Fig.

2.2)

Figure 2.1: Illustration of phospholipid bilayer membrane (Adapted from [37])

15

Figure 2.2: Equivalent circuit representation of cell in suspension (Adapted from [33])

Electroporation

In the presence of an external electric field, the membrane is polarized and dipoles are formed

either within or at the interfaces of the membrane and aqueous environments, inducing a

transmembrane potential, ΔVm, which leads to electroporation. This process relies on the weak

nature of the hydrophobic-hydrophilic interactions in the phospholipid bilayer. Several

theoretical models exist to explain the process of electroporation; however, the most commonly

used is the transient aqueous pore mechanism hypothesis proposed by Weaver et al [38] and

expanded by others [35, 39], in which a pulsed external electric field rapidly rearranges the

localized structures, polarizing the membrane and increasing its electrical conductivity while

inducing thermal fluctuations. As a result, hydrophobic pores appear randomly on the surface of

the membrane, and transition under the stress of the transmembrane potential, becoming aqueous

pathways, or reversible hydrophilic pores, typically at a threshold ΔVmem of 0.5-1.2V. For a

spherical cell, assumed to have a membrane that is a pure dielectric, ΔVmem, can be obtained by

Equation 2.1 when under the influence of a DC field, and Equation 2.2 for an AC field [35, 40].

Where:

RE – Resistance of extracellular medium

[Ω]

CE – Capacitance of extracellular

medium [F]

RC – Resistance of cytoplasm [Ω]

CM – Capacitance of cell membrane [F]

16

(

)

(

)

(

⁄ )

Where:

E is the value of the electric field in the extracellular environment [V·m-1

],

r is the radius of the cell [m],

θ is the polar angle measured between the centre of the cell and the direction of the electric

field [0],

t is the time/pulse duration of the electric field [s],

f is the frequency of the applied AC field [Hz],

τc is the charging time constant of the plasma membrane [s-1

],

Cmem is the capacitance of the plasma membrane per unit area [F],

ρi is the electrical resistivity of the intracellular environment, particularly the cytoplasm

[Ω·m],

ρe is the electrical resistivity of the extracellular environment [Ω·m],

Figure 2.3: Polarization of plasma membrane under the influence of an electric field

17

The expression for the time constant, τc, is obtained by modelling the system as a parallel plate

capacitor (Fig. 2.2) surrounded by two layers of extracellular environment. Based on accepted

values for Cmem, ρi, and ρe [33, 41], τc is usually ~ 100ns, thus in cases where the pulse duration

is much greater than the charging time of the plasma membrane (t >> τc), Equations 2.1 and 2.2

can be reduced to Equations 2.4 and 2.5 respectively.

An extension of this theory, following Equations 2.4 and 2.5, suggests that intracellular

organelles, such as the nucleus and mitochondria, have a unique transorganelle membrane

potential, which can be exploited to permit electroporation of organelles, without affecting the

integrity of the plasma membrane [28]. This is due entirely to their smaller radii when

considering a DC field, while the radii, membrane capacitance and charging time constant are

important factors when an AC field is applied. Under the influence of an AC field, selective

electroporation of intracellular organelles is only possible using ultrashort pulse durations (~ns),

which prevent the charging time of the plasma membrane to be reached (t < τc), coupled with

very high applied voltages to create a larger electric field across the cell. Equation 2.6 and 2.7 are

modifications of 2.4 and 2.5 respectively, and determines the transorganelle membrane potential,

where subscripts org represents within or of the organelle, and int, represents intracellular.

√ ( )

(

⁄ )

18

Applications of electroporation

Common applications of electroporation are focused on the creation of a few holes at the cells

equator, relative to the electric field (θ = 00), to enable transport into the cell. Whereas pores on

the plasma membrane are rendered open in microseconds, resealing of the pores occurs over a

range of a few minutes and during these times, foreign molecules, DNA and drugs, among

others, can be introduced to the target cell. The most common use of electroporation is DNA

transfection whereby a specific gene is introduced to the host cell via a plasmid in order to

investigate a particular function or structure. Similarly, plasmids can be transferred between cells

incubated together to exchange desirable features when pores are open. Clinically, due to the

stability of DNA, vectors containing genes can be delivered during gene therapy to treat a

genetic disorder or to replace a defective gene. Prausnitz et al [42] showed that electroporation

can be effective in transdermal drug delivery by forming pores in the stratum corneum – the

outermost layer of epidermis, which then allow drugs to reach a target tissue. This method has

also been extended to cancer tumour electrochemotherapy where disruption of the tumour cell

membrane increases the amount of drug than can be delivered.

2.1.2 Electrical lysis

Selective lysis by direct and alternating current sources

The work in this thesis extends the mechanism of reversible electroporation, in which membrane

pores are re-sealable, to a regime of irreversible electroporation, where pores become too large to

be re-sealed, referred to as electrical lysis. The plasma membrane is permanently ruptured when

an applied voltage creates an external electric field across the membrane, inducing a

transmembrane potential in excess of the threshold value, ΔVmem. By exploiting the differences

in size and electrical properties mentioned above, a specific electrical pulse can lyse the plasma

membrane with minimal impact on intracellular organelles.

An electric field produced by a direct current (DC) source is the simplest and most

obvious choice for selective lysis, since the subcellular organelles would experience no

transorganelle membrane potential, while the plasma membrane experiences a large

transmembrane potential. However, while DC sources have been used predominantly for cell

lysis, specifically in microfluidic devices for single cell lysis [24, 43, 44], the generation of a

large, continuous electric field requires the application of a high applied voltage. As a result, the

19

water electrolysis threshold (~ 1V) is usually exceeded, leading to the formation of bubbles and a

change in pH near the electrodes which may interfere with subsequent sampling processes. Using

an alternating current (AC) source to generate the electric field would minimize the effects of

water hydrolysis; however, Lu et al [28] have shown that an optimal frequency, in the range 1-

100 kHz (Fig. 2.4), must be reached to be able to perform selective lysis. Consequently, to avoid

the negative effects of using a DC source, and the requirement to find an optimal frequency with

an AC source, a pulsed DC source will be used in this project.

Figure 2.4: Modeled results for ΔVmem and ΔVorg, showing optimal frequency region (Adapted from [28])

Comparison of lysis techniques

Several alternative techniques exist to perform lysis on single cells, each with unique advantages

and disadvantages. The most widely used method for bulk assays which translates well to single

cells, is the introduction of a chemical detergent which solubilizes lipids and proteins in the

plasma membrane, creating pores which leads to complete lysis. Two common detergents, Triton

X-100 and SDS, each exhibit different lysis capabilities, with the former typically inducing lysis

in ~30s while preserving enzyme activity [45]; whereas, the latter usually results in faster lysing

(< 2s) but denatures membrane and cellular proteins [32]. Denatured proteins are typically

unfavourable as they quickly aggregate, forming an insoluble, randomly organized structure.

While chemical lysis does not require specialized equipment apart from a mixing method, and

various detergents exist to enable selective lysis, it is evident that the detergents can significantly

impact the outcome of the experiment due to long times to lyse, leading to excessive diffusion of

cellular content. Detergents also tend to denature and break up protein complexes, while also

adding an additional reagent that may eventually need to be removed from the cellular analytes

prior to a specific assay.

20

Optical lysis techniques also exist where a pulsed (~ns) laser microbeam generates a

shockwave in the vicinity of the cell, followed by the formation of a cavitation bubble which,

upon expansion or collapsing, ruptures the plasma membrane. The lysing speed in this method is

dependent on the position of the focal point of the laser pulse and can have a range of 1 – 400µs

[46], which allows cellular content to be released quickly instead of over a lengthy period,

making this method ideal for studying highly dynamic cellular processes. While there is no

literature available on the use of this method to selectively lyse a membrane, when leaving other

organelles intact, it is theoretically possible by adjusting the pulse duration of the beam and

numerical aperture of the objective used. By employing femtosecond pulses at a high repetition

rate, instead of single nanosecond pulses, the energy deposited to the cell can be significantly

reduced, enabling selective membrane lysis.

Single cell lysis has also been demonstrated by mechanical and acoustical methods. The

former subjects a cell to a physical force, such as compression within a confined region [47],

where the mechanical stress results in membrane rupture, or to sharp, physical structures which

inhibit a cell‟s path and punctures the membrane in a uncontrolled manner [48]. The

compression method, while capable of producing fast (~ms), complete lysis, results in a non-

uniform diffusion of cellular content, and the possibility of cellular debris adhering to parts of the

compression region. Similarly, mechanical lysis against physical structures typically results in

cellular debris sticking to the physical structures and poor diffusion due to incomplete membrane

rupture. Additionally, neither of these mechanisms lends itself to selective lysis.

Acoustical lysis by sonication utilizes ultrasonic waves to shear a cell by generating

cavitation in high pressure areas. The major limitation of this method is the long time required

for complete lysis (3 – 50s), which can result in thermal damage to the cell over extended times

and excessive diffusion of cell contents [49]. Furthermore, localization of the ultrasonic wave to

lyse a single cell poses an engineering challenge and selective lysis of a cell would require

significant refinement of current techniques.

Based on the limitations of other lysis techniques, and their incompatibility with other

aspects of the microfluidics program, electrical lysis was chosen as the best possible technique

for this project as there is strong theoretical evidence supporting selective lysis of the plasma

membrane in a controlled manner due to the cosθ dependency (Equation 2.4), while other

organelles remain unharmed. Additionally, electrical lysis can typically occur within

21

milliseconds after the application of a pulse and with adequate confinement, diffusion of cell

contents can be made relatively uniform, while denaturation of proteins is usually not evident.

Electrical lysis in microfluidic devices

Electrical lysis in the confines of a microfluidic device is advantageous over macroscopic

systems primarily by decreasing the voltage requirements to perform lysis, due to the

significantly reduced inter-electrode distance, d, as can be generally inferred by Equation 2.9,

where E is the electric field, and Vapp, the applied voltage. Additionally, with the selection of

appropriate materials for device fabrication, as will be discussed in Chapter 3, heat dissipation is

minimal, thus reducing thermal effects on the plasma membrane. However, the design of the

electrodes is also critical to the shape and homogeneity of the electric field produced, along with

the effects of electrode – membrane interaction.

Many researchers have fabricated devices in which the electric field used for lysis

depends on the media through which the cells are flowing [24, 43, 44, 50], with the field existing

within the fluidic medium. While this is a straightforward approach, requiring simple fabrication

methods and possible geometric variation in the fluidic channels, the applied voltage is relatively

high due to large inter-electrode distances (~1cm).

Electrodes have also been designed in a 2-D manner, where a cell is positioned directly

on top of an electrode, while the second one is located above the cell [51, 52]. The integration

and positioning of these electrodes directly above a cell poses an engineering challenge and in

cases where the upper electrode is a capillary tip [52], creates a heterogeneous electric field,

having a larger field strength on the upper surface than the bottom of the cell, leading to greater

pore creation on this portion of the plasma membrane. As a result, diffusion of the cytosol in the

channel may be unpredictable. The possibility exists also that a cell being positioned directly on

top of an electrode may lead to direct charge transfer and hence heating of the membrane. Due to

these factors and the incompatibility of 2-D electrodes with other components of the overall

microfluidics program, 3-D electrodes were explored for this project.

3-D electrodes offer an improvement on the quality and homogeneity of the electric field;

however, they are often integrated directly into fluidic channels [28, 53, 54], which pose a

22

problem due to contact with the plasma membrane. Thermal effects, due to Joule heating of the

cell, would adversely affect lysis, as it would be difficult to determine whether damage to the

membrane is attributed to electrical or thermal mechanisms. In Joule heating, the cell, having

high resistance, effectively becomes part of the electrode circuit, and electric current passing

through it is dissipated as heat, which may also lead to denaturation of analytes of interest within

the cell. This chapter aims to simulate a common 3-D electrode design and the intended design

for this project to determine the best possible solution to create a homogenous electric field in a

confined region while minimizing thermal effects on the plasma membrane. Determination of the

required applied voltage to produce a suitable lysing electric field will also be investigated.

Figure 2.5: Illustrations of various electrode configurations used in microfluidics research (Images adapted from

[55]). Designs used by: (a) [24, 43, 44, 50], (b) [51], (c) [28, 53]

2.2 Materials and Methods

2.2.1 Electric field simulations

To induce transmembrane potentials ranging from 0.5-1.2V, an estimation of the minimum

electric field required to produce electroporation (θ = 00) of the plasma membrane of a cell with

diameter ranging from 8-12µm can be obtained from Equation 2.4. This yields fields in the range

0.33-1kV·cm-1

at the poles of the cell closest to the electrodes and was used as a minimum basis

in simulations.

Estimations of the electric field distribution produced by different applied voltages were

numerically simulated by finite element methods using COMSOL Multiphysics 3.4 (COMSOL

Inc., Burlington, MA, USA). The software‟s conductive media DC model was used for static

state situations and solves Maxwell‟s differential equations for 3-D electrode geometries as

(a) (b) (c)

23

previously described (Section 2.1.2). Additionally, the simulations were able to determine the

effects of electrode and channel geometry on the electric field distribution, while as a post

processing feature of COMSOL, the thermal effects of the electrodes were also obtained within

the region between the two electrodes.

Simulations were initially performed using typical channel geometries employed by other

researchers for 3-D electrodes [28, 53], and the intended channel geometry to be used for this

project (Fig. 2.6), which will be discussed in detail in Chapter 3. Regions of the microfluidic

device in which the electrodes were embedded, were modeled as known electrical insulators

traditionally used to fabricate devices, while the electrical properties of electrodes were modeled

similarly to that of ITO, a common electrode material in microfluidics. The electrical properties

of alpha MEM, which will be used as the extracellular medium for in vitro studies, was also

taken into consideration for all models. The discretization sizes for numerical modeling were

chosen to be 1-2µm, which was smaller than the smallest structures of the system, while the

material properties are indicated in Fig. 2.6 for the intended geometry for this project.

Figure 2.6: Geometric simulation of intended microfluidic chip including material properties

24

2.3 Results and Discussion

2.3.1 Electric field distribution dependency on geometry

Teeth-like electrode structures

3-D electrodes having a tooth-like structure demonstrate the ability to create a homogenous

electric field directly between the tips of the electrode as shown in Fig. 2.7 (a) when a small,

arbitrary voltage of 4V is applied. However, for effective electrical lysis to occur, cells would be

required to be directly between a pair of tips, since the region between teeth does not

demonstrate a homogenous field. While lysis may occur when a cell is anywhere within the

region of the teeth, different parts of the plasma membrane would experience different field

strengths, thus making the lysis process unpredictable, as expected by Equation 2.4.

Additionally, an electric field of this nature does not lend itself to selective lysis due to its overall

non-homogeneity. In Fig. 2.7 (b), simulations show that cells passing through the teeth-like

electrode structure also experience a significantly high resistive heating due to direct transfer of

charge carriers through the cell as there is no insulating structure between the cells and the

electrodes.

(a)

25

Figure 2.7: a) Simulation of electric field distribution between teeth-like electrodes; b) Resistive heating between

electrodes. Tip-tip distance in x-axis = 40µm, in y-axis = 15 µm

Tapered fluidic channels with electrodes embedded into microfluidic structure

Simulations reveal that the electric field distribution within the microfluidic channel in the region

between the embedded electrodes is homogenous throughout the height, width and length of the

channel, as demonstrated by multiple slices (Fig. 2.8 (a)), while arrows in the simulation also

indicate the direction of the electric field. The evidence provided by these simulations of the

intended microfluidic design indicate that the electric field distribution is related to the geometric

and structural design of the electrodes and microfluidic channel, while also confirming that the

choice of tapered channels will not negatively impact the quality of the electric field.

Furthermore, Fig. 2.8 (b) shows that the resistive heating in the region of the microfluidic

channel close to the electrodes is significantly lower when a voltage of 4V is applied compared

to the teeth-like electrode model. This is most likely due to the thermally insulating nature of the

material in which the electrodes are embedded, in this case, a common fabrication material in

microfluidics, PDMS, having thermal conductivity of 0.18W·m-1

·K-1

[56].

(b)

26

Figure 2.8: a) Simulation of electric field distribution between embedded 3-D electrodes, with arrows shown in

insulating region between electrodes and channel; b) Resistive heating between electrodes. Inter-electrode x-axis

distance = 55µm, region of interest of channel in y-axis = 150µm, channel height = 20µm.

(a)

(b)

27

2.3.2 Electric field dependency on applied voltage

While the colour map simulations in Fig. 2.8 show maximum and minimum values of the electric

field distribution in the fluidic channel, the electric field along a line of interest, drawn along the

x-axis from the y-axis point of one electrode face nearest to the narrowing point of the channel to

the opposite electrode face through the z-midpoint of the channel, yielded a better approximation

of the electric field within the channel (Fig. 2.9).

Figure 2.9: Electric field strength as a function of distance in x-axis, showing regions of the fluidic channel and

insulating regions at an applied voltage of 4V

The common method of determining the electric field across a microfluidic channel is to use

Equation 2.9 [46], which would yield a field of 0.727 kV·cm-1

. However, this method is unable

to consider the effects of the properties of the insulating regions between each electrode and the

channel, along with the electrical properties of the extracellular media which would fill the

channel. The numerical simulation considers all aspects of the system and therefore yields a

better approximation of the electric field, 0.563 – 0.609 kV·cm-1

, across the width of the fluidic

channel as extracted from Fig. 2.9 when 4V is applied across the electrodes. Table 2.1 coupled

with Fig. 2.10 summarize the variation of the range of the electric field within the fluidic channel

as a function of applied voltage.

Fluidic channel

Insulating region Insulating region

28

Applied

Voltage [V]

Minimum

Electric Field

[kV·cm-1

]

Maximum

Electric Field

[kV·cm-1

]

Range, [kV·cm-1

]

4 0.563 0.609 0.046

8 1.126 1.219 0.093

12 1.689 1.828 0.139

16 2.252 2.437 0.185

20 2.815 3.046 0.231

24 3.377 3.656 0.279

28 3.940 4.265 0.325

32 4.507 4.532 0.025

36 5.439 5.473 0.034

40 6.043 6.093 0.050

44 6.592 6.702 0.110

48 7.252 7.311 0.059

Table 2.1: Dependency of electric field range on applied voltage

29

Figure 2.10: Plot of electric field dependency on applied voltage as obtained from COMSOL simulations

Based on the numerically approximated values in Table 2.1, the applied voltage chosen for the in

vitro studies in the project was 32V since it yields the least variation of the electric field across

the width of the channel and exceeds the minimum required field (0.33-1kV·cm-1

) at the poles of

the cell closest to the electrodes to initiate irreversible electroporation. Furthermore, by Equation

2.4, the field produced by this applied voltage is capable of inducing transmembrane potentials

greater than the required threshold for lysis at positions on the plasma membrane further away

from the electrodes, up to ~ θ = 820. This results in pore creation on ~90% of the cell‟s surface,

assuming the cell to be spherical. When considering nuclear membrane integrity, the electric

field generated by this applied voltage, would generate a maximum transorganelle potential of ~

0.9V on a 2µm sized nucleus, which is insufficient to cause rupture.

2.3.3 Determination of heat transfer to cell during lysis

Modeling the system as a capacitor

While COMSOL simulations have determined there is minimal resistive heating to the region of

the channel between the electrodes, if the electrode – insulator – microchannel system is

considered as a parallel plate capacitor (Fig. 2.11), with each region between the electrodes a

different dielectric material, then the amount of energy deposited to a cell in that region can be

30

determined. This is only possible if the following assumptions are made:

i) the extracellular media is comprised primarily of water,

ii) all of the energy required to charge the capacitor is transferred to a cell as heat,

iii) the cell is comprised primarily of water.

Figure 2.11: Parallel plate capacitor model of electrode – insulating region (C1 and C3) – channel system (C2),

showing insulating regions and channels as capacitors, with corresponding thicknesses

The capacitance, C, of each section can be calculated using Equation 2.10, where A is the

surface area of the capacitor plate, ε0 is the permittivity of free space (8.854 x 10-12

F·m-1

), εr is

the relative permittivity of the dielectric of interest, and d is the thickness of the dielectric. Table

2.2 summarizes the values of the capacitance for each section, assuming C1 and C3 to be PDMS,

the intended fabrication material for this project, and C2 to be water at 200C. Meanwhile, the

work done in charging the capacitor, W, is given by Equation 2.11, where CT is the total

capacitance and Vapp is the applied voltage across the plates.

C3 + - C1 C2

d1 = 20µm d2 = 15µm d3 = 20µm

31

Material Relative

permittivity,

εr

Surface area (A) of

electrode (150µm x

20µm) [m2]

Thickness (d)

[m]

Capacitance [F]

C1 2.67 3 x 10-9

20 x 10-6

3.54 x 10-15

C2 80.1 3 x 10-9

15 x 10-6

1.42 x 10-13

C3 2.67 3 x 10-9

20 x 10-6

3.54 x 10-15

Table 2.2: Calculated values of the capacitance of each dielectric layer

The total capacitance is 1.75 x 10-15

F and with an applied voltage of 32V, Equation 2.11

determines the charging energy to be 8.96 x 10-13

J. Based on assumption (ii), Equation 2.12

calculates the change of temperature, ΔT, of a cell within the region of the microchannel between

the electrodes to be 2.14 x 10-4

K, when the mass of the cell, m, is assumed to be 1ng and its

specific heat capacity, c, to be 4186 J·kg-1

·K-1

. This estimation confirms that when the system is

modeled as a parallel plate capacitor, even in an extreme case where all of the energy stored in

the capacitor is transferred to the cell, the corresponding change in temperature is negligible.

Consequently, the electric field required to perform plasma membrane lysis is not expected to

produce thermal effects in the cell such as protein denaturation.

[ ]

Modeling the cell as a resistor

Simulations have shown that due to the insulating nature of the material separating the electrodes

from the microchannel, there is negligible resistive heating; however, the intended material is a

dielectric and has a breakdown voltage of 21.2V·µm-1

[56]. As a result, the extracellular media

and the cell can become a part of the current carrying circuit when the breakdown voltage is

exceeded, which would require a voltage of 424V since the material in the electrode to channel

gap is modeled as 20µm. Equation 2.13 is a combination of Joule‟s law and Ohm‟s law and

32

determines the energy, Ec, dissipated to a cell, of resistance, R, in time, t, when a current, I,

passes through the cell.

Assuming a short time duration of 100µs as commonly used in electroporation, an arbitrary

current of 5mA, and cell resistance of ~ 1 x 104Ω, the energy dissipated is calculated to be 2.5 x

10-5

J, which would result in a temperature increase of 5972.3K. While this value is extremely

high, it is based on the assumption that all of the energy dissipated to the cell is transformed into

heat and that the system is 100% efficient. Since the applied voltages anticipated to be used in

this project do not approach the breakdown voltage of the dielectric, the system is not expected

to act as a complete current carrying circuit, thus the cell would not experience any resistive

heating. Additionally, pulse durations of 100µs to be used in this project are much greater than

the charging time of the plasma membrane (~100ns), and are yet small enough to prevent a large

amount of energy to be deposited to the cell.

2.4 Conclusion

This chapter has explored the theoretical basis for selective lysis of the plasma membrane via

electrical lysis and has compared this method to other current techniques available for single cell

lysis. By gaining an understanding of electrical lysis by both DC and AC electric fields and

weighing their pros and cons, a DC electric field was chosen for all in vitro work, to be discussed

in Chapter 4. COMSOL simulations provided a numerical approximation of the electric field

distribution within a microfluidic channel due to an applied voltage across a pair of electrodes,

while also demonstrating the heterogeneity and homogeneity of the electric field based on

commonly used 3-D electrode structures, and those intended for use in this project. Furthermore,

the simulations show that at an applied voltage of 32V, the electric field produced is large

enough to surpass the threshold transmembrane potential required for lysis, and there is minimal

variation of the field across the width of the channel. Consequently, this applied voltage, with a

pulse duration of 100µs, which is greater than the required charging time for the plasma

membrane, will be used for all in vitro studies in this project.

33

Chapter 3 Fabrication of microfluidics device with integrated electrodes for

single cell lysis

3.1 Introduction

3.1.1 Design requirements of a single cell microfluidic device

Channel geometry for cell confinement

Microfluidic devices designed and developed for the purpose of single cell analysis should

ensure that the fluidic channels are slightly wider than the diameter of the cells of interest to

assist in the confinement of cell movement to one direction along the channel. Additionally, in

regions where cells may be interrogated and their contents analysed, further narrowing of the

fluidic channel would encourage analyte flow along the channel and reduce diffusion laterally

within the channel.

There exists an abundance of microfluidic devices designed for single cell capture,

ranging from microfabricated physical cell traps situated both perpendicular to and lying on the

base of the fluidic channel [57, 58], micro-pillars standing vertically in the channel [54],

complex systems of pneumatic pumps and valves that control cell positioning within channels

[23], tapered fluidic channels that confine cell movement and act as a trap [24], dielectrophoretic

traps [25], and optical tweezers [24]. The challenge posed to the former devices is the ability to

confine analytes and limit diffusion after the cell has been lysed or interrogated in some way, due

to the fabrication limits of the device. Meanwhile, tapered channels limit the flow of the buffer

solution adjacent to the cell and limits transverse diffusion of analytes while promoting flow

longitudinally. This chapter will present a tapered channel geometry with the inclusion of a

region where cell lysis and future analyte separation will occur, referred to as the injector

structure.

Integration of electrodes

For the purposes of electrical lysis, integrated electrodes in a microfluidic device are required to

either pass a direct current across the cell by being in contact with the cell [54], or by creating an

electric field in the vicinity of a cell [27]. As outlined in Chapter 2, integrated electrodes are

34

advantageous if designed to be 3-D structures [28, 53] to create an electric field which interacts

with a larger cell surface as opposed to electrodes which are 2-D producing fields that only

impact one part of a cell [27]. Most 2-D electrodes are fabricated by a deposition process or

excimer laser mediated cutting as demonstrated by Xu et al [59] using ITO and other similar

materials, while 3-D electrodes usually require a complex fabrication method to place platinum

or titanium wires, or photolithographic development of chromium and silver electrodes onto

glass [54]. In this chapter, a novel method of integrating 3-D microelectrodes in close proximity

to fluidic channels and capable of extending a homogenous field over a larger cell surface of a

single cell will be demonstrated.

3.1.2 UV photolithography as a fabrication tool

The process of photolithography entails the selective removal of parts of a thin photo sensitive

film, referred to as a photo resist, by transferring desired patterns from a photo mask using light

of a specific wavelength. Prior to exposure, the resist is distinguished as being either positive or

negative, depending on the way it is developed following irradiation, with the former becoming

insoluble in photopolymerized regions, and the latter soluble where exposed. Viscosity of the

resist is a significant determining factor of film thickness, which in turn determines the aspect

ratio, referring to the ratio of the width to height of the structures being fabricated [60].

This method has been used for over a decade as a fabrication tool for microfluidic

devices [61], with the resist being deposited on a variety of substrates including glass [62],

silicon wafers [63], and metals [64]. While the latter two have shown good adhesion and thermal

expansion coefficient match to photo resists, they are significantly more expensive, while glass

can be treated to improve its substrate qualities and is less expensive. In the same respect, a

plastic photo mask is often preferred [63] since it is cheaper and allows more masks to be created

and designs to be implemented for the equivalent cost of having a single metal mask, typically

quartz-chrome, designed for one geometric pattern [65]. However, plastic masks, produced by a

printing process, are unable to achieve the same spatial resolution and fine details as metal masks

which are produced by laser writing.

Following photolithography, structures can either be used directly as microfluidic devices

[66] or in a soft lithography process in which another material, commonly PDMS, is casted onto

the photo resist to create an inverse of the structures [63]. PDMS, while naturally hydrophobic,

35

has demonstrated good biological compatibility due to it being non-toxic [67, 68], structurally

flexible, inert, allows for rapid prototyping [63], and consequently has become the most

commonly used material in which microfluidic devices are fabricated.

Other fabrication tools exist to create microfluidic devices, including laser

micromachining [24], hot embossing [69] and direct write laser photolithography [70] among

others; however, while the latter of these methods offer better resolution than ordinary

photolithography, they are expensive, time consuming and for the purposes of this project,

incompatible for the overall program. This chapter reports on an improvement to the standard

photolithography procedures yielding minimum structure sizes of 5µm using a plastic photo

mask and glass substrate.

3.2 Materials and Methods

3.2.1 Photo mask production and optimization

Mask lay-out as produced by computer-aided design

Designs were sketched using closed rectangles and circles in commercial AutoCAD 2008

software (Autodesk, Inc., San Rafael, CA, USA), as described similarly by the Whiteside group

[61, 71]. Two-dimensional structures were drawn to represent the outline of the intended device

with fluidic channels targeted to be 35µm wide, at least twice the diameter of a typical

eukaryotic cell (~10-15µm) and to enable pressure driven flow. In the region designated as the

injector structure, the channel was reduced to a width of 15µm over a distance of 100µm to

permit single cells to sequentially enter the electrode zone at any given time. Beyond the

electrode zone the channel was further tapered to 7µm over 250µm to confine the flow of

analytes post lysis specifically to confine horizontal diffusion and encourage diffusion along the

channel. For this work, a funnel shaped design allowed a single cell to be held in place at the

centre of the electrodes as depicted in Fig. 3.1(a). The overall length of the fluidic channel was

targeted to be 10mm with a port hole, 1.25 mm in diameter, drawn at one end to be used for

fluidic access.

Channels to be converted into electrodes were designed perpendicular to the injector

structure and offset from the fluidic channel by 20µm along the x-axis. A width of 150µm,

centred at the funnel shaped region of the fluidic channel, was selected to enable the resulting

36

electric field to completely encompass a cell being held in this region as shown in Fig. 3.1(b).

The length of each electrode was planned to be 4.25mm having a 1.5mm diameter filling port

and a 260µm x 40µm air flow channel angled at 550 to the end closest to the fluidic channel. The

air flow channel terminates with a 1mm diameter port hole and along with the initial 400µm

wide region section of each electrode will allow for easier integration of the electrode material

upon device fabrication.

Figure 3.1: (a) Schematic AutoCAD drawing of integrated microfluidic device (b) Injector structure region

Printing and characterization of photo masks

Three different commercial printing companies were solicited, namely Norwood Graphics Inc.

(Toronto, ON, Canada), Pacific Arts and Designs (Toronto, ON, Canada) and The Photoplot

Store (Colorado Springs, CO, USA), to print the mask lay-out designed in AutoCAD at the

highest possible resolution to accommodate the micron sized features and gaps on the mask.

Each vendor printed the photo masks using particular photoplotters and photo film providing

varying degrees of resolution. Figure 3.2 shows a typical photoplot file that was converted from

the initial CAD drawing prior to the production of the true photo mask with the white regions

representing the transparent region through which light will pass.

(b)

Electrode Cell funnel

(a)

4.25mm

1.25mm

150µm

37

(b)

Figure 3.2: (a) Pre-printing illustration of microfluidic device (b) Injector structure region

Vendor DPI

Minimum

Line/Space

Width [µm]

Type of Photo

film

Refractiv

e Index

(n)

Norwood Graphics

Inc.

2540 10±5 NA ~1.53

The Photoplot Store 5080 5±3 Kodak Accumax

ARD7

~1.55

Pacific Arts and

Design

2000 12.7±5 Fuji Phototool

Satisfine HPR-7S

~1.57

Table 3.1: Summary of printing resolution and photo films used

Table 3.1 summarizes the printing capabilities of the three suppliers based on information that

each has provided.

The criterion used for choosing the best possible photo mask, in which i) and ii) are necessary

features, while iii) and iv) are considered desirable, were as follows:

i) minimum line/space width printing resolution < 10µm;

40µm 500µm (a)

38

ii) minimum ink spray onto clear regions targeted as microstructures;

iii) high OD at 365nm in the region of the printed black ink, with %T < 0.01%;

iv) low OD at 365nm in the clear transparent region, with %T > 50%;

Measurements of the light absorption of each type of photo film at 365nm were made so

as to determine which would allow the least amount of attenuation of the UV beam during

irradiation. A thin strip of each type of photo film was positioned on the inner wall of a 4.5ml

PMMA cuvette (VWR International, Mississauga, ON, Canada) and placed into a CARY 300

Bio spectrophotometer (Varian, Inc., Palo Alto, CA, USA) to measure the OD of the film. The

absorbance of both the transparent and the black printed parts of the photo mask were quantified

over the range 250nm to 500nm to determine the effect of both the film and the ink on the

transmission of light at the wavelength of interest, 365nm.

Using an Axiovert 200M (Carl Zeiss, Toronto, ON, Canada) inverted microscope with a

mounted CoolSNAP Pro camera (Media Cybernetics, Bethesda, MD, USA), offering an

additional 10X magnification for image acquisition and packaged with Image-Pro Plus analysis

software, images were acquired of the microstructures on the printed photo mask. The software

allowed measurements to be made of structure widths and lengths which were then compared to

the CAD drawn features to determine how well the features are reproduced. Additionally, the

images highlighted regions in which the ink spray had spread to the microchannel regions during

printing and combined with the measurements would give a more accurate view of what the true

resolution of each mask is.

3.2.2 Preparation of glass substrates

Piranha treatment of glass slides

Soda-lime glass, primarily composed of SiO2 (~73%), of dimensions 75mm x 20mm x 1mm

(Ted Pella Inc., Redding, CA, USA) were used as the substrate for photolithography. Since glass

is hydrophobic and manufactured slides may have organic residues due to handling, slides were

first exposed to high pressure air to remove dust particles and cleaned by immersing in a piranha

solution, a 3:1 volumetric ratio of 99% H2SO4 (Sigma-Aldrich Ltd., Oakville, ON, Canada) and

30% H2O2 (Sigma-Aldrich Ltd., Oakville, ON, Canada) [72, 73]. The solution hydroxylates the

glass surface thus making it hydrophilic and increasing the number of silanol groups [74]. While

working in a chemical fume hood, slides were placed in a large Pyrex baking dish and

39

submerged with 150ml of H2SO4 after which 50ml of H2O2 was poured into the dish creating a

highly exothermic reaction reaching about 1200C. After 10 minutes of piranha treatment, the

slides were removed individually from the solution and washed in 2 sequential baths of

18.2MΩ·cm deionized water for 1 minute each. This step aids in the removal of any residual

traces of the H2SO4 - H2O2 reaction. Slides not being used immediately were stored in a large

beaker with deionized water and sealed with parafilm. Prior for use as photolithographic

substrates, glass slides were submitted to a dehydration bake on a hotplate at 2000C for 30

minutes.

Spin coating photoresist with improved adhesion

SU-8 2025 (MicroChem Corp., Newton, MA, USA) was the resist used for UV

photolithography. It is a thick, negative-tone, epoxy-photoplastic that is capable of reproducing

structures with high aspect ratio [60, 75]. It has been established that to obtain a homogenous and

stable coating of SU-8 onto the substrate, there must be sufficient wetting [76], as untreated glass

has poor adhesion properties with SU-8 [66, 75] causing structures to become tilted and to

delaminate easily. Others have attempted to circumvent this problem by using a very thin (~2-

5µm), highly polymerized SU-8-2 layer, referred to as a seeding layer, between the true SU-8 to

be used in fabrication and the glass [77]. However, a known adhesion promoter [78], OmniCoat

(MicroChem Corp., Newton, MA, USA), was used as a sacrificial layer between the glass

substrate and the SU-8. While a complete understanding of chemical nature in which OmniCoat

aids in adhesion is difficult due to commercial reasons, it is generally inferred that its purpose is

to decrease the interfacial stress, which is a known delaminating factor between the SU-8 and the

glass substrate.

Using a WS-400-6NPP-LITE spin coater (Laurell Technologies Corp., North Wales, PA

USA), several drops of OmniCoat, amounting to 2ml, were placed on the glass substrate to

ensure maximum surface coverage as its thickness is of no importance [75]. It was spun for 5

seconds at 500 rpm with an acceleration of 100 rad·s-1

followed by a second spin cycle of 30

seconds at 3000 rpm accelerating at 300 rad·s-1

. These cycles created an OmniCoat layer of

approximately 20-30nm in thickness. The adhesion promoter deposition process was completed

by baking the substrate on a hotplate at 2000C for 1 minute and allowing it cool to room

temperature.

40

To achieve 20µm high structures within SU-8, 3ml of SU-8 2025 was deposited onto the

centre of the glass substrate over a sacrificial OmniCoat layer. The sample was spun at 500 rpm

for 15s with an acceleration of 115 rad/s followed by 4000 rpm for 30s with an acceleration of

342 rad/s as recommended by MicroChem Corp. (see Fig. 3.3).

Figure 3.3: Spin speed vs. film thickness curve for various photoresists (Adapted from [60])

Subsequently, the substrate then underwent a soft bake process at 650C and 95

0C, however, the

suggested times of 1 minute and 6 minutes respectively, were extended to 3 minutes and 20

minutes. The prolonged soft baking time allowed for maximum solvent evaporation which

avoids high film stress during post-exposure baking [75, 76, 79]. Following UV

photopolymerization and post-exposure baking, samples of the thin SU-8 film were removed

with a scalpel and attached to a 3mm thick slab of PDMS using a drop of glycerol to promote

adhesion. The combination piece was then placed on its thin edge (3mm) on a no. 1 glass cover

slip (~0.13-0.17mm) and observed using the Axiovert 200M microscope and measurements of

the film thickness were made using Image-Pro Plus.

41

3.2.3 SU-8 exposure and development

UV exposure with reduced diffraction effects

Prior to UV exposure, samples were kept in a Petri dish wrapped in aluminum foil to avoid

premature photopolymerization and degradation from environmental light sources. The set-up of

the substrate and mask during UV irradiation is depicted in Fig. 3.4 showing first a 25mm x

80mm x 3mm (x, y, z) base layer of PDMS which acted as a dampener to the glass substrate in

the event of external vibrations. The next layer was a piece of printed black transparency of

equivalent x, y dimensions to the PDMS whose purpose was to reduce retro-reflection of the UV

light as it passed through the substrate. Figure 3.4(a) shows one configuration in which the photo

mask was placed directly on top of the SU-8 substrate with a small air gap in between, while (b)

shows an alternative method of having a thin layer of glycerine (Sigma-Aldrich Ltd., Oakville,

ON, Canada) acting as a gap and refractive index compensating media between the SU-8 and the

mask [80, 81]. The significance of this step will be discussed below in Section 3.3.3. Small

droplets of the glycerine were deposited on the underside of the mask which would be in contact

with the SU-8 and spread evenly using a lint free swab. The mask was then pressed firmly onto

the SU-8 to remove pockets of air and to ensure uniform flatness.

The light absorption by glycerine was quantified over the 250nm to 500nm range using a

CARY 300 Bio spectrophotometer. The liquid was placed into a 4.5ml PMMA cuvette, with path

length 10mm, and the OD at 365nm was determined.

Figure 3.4: Set-up of SU-8 substrate with photo mask for UV exposure with (a) air gap, (b) glycerine. Numbers in

bracket represent thickness of each layer and corresponding refractive index

42

A SÜSS MA6 mask aligner (SÜSS MicroTec, Garching, Germany) producing an output

of 16.5 ± 0.5 mW·cm-2

was used in soft contact mode to perform UV exposure of the substrate.

This mode caused the top surface of the mask to just touch the aligner‟s heavy glass plate

without any decompression of the SU-8 substrate. Different exposure times of 10s, 12.5s, 15s,

and 17.5s were tested for photopolymerization of SU-8 to determine an optimal time with or

without the glycerine layer that would yield structures of the highest resolution. The

recommended radiant energy per cm2 for a 20µm thick film was 150 - 160 mJ·cm

-2 [60] and

based on Equation 3.1, where ERAD represents the radiant energy, and P the power, this

corresponds to an exposure time, texp, of ~9.1 - 9.7s. Meanwhile, the exposure times investigated

correspond to energies of 165 mJ·cm-2

, 206.25 mJ·cm-2

, 247.5 mJ·cm-2

, and 288.75 mJ·cm-2

respectively. These values represent the exposure before attenuation by the photo mask and

glycerine.

[mJ] (3.1)

Post exposure baking and substrate development

Immediately following UV exposure, SU-8 substrates were placed on a hotplate at 650C, for 1.5

minutes, after which they were transferred to a second hotplate at 950C for a baking time of 30

minutes. These times were longer than the recommended times from MicroChem Inc. of 1

minute and 6 minutes respectively to accommodate the thermal polymerization process that

would improve the resolution of the SU-8 structures in regions that were not adequately

photopolymerized, by continuing the cross-linking process [76, 79]. Since there is a large

thermal expansion coefficient mismatch between SU-8 (52 ppm K-1

) and glass (9 ppm K-1

), for

cool-down, substrates were placed on a hotplate at 650C for 15 minutes to minimize stress being

imprinted in SU-8 at the material interface [75] before being placed into a Petri dish at room

temperature.

Chemical development was performed by immersing substrates individually in 10ml of

SU-8 developer (MicroChem, Newton, MA, USA) for 1 minute while agitating rigorously. This

was followed by a spray rinse with 99% isopropanol (Sigma-Aldrich Ltd., Oakville, ON,

Canada) for 10-15 seconds and a final controlled high pressure air drying step to dry the

substrate and remove residual isopropanol while ensuring no SU-8 lift off from the substrate.

43

Prior to the PDMS microchip development, the substrate, referred to henceforth as the SU-8

master, was examined under the Axiovert200M microscope to determine the resolution of the

structures and the shrinkage of structure width which may have occurred during exposure and

development.

Measurement of side wall angle of SU-8 structures

Due to the rigidity of the SU-8 microstructures, a direct measurement of their x-z angularity was

not possible without damaging and delaminating the structures in the process. Instead, the SU-8

master was placed on a flexible polycarbonate sheet with the structures facing upward and a rigid

acrylic frame, 4mm deep, surrounded the master. 20ml of PDMS mixture was made from a 10:1

(V:V) base to curing agent of Sylgard 184 elastomer (Paisley Product Inc., Toronto, ON,

Canada) and degassed using a vacuum desiccator (Bel-Art Products, Pequannock, NJ, USA) for

20 minutes. PDMS was cast onto the SU-8 master and 350g aluminum weights were used to

keep the frame in place and to prevent prepolymer PDMS from leaking out while the

polycarbonate sheet (P & A Plastics Inc., Hamilton, ON, Canada) was placed on a hotplate at

700C for 1.5 hours. The flexible sheet enabled the SU-8 master with the casted PDMS to be

easily removed without damaging the SU-8 master or the PDMS. The cured PDMS piece,

referred to as the PDMS master, was 4mm in height given by the depth of the acrylic frame and

was slowly peeled away from the SU-8 master to prevent the SU-8 structures from lifting off the

glass substrate and becoming embedded in the PDMS.

A transverse slice was made through the PDMS master to obtain a cross sectional view of

the fluidic and electrode channels and observed using the light microscope. Using the ImagePro

Plus software, the side wall angles were measured as illustrated in Fig. 3.5. To properly

characterize the variation of the side wall angle as a function of channel width, SU-8 structures

of widths ranging from 5µm to 100µm were subjected to the above mentioned procedure and a

graph of channel width versus side wall angle was plotted for structures created with and without

glycerine as the gap compensating media.

44

(

)

Figure 3.5: Illustration of cross sectional view of a microfluidic channel and method of measuring side wall angle

3.2.4 Device fabrication by rapid prototyping of PDMS

Production of rigid epoxy master

The SU-8 master was replicated in PDMS by casting as previously described, (Section 3.2.3)

producing a PDMS master, inverse to the SU-8 master, with valleys or channels where ridges

and structures exist on the SU-8 master. Due to the fragile nature of the SU-8 master, a rigid

epoxy master was fabricated which would maintain the resolution of the original master and

could be used repeatedly without degradation of microstructures. With the channels facing

upwards, the PDMS master was placed in a 6mm deep rectangular receptacle made of the same

elastomer, and 20ml of a single component UV curable epoxy, OG 169 (Epoxy Technology,

Billerica, MA, USA), was poured onto the master. This was cured using an in-house UV light

box with an irradiance of 2.0 ± 0.2 mW·cm-2

for 45 minutes to create the epoxy master. Due to

the flexibility of the PDMS receptacle, the final master was easily removed after curing.

PDMS

45

Figure 3.6: Illustration of multi-step soft lithography process

With this approach, rapid production of PDMS microfluidic chips was achieved in less

than 1.5 hours, first by pouring 10ml of prepolymer PDMS into the epoxy master and degassing

for 20 minutes, immediately followed by a 1 hour baking period at 700C, and careful removal

from the epoxy master, to ensure all fine details of the fluidic channels remain intact. Figure 3.6

illustrates the complete soft lithography process which allows rapid prototyping of the PDMS

chip. Access ports for fluidic entry and control, air flow exit and electrode filling were created

using Harris Uni-core punches (Ted Pella Inc., Redding, CA, USA) of internal diameters 0.5mm,

1.5mm and 2.0mm respectively, while a 3.0mm punch was used to create a reservoir at the

opposite end of the fluidic channel away from the entry and control port.

Verification of wall uniformity by confocal microscopy

While the method described in Section 3.2.3 allows only a small number of single point views

and measurements of the side wall angle, multiple 3-D images of the microfluidic channels were

obtained using confocal microscopy after filling the fluidic and electrode channels with different

fluorescent dyes. The electrode and their air outlet channels were filled with a 0.1mM solution of

Rhodamine 123 (excitation = 505nm, emission = 560nm) (Invitrogen Inc., Burlington, ON,

46

Canada), while a 0.1mM solution of Hoechst 33342 (excitation = 350nm, emission = 461nm)

(Invitrogen Inc., Burlington, ON, Canada) filled the fluidic channel. Using a Zeiss LSM 510

META NLO inverted microscope (Carl Zeiss, Toronto, On, Canada) with a 10x objective, a Z-

stack of the fluorescent channels in the vicinity of the injector structure region was obtained at

2µm slice intervals.

PDMS bonding by air plasma

To enclose the network of channels, a fourth surface was required as the moulded PDMS chip

provides only three walls. This surface was fabricated by spin coating prepolymer PDMS for 40s

at 750rpm onto a no. 1 glass cover slip to create a 100µm thin membrane [82] which was baked

on a hotplate at 700C for 1 hour to cure. The thickness of the membrane was verified using

Image-Pro Plus as previously described (see Section 3.2.2). Due to the high pressures that can

exist within the channels, especially during the electrode filling process, an irreversible seal was

necessary between the PDMS chip and PDMS cover slip. By exposing the two surfaces to an air

plasma treatment, an increase in the surface free energy and Silanol groups are created,

permitting covalent bonding when the two PDMS substrates are brought into conformal contact,

thus forming a tight, irreversible seal [68, 83].

Traditionally, vacuum plasma systems with a controlled oxygen flow are used for this

bonding process, however, a less cumbersome, hand held corona treater was used in this work

which operates in room air at atmospheric pressure [84]. In the confines of a fume hood, a BD-

20AC portable device (Electro-Technic Products Inc., Chicago, IL, USA) was held in a fixed

vertical position with a retort stand (Fig 3.7) while the two PDMS parts were placed flat on a

motorized stage moving back and forth at a speed of 10 ± 1 mm·s-1

. The PDMS surfaces were a

distance of 4.0 ± 0.2mm from the wire electrode of the corona treater and exposed to the plasma

for 8 passes, totalling 45 seconds, after which the PDMS chip was manually pressed lightly

against the thin membrane to complete the sealing process. These PDMS devices are rendered

hydrophilic for a brief period after the plasma treatment and a 12 hour period was allowed before

they were used for any microfluidic experiments.

47

Figure 3.7: Corona treater with PDMS chip and spin coated cover glass on a motorized stage

3.2.5 Material characterization of integrated electrodes

Composite PDMS with carbon

The design of the microfluidic chip in this project allows for the integration of electrodes by

filling a suitable material into the channels intended as electrodes. Xiu et al [85] reported a

method of incorporating carbon black particles, which are known to have good electrical

properties (ρ ≈ 0.2Ω·cm) [86], into prepolymer PDMS to create conducting composites. Since

PDMS, even at higher than normal viscosities after mixing with carbon, would be straight

forward to integrate into the electrode channel to create an almost seamless electrode, a similar

method was followed by mixing Vulcan XC72R carbon black (Cabot Corp., Boston, MA, USA)

into prepolymer PDMS with concentration based on weight. Composites of mixing ratios 5%,

10% and 15% by weight of carbon to PDMS were created by adding the powdery carbon black

in 0.1g increments to the PDMS and mixing vigorously to promote uniform dispersion of the

carbon particles within the composite. This was necessary since carbon black has a density of

0.096g·cm-3

compared to that of PDMS, 0.965 g·cm-3

.

Corona treater

Motorized stage

Electrode wire

PDMS chip

Spin coated

cover glass

48

Different lengths of tubing (Cole-Parmer Inc., Montreal, QC, Canada), ranging from 3cm

to 12cm, each with an inner lume of 0.81mm, were filled with each of the composites. A 20mm

length of copper wire of diameter 250µm (Consolidated Electronic Wire & Cable, Franklin Park,

IL, USA), was inserted at each end and cured on a hotplate at 700C for 2 hours to create carbon

based PDMS wires (Fig. 3.8a), and their resistance was measured using a digital multimeter

(Fluke Electronics, Mississauga, ON, Canada) . By plotting the resistance, R, as a function of

length, l, the electrical resistivity, ρ, was determined by multiplying the slope of the graph and

the cross sectional area, A, of the tubing, following Equation 3.2, which assumes that the

composite is a uniform electrical conductor . Equation 3.2 does not consider the random

arrangement of the carbon particles within the PDMS matrix.

The integrated electrodes were fabricated by filling the carbon-PDMS composite into the

electrode channels of the microfluidic device using a 10ml syringe under high pressure via the

designated filling ports, inserting copper wire and curing as described above. Effectiveness of the

filling process throughout the electrode was observed under a light microscope.

Electrically conductive epoxies and pastes

Electrical properties of manufactured compounds embedded with metals, giving them the ability

to conduct, were also investigated and integrated into the microfluidic device. Table 3.2

summarizes the properties of the different epoxies and pastes tested as given by the

manufacturers that were used as integrated electrodes.

49

Property

Product Name

SS-25M SS-25M + OMS Silver Paste Plus Silver Conductive

Epoxy

Manufacturer

Silicone Solutions,

Twinsburg, OH,

USA

Silicone Solutions,

Twinsburg, OH,

USA + Atelier

D‟art Hade &

Rodier, Granby,

QC, Canada

SPI

Supplies/Structure

Probe Inc., West

Chester, PA, USA

MG Chemicals,

Surrey, BC,

Canada

Description

-Single part epoxy

-Moisture curing

RTV

-Single part epoxy

-Moisture curing

RTV

-1:5 volumetric

ratio of SS-25M to

OMS†

-Single part paste

-Uniform particle

distribution

-Moisture curing

- Two part epoxy

- Moisture curing

Conductive Filler 98% Nickel and

Graphite

Nickel and

Graphite

~72% Silver ~45% Silver

Particle Size [µm] 2-15 2-15 0.3-0.6 1-5

Viscosity [cps] 100,000 NA ~70,000 ~60,000 (mixed)

Electrical

Resistivity (ρ)

[Ω·cm]

6 x 10-2

NA 3 x 10-5

3.8 x 10-1

Electrical

Conductivity (σ)

[S·cm-1

]

16.7 NA 3.33 x 104 2.63

† - odourless mineral spirit

Table 3.2: Properties of electrically conductive epoxies and pastes used as integrated electrodes

50

Each compound was filled into different lengths of tubing and underwent an overnight

curing period, followed by a measurement of the resistance as described previously in Section

3.2.5. Their resistivity was also extracted from the slope of the resistance as a function of length,

according to Equation 3.2. The filling process was carried out in the same manner as described

for the carbon-PDMS composite and the resistance of the integrated electrode was also measured

within the microfluidic chip. Fig 3.8c illustrates the approach selected by inserting a 20mm

length of copper wire (diameter) through the PDMS to make contact with the tip of the electrode

proximal to the fluidic channel.

Figure 3.8: a) Tubing filled to measure resistance b) Cross sectional view and c) top view of chip with electrode

filled; d) Cross sectional view of chip showing resistance measurement with inserted copper wire

Injection moulding using molten solders

A modification to the system of fabricating micro electromagnets within a PDMS environment

as described by Siegel et al [87] allowed micro wires to be embedded in the previously described

PDMS microfluidic device creating the integrated electrodes. This is possible as PDMS is

thermally and mechanically stable up to 3000C [88]. A 0.1M solution of 3-

mercaptopropyltrimethoxysilane (Sigma-Aldrich Ltd., Oakville, ON, Canada) in acetonitrile

(Sigma-Aldrich Ltd., Oakville, ON, Canada) was injected into the intended electrode channel

and stored at room temperature for 1 hour to allow the solvent to evaporate. This silane

compound coats the channel, reducing its surface free energy and making it wettable to the

molten solder.

l

A

(a)

(b) (c)

51

Concurrently, 2g of 99.9% In solder (AIM Solders Inc., Cranston, RI, USA) was placed

in a metal tipped 10ml glass syringe (Cole-Parmer Inc., Montreal, QC, Canada) wrapped in

silicone heating tape (HTS/Amptek Company, Stafford, TX, USA) and heated to ~2000C,

melting the solder (melting point = 1570C). After the 1 hour silanizing period, the microfluidic

device was placed on a hotplate at 2250C and molten solder was injected into the electrode

channel by inserting the tip of the syringe into the filling port and applying manual pressure to

the syringe. After complete filling is observed, a 20mm length of 250µm diameter copper wire

was inserted into each port and the device was removed from the hotplate to cool at room

temperature bonding the copper wire firmly. This method was repeated using 57Bn25In17Sn

solder (AIM Solders Inc., Cranston, RI, USA) which has a melting point of 790C.

Since it was difficult to manipulate molten solder, the method of measuring the resistance

of the material by inserting it into different lengths of tubing (Section 3.2.5) was not applied

here. Instead, the resistance of a solid bulk solder block, 55mm x 4mm x 3mm, was measured

and the resistivity calculated using Equation 3.2. The resistance of the integrated electrode was

measured by making contact with the electrode tip by inserting a piece of copper wire through

the PDMS as previously described.

52

3.3 Results and Discussion

3.3.1 UV absorbance and micrograph resolution of photo masks

The thickness of each type of photo mask investigated, along with their absorbance at 365nm

extracted from Figures 3.9 (b), is shown in Table 3.3

Figure 3.9: a) Absorbance as a function of wavelength for printed parts of photo mask, b) Absorbance as a function

of wavelength for transparent part of each photo mask

a) b)

53

Vendor Thickness/µm OD365nm

(Black ink)

OD365nm

(Clear

transparency)

Norwood Graphics

Inc.

85±5 6.014 0.235

The Photoplot Store 175±5 5.950 0.202

Pacific Arts and

Design

175±5 6.723 0.255

Table 3.3: Thickness and OD measurements of 3 different photo mask

Each of the three photo masks demonstrate negligible light transmission (~0.0001%)

through the black ink while the clear region transmits >55% of the light, thus satisfying the two

secondary criteria. As a result, the determining factor for the mask choice was made based on

the micrograph images of the masks (Fig. 3.10) which determined whether the imperative criteria

were met. These images showed that while the Norwood mask was reported to have a minimum

resolution of 10µm, the edge features along the intended channel region were poor and there was

significant ink spray in clear regions. The Pacific Arts mask had poor resolution at 12µm and

was unable to achieve the targeted CAD features less than 10µm. Meanwhile, the Photoplot

Store mask exhibited significantly better edge features, minimal ink spray and combined with a

minimum line/width resolution of 5µm was determined to be the closest match to the imperative

criterion and was selected for all photolithographic experiments.

54

Norwood Photoplot Store Pacific Arts

Figure 3.10: Micrograph images of mask features from 3 suppliers (top - 10x objective; bottom - 20x

objective)

3.3.2 SU-8 spin coating and exposure

SU-8 spin coated thickness

The photo resist thickness was measured once monthly for 4 consecutive months and 5 samples

each time, to determine the consistency of the spin coater in producing a targeted 20µm

thickness of SU-8. Results indicated that the average thickness when spin coating was executed

as recommended in Fig. 3.3 was 20.6 ± 0.2µm.

Optimization of exposure times

The OD of the chosen photo mask (Photoplot Store) at the wavelength used for UV

photolithography was determined to be 0.202 (Table 3.3) in the clear region through which light

is transmitted. Meanwhile, the OD, AOD, of glycerine at 365nm for a 10mm path length, l, was

determine to be 0.0351, giving an absorption coefficient, α, of 0.00351mm-1

from Equation 3.3a.

By assuming that the layer of glycerine between the photo mask and the SU-8 is ~10µm, and the

experimentally determined absorption coefficient, the OD of the glycerine layer at 365nm was

3.51 x 10-5

. This confirms that the photo mask impacts the incident light reaching the SU-8 layer

40µm Bottom Images 140µm Top Images

55

due to absorption, while neither an air gap, nor a glycerine layer between the mask and the SU-8

results in any significant absorption.

(3.3a)

[mW.cm

-2] (3.3b)

It was previously stated that the irradiance of the mask aligner was 16.5mW·cm-2

.

However, according to one form of the Beer-Lambert law (Equation 3.3b), the irradiance

reaching the SU-8 substrate was calculated to be ~13.48mW.cm-2

. Consequently, the radiant

energy per cm2 for each of the times used during SU-8 photopolymerization determined by

Equation 3.1 has been summarized in Table 3.4.

Exposure Time

(s)

Radiant Energy per cm2

on photo mask [mJ·cm-2

]

(16.5mW.cm-2

irradiance)

Radiant Energy per cm2

on SU-8 [mJ·cm-2

]

(13.48mW.cm-2

irradiance)

10 165 134.8

12.5 206.25 168.5

15 247.5 202.2

17.5 288.75 235.9

Table 3.4: Initial and final radiant energy per cm2 due to absorption in photo mask

The recommended radiant energy per cm2 for a 20µm layer of SU-8 is 150-160mJ·cm

-2,

which would have required an exposure time of 11.1s-11.9s according to the adjusted irradiance

of 13.48mW·cm-2

. Three of the four exposure times used were greater than the recommended

value and as a result there is an overcompensation of the required radiant energy per cm2. This

ensures that there is adequate photopolymerization and cross-linking of the compounds in the

SU-8 photoresist. Based on the calculated radiant energies, an exposure time of 12.5s yields the

closest match to the recommended radiant energy, while it could be expected that the longer

56

exposure times may cause over-exposure of the SU-8 film, resulting in loss of feature

delineation and hence resolution [75].

Improvement of SU-8 resolution using glycerine

After developing the SU-8 structures, it was observed that SU-8 films subjected to 10s of

exposure time easily delaminated during the drying process, presumably due to inadequate

photopolymerization of the photoresist, particularly near the SU-8 – glass interface. Meanwhile,

micrograph images (Fig. 3.11a) of the developed SU-8 structures confirm that the edge features

of over-exposed exhibit poor resolution at times exceeding 12.5s when there is an air gap

between the photo mask and the SU-8 surface. However, as previously shown by Kang et al

[81], results indicate that the use of glycerine (Fig. 3.11b) as a gap and refractive index

compensating media improves the resolution of edge features by reducing diffraction effects

and allows minimum line widths of 5µm for the Photoplot Store mask to be maintained. Those

diffraction effects will be explored in greater detail when discussing the side wall angle

measurements.

12.5s 15s 17.5s

a) Without Glycerine

b) With Glycerine

40µm

Figure 3.11: Resolution improvement of SU-8 structures using glycerine for different exposure times

57

3.3.3 PDMS device development

Side wall angle and channel uniformity measurements

A significant improvement in the side wall angle of the PDMS channels was observed when a

thin layer of glycerine was used as a compensator removing the air gap between the photo mask

and the SU-8 surface. Micrograph images (Fig 3.12) provide examples of this phenomenon while

the plot of side wall angle as a function of channel width for the three exposure times (12.5s, 15s,

17.5s) confirms the angle improvement (Figure 3.13).

Figure 3.12: Micrograph images of PDMS channels showing side wall angle improvement with glycerine at an

exposure time of 12.5s

Figure 3.13: Side wall angle as a function of channel width for different exposure times

The ideal side wall angle, υ, is 900, and was not achieved for any of the exposure times, while

the angle increased with channel width. It asymptotically approached 86.50, 85

0 and 85

0 for

exposure times of 12.5s, 15s and 17.5s respectively while the slope of wall angle versus channel

width approached 0 for glycerine use compared to a gradient when an air gap was present.

Meanwhile, the terminal slope of υ as a function of channel width is still different from zero for

PDMS – Without Glycerine PDMS – With Glycerine

50µm 50µm

Without Glycerine With Glycerine

58

the samples without glycerine. The common feature with samples with and without glycerine

appears to be a lower side wall angle for structures with aspect ratios 1:2 or greater, representing

a narrower channel base than wall height.

With an air gap between the mask and SU-8 layer, the UV light passes from a refractive

index 1.45 (photo mask), to 1 (air), and finally 1.65 (SU-8). Refraction of the UV light due to the

refractive index mismatch between the layers does not apply to explain a wider upper region near

the surface of the photoresist, and a narrower structure near the base as the UV beam is well

collimated. Chuang et al [80] proposed an explanation for the side wall angle phenomena based

on Fresnel diffraction. The thinness of the photo mask and its lack of flatness creates an air gap

between the mask and the SU-8 surface while the edges of the mask patterns cause diffraction

effects, with a Fresnel number, u, determined by Equation 3.4, where x represents the width of

the slit, y is the vertical distance to the photo mask, and λ is the wavelength.

(3.4)

Due to a large y value when an air gap is present, the Fresnel number is close to 10,

resulting in significant diffraction [89], accounting for the „top hat‟ effect when exposure extends

laterally from the slit. With the air gap eliminated by glycerine (n = 1.47), which has a refractive

index close to that of the photo mask, the distance between the mask and the SU-8 surface is

reduced considerably, resulting in a larger Fresnel number which yields a more uniform

diffraction pattern. Equation 3.4 also supports both air gap and glycerine experimental

observations that the side wall angle, which is indicative of the degree of diffraction, is greater

when the slit size is small, since at these widths the Fresnel number will be even closer to 1. It

should be noted that while the glycerine decreases the Fresnel diffraction effects, it does not

completely eliminate it since a perfectly uniform gap distance, and infinitely long slit size would

be required to create side wall angles of 900 [81].

Z-stack analysis of channel uniformity

The 3-D images obtained by confocal microscopy verified that in the vicinity of the injector

structure, where the electrode channel is closest to the fluidic channel, there were no significant

structural deformities or aberrations along the viewable lengths of either channel that may cause

non-uniform fluid flow or obstruct cell movement. This was evident even in the narrowest fluidic

59

channel region (~5.5µm). Primarily, the images and cross sectional profile (Fig. 3.14) of the

intensity measured transversely across a channel, further confirmed that the side wall angle

remains uniform along the length of the channel due to the negligible ascending and descending

slope of the profile. These images also show that there were not leakage points between the

PDMS-PDMS bond on the base of the device, nor from the fluidic or electrode channels into the

surrounding PDMS structure.

Figure 3.14: (a) Central plane in the x-axis; (b) Central plane showing z-axis view (top), and y-axis view through

the fluidic channel (right); (c) Sample profiles through various regions

Resolution losses from CAD to PDMS device

The fabrication process to develop the PDMS microfluidic device is susceptible to several

possible stages of line width resolution losses. From the CAD design to the printed mask, losses

are expected due to the printer‟s inability to fully capture the fine features, while SU-8 expansion

due to the cross-linking and development process would also change the feature sizes [76]. Table

3.5 shows a comparison of the channel widths and separator gaps for the various steps of the

fabrication process.

(a)

(b)

(c)

150µm

150µm

FWHM = 5.3µm

FWHM = 28.8µm

FWHM = 139.7µm

60

CAD Width/

Separator

[µm]

Mask

Width/

Separator

[µm] ±

1.5

SU-8

Structure

Width/

Separator

[µm] ± 1.5

PDMS

Structure

Width/

Separator

[µm] ± 1.5

Total Change of

Width/

Separator [µm]

%

Difference

7 (channel) 4.5 5.5 5.5 -1.5 21.4

15 (channel) 10.5 11.25 11.25 -3.75 25

20 (separator) 13.75 12.5 12.5 -7.5 37.5

40 (channel) 28.5 30 30 -10 25

200 (channel) 138.5 142.5 142.5 -57.5 28.75

Table 3.5: Variation of channel widths and gaps during fabrication process

The data suggests that the average difference from CAD drawing to the final PDMS structure is

27.5%, of which a majority is due to the printing process while the SU-8 structures undergo a

small expansion during the fabrication process and are within ~10% of the photo mask

structures. As expected, the PDMS channels are exact copies of the SU-8 structures, yielding no

resolution losses which will be important in the future if structures with resolution < 5µm are to

be developed. It should be observed that while the gap separating the electrode from the fluidic

channels decreases, this is due to the expansion of the surrounding SU-8 structures, particularly

the intended fluidic and electrode channels, thus causing a shrinkage of the inter channel

distance.

3.3.4 Electrode material characterization

Physical description of integrated electrodes

Carbon filled PDMS composites demonstrated poor syringing properties due to its high

viscosity, having a thick, paste-like consistency. The high pressures required to fill the channels

with these compounds often resulted in blistering, in which the PDMS seal was irreversibly

61

broken. Additionally, the manual mixing process proved difficult due to the low density and

powdery nature of carbon black. While the mixing and filling processes were non-ideal, the

composites, however, showed good integration in the electrode channels having also been made

with PDMS (Fig. 3.15a).

In its original form, the nickel based electrode material was also difficult to fill into the

electrode channels, however, when its viscosity was lowered using the odourless mineral spirit,

the filling process was significantly easier. Micrograph images (Fig. 3.15b) show that due to the

size of the nickel pieces, the electrode matrix lacked continuity as large gaps existed between

particles, while the low viscosity form of this epoxy caused the particles to be spread even

further apart. This would negatively impact the electrical conductive properties of the electrode

as the particles should be closely packed to ensure a continuous current flow.

Figure 3.15: Physical properties of integrated electrodes made of different material, with (a) carbon-PDMS; (b)

nickel paste; (c) silver paste

The single part silver paste filled the channels easily without requiring excessive

syringing pressure while particles appeared to be in a compact matrix. However, due to the

alcohol base in which this paste is produced there is a condensation of the electrode during the

curing process, causing it to pull the PDMS gap that separates the electrode from the fluidic

channel resulting in an expansion of the injector structure region (Fig. 3.15c). This would

significantly impact the device‟s ability to hold a single cell in place during lysis. Meanwhile, the

two part silver epoxy was difficult to apply via a syringe and fill the channels due to its high

viscosity but particles appeared to be packed closely together.

High pressures were required to start the filling process of both types of molten solder;

however, once the solder enters the channel, the filling process is completed in about 5 seconds

aided by capillary forces drawing the liquid in. Gap formations within the solder in the channel

(a) (b) (c)

62

were observed when the pressure being applied to the syringe was not consistent. Some of these

gaps were eliminated by increasing the temperature of the hotplate to 2500C while applying

syringe pressure to encourage reflow of the molten solder to fill the gaps. The Indium solder,

while having a high melting point, filled the channels uniformly, unlike the BnSnIn combination

which has a lower melting point but upon cooling became brittle. This led to poor bonding with

the copper wire inserted in the port and cracking along the length of the electrode, with complete

loss of conductivity.

Electrical properties of electrode composites, pastes and epoxies

Figure 3.16: Resistance as a function of tubing length for electrode materials

The plots in Fig. 3.16 show the results of the resistance measurements in the various lengths of

tubing. This was necessary to verify the resistivity and conductivity of the electrode material and

to determine which would be suitable as an integrated electrode. Table 3.6 summarizes the

information extracted from Fig. 3.16, with the resistivity (ρ) calculated by taking the slope of

each plot (R/l) and multiplying it by the cross sectional area of the tubing as shown in Equation

3.2, while the conductivity (σ) was determined as the inverse of the resistivity.

63

Electrical

Property

Product/Material Name

C-

PDMS SS-25M

SS-25M

+ OMS

Silver

Paste

Plus

Silver

Conductiv

e Epoxy

Indium

Solder

Bn-In-

Sn

Solder

Manufacturer‟s

Resistivity

(ρ)/Ω·cm

NA 6 x 10-2

NA 3 x 10-5

3.8 x 10-1

8.37 x 10-5

NA

Measured

Resistivity

(ρ)/Ω·cm

5%:

1998.2

10%:

1367.7

15%:

771.1

4.9 x 10-2

248 2.63 x 10-3

2.51 x 10-1

†4.36 x 10

-3

†7.4 x 10

-2

Manufacturer‟s

Conductivity

(σ)/S·cm-1

NA 16.7 NA 3.33 x 104 2.63 1.19 x 10

4 NA

Measured

Conductivity

(σ)/S·cm-1

5%:

5.0 x 10-4

10%:

7.3 x 10-4

15%:

1.3 x 10-3

20 4.03 x 10-3

3.81 x 102 3.98 2.29 x 10

2 13.5

Microelectrode

Resistance/Ω NA 5.78 x 10

4 24 x 10

6 2.87 x 10

3 2.69 x 10

5 4.61 x 10

3 7.95 x 10

4

† - obtained from resistance measurements of solder block

Table 3.6: Electrical properties of all conductive materials used as integrated electrodes

The table above indicates that the Silver Paste Plus single part paste had the best conductivity

and lowest resistance at the microelectrode level. However, the process of measuring the

resistance of the microelectrode often led to damages to the electrode due to the copper wire

breaking the paste matrix or compressing it in an irreparable manner. This was observed for all

64

integrated electrodes in which a paste or epoxy was used. Meanwhile, the indium solder had a

comparable electrical conductivity and microelectrode resistance, and while measurement of the

microelectrode resistance also punctured the embedded solder, the device was placed on a

hotplate at 2500C to re-melt the solder, causing reflow and fixing any damaged regions. As a

result, the indium solder was determined to be the preferred choice for the integrated electrode

material and was used for fabricate all microfluidic devices for in vitro studies.

3.3.5 Microfluidic device with integrated electrodes

Figure 3.17: Final microfluidic device with integrated solder electrodes

100µm

65

3.4 Conclusion

The work in this chapter has demonstrated the ability to improve the resolution and feature size

of structures fabricated via UV photolithography using a plastic photo mask. The minimum

feature size of 5.5µm achieved, with near straight side walls, is significantly better than those

fabricated by similar methods as reported in the literature [90] and enabled the development of a

reproducible microfluidic device capable of confining a single cell while restricting the diffusion

of analytes. Additionally, a unique integration of 3-D electrodes, close to, but not in direct

contact with the fluidic channel was also implemented through a combination of the optimized

fabrication process and injection moulding of good electrically conducting solder.

66

Chapter 4 In vitro experimental verification of plasma membrane lysis

4.1 Introduction

Verification of selective electrical lysis of the plasma membrane, while the nuclear membrane

remains intact, should be facilitated with normal and diseased cells to determine the effectiveness

of the lysis method on each type. The work in this thesis employed 3T3 rat fibroblasts as a

normal model, and 9L rat gliosarcomas as a diseased brain tumour model.

3T3 cells

Established in 1962, the name refers to “3-day transfer, inoculum 3 x 105 cells”, and due to the

large volume of data available on this cell line, it has been used as a model system extensively in

experimental research. Furthermore, these adherent cells have been used repeatedly in

microfluidics and have demonstrated the ability to maintain viability in PDMS structures [91].

Electroporation of single 3T3 cells has been reported to occur with an applied electric field

strength of under 4 kV·cm-1

[92] and are typically 8-15µm in size.

9L cells

These rare, malignant form of gliomas are adherent and have sizes between 10-12µm. These

cells closely simulates glioblastoma multiform when implanted in vivo and consequently has

been used often in photodynamic therapy studies [93] and more recently in detecting cancer

stem-like cells [94]. While no literature exists on electroporation of single 9L cells, they are

typically lysed by ~1 kV·cm-1

electric fields.

67

4.2 Materials and Methods

4.2.1 Experimental set-up

Fluidic system and flow control

Common flow control features in microfluidic devices often include syringe pumps [95], on-chip

micropumps, microvalves and actuators [96, 97]. Initially for this project, an NE-1000 syringe

pump (New Era Pump Systems Inc., Wantagh, NY, USA) was used; however, the minimum

flow rate was limited to ~0.2nl·s-1

for a 1ml syringe. The total volume of the fluidic channel

between the entry port and the injector structure was ~1nl, thus only a 5s window was available

for fluid manipulation. As a result, a system relying on Bernoulli‟s principle was adopted for this

thesis. In fluid dynamics, this principle states that a change in the speed of a fluid undergoing in-

viscous flow occurs simultaneously with a change of its potential energy or its pressure [98].

Noguchi et al [99] presented a thorough analysis of flow dynamics of model particles (vesicles)

governed by a modified Poiseuille flow in microfluidic channels due to rectangular channels

compared to normal cylindrical channels in which the flow was controlled by Bernoulli‟s

principle with a height varying system. A modification to their system was implemented to allow

flow manipulation in this thesis.

The first part of the fluid control mechanism comprised a step to flush the fluidic channel

with a 3ml syringe with a 23 gauge needle attached. The needle was sheathed by Tygon flexible

plastic tubing (Cole-Parmer Inc., Montreal, QC, Canada), 60cm in length, with inner diameter

0.51mm and outer diameter 1.52mm. At the opposite end of the tubing, a 12.7mm length of

stainless steel tubing (New England Small Tube Corp., Litchfield, NH, USA) with a 0.43mm

bore and outer diameter of 0.635mm was fitted, and inserted into the fluidic entry port of the

microfluidic chip. A buffer solution containing 0.01% Pluronic (Sigma-Aldrich Ltd., Oakville,

ON, Canada) was used to fill the channel using the syringe set-up, after which the needle was

removed and submerged in a 15ml Falcon tube containing the same solution. The tube was

secured to a height varying stage controlled by a z-axis travel translation stage (Thorlabs,

Newton, NJ, USA) as shown in Fig. 4.1 initially set to an equilibrium state of zero flow. The

microfluidic chip was secured to an in-house fabricated microscope stage using a pair of metal

clamps to ensure chip stability during experiments and observations were made using an

Axiovert 200M inverted microscope with a 10X Fluar objective (Carl Zeiss, Toronto, ON,

Canada)

(a)

68

Figure 4.1: Image of flow control system

Cell preparation and fluorescent labelling of cytoplasm and nucleus

Two different cell lines were used for in vitro experiments on chip, namely 3T3 fibroblasts and

9L gliosarcoma cells. Each cell line was cultured using Dulbecco‟s Modified Eagle‟s (DME)

Medium formulation (Invitrogen Inc., Burlington, ON, Canada) containing physiological

components including vitamins, amino acids, sugars, inorganic salts and phenol red to determine

the presence of any chemical reactions within the media which may affect pH. The typical

physiological pH of cells is ~ 7.2-7.4 which produces red colour in the cell culture media;

however, a change in the pH to more basic or more acidic would be accompanied by a change in

colour. The media was also supplemented with 10% fetal bovine serum (FBS) which contains

essential growth factors aiding in stimulation of cellular growth. Antibiotics in the DME, such as

streptomyocin and penicillin, also help to prevent the growth of gram positive and gram negative

organisms which may infect or contaminate the cell culture. Cells were incubated in standard cell

culture flasks at 370C/5% CO2.

Specialized

microscope

stage Translation

Stage Translation

Stage

69

The membrane permeable dye, calcein AM (Invitrogen Inc., Burlington, ON, Canada), excited at

495nm and emits at 515nm, was used as a fluorescent marker for the cytoplasm of viable cells.

Upon entering the cell, the acetomethoxy group of the dye (Fig. 4.2) is hydrolyzed by

intracellular esterases converting the non-fluorescent calcein AM into a strong green-fluorescent

calcein. Cells were trypsinized and re-suspended (~1-2 x 106 cells·ml

-1) in buffered media of pH

7.2 with phenol red free media (alpha MEM), after which 1mM of the dye was administered and

incubated for 1 hour at 370C, allowing adequate transport through the membrane and hydrolysis

in the cytoplasm.

Figure 4.2: Calcein AM molecule (C46H46N2O23) and excitation/emission spectra (Adapted from [100])

Following incubation, cells were centrifuged at 2000 rpm, the supernatant removed, and

re-suspended in clear alpha MEM. To tag the nucleus, Hoechst 33342 (Invitrogen Inc.,

Burlington, ON, Canada) was used and is excited at ~350nm and emits at 461nm (Fig. 4.3). The

dye is rendered lipophilic by its ethyl group, making it permeable to the plasma membrane, and

binds to the A-T base pairs of DNA. A concentration of 5µM of Hoechst was added to the

suspended cells and incubated for at least 30 minutes at 370C to allow diffusion of the dye. After

this incubation period, cells were again centrifuged at 2000 rpm, the supernatant removed and a

final re-suspension in clear alpha MEM was carried out.

70

Figure 4.3: Hoechst 333342 molecule (C27H37Cl3N6O4) and excitation/emission spectra (Adapted from [100])

On the microfluidic chip, the cell reservoir, located at the opposite end of the fluidic

channel to the control port (Fig. 3.1), was filled with 50µl (~0.5 – 1 x 105 cells) of the cell

suspension and the height of these reservoirs represented the zero potential. Fine adjustments

were made to the z-stage, varying the Falcon tube holder by 1mm increments to create a pressure

gradient between the reservoir and the fluidic control port, permitting adequate control of the

flow, and allowing a single cell to be stopped completely in the region of the injector structure,

eliminating mechanical stress during lysis. While optical tweezers were available to facilitate

single cell selection and transport from the cell reservoir to the injector structure, the distance of

~3mm would have required too much time.

Electronic set-up for electrical lysis

According to simulations in Chapter 2 Section 2.3.2, an electric field in the range 4.507-

4.532kV·cm-1

is generated when a potential difference of 32V is applied across the electrodes,

thus to examine the effect of varying the magnitude of the electric field on cell lysis, a DC power

supply capable of achieving a range of at least 0 – 50V was selected. Additionally, a function

generator or arbitrary waveform generator was needed to create a pulsed DC source to

effectively induce electroporation as explained in Chapter 2, Section 2.1.2 and an Agilent

33220A (Agilent Technologies Inc., Mississauga, ON, Canada) was used for this requirement.

Since this function generator has a maximum peak voltage output of 10V, an external DC power

supply was required to generate a DC offset and produce a new maximum peak voltage of at

least 32V. This was achieved using an Agilent HP 6209B DC power supply (Agilent

Technologies Inc., Mississauga, ON, Canada) with a voltage output range of 0 – 320V,

71

connected in series as shown in Fig. 4.4, to the function generator and the load, in this case, the

micro electrodes, using a method suggested by Agilent Technologies [101]. The electrical

connection to the copper wire electrode contacts was made using a BNC cable modified with a

pair of mini grippers (Mouser Electronics, Mansfield, TX, USA) at one end while the opposite

end was connected to the function generator.

Figure 4.4: Circuit diagram of function generator with external DC offset

4.2.2 Visual monitoring of electric field induced cell lysis

Quantification of photobleaching effects

With the electrical components connected, the translation stage was adjusted to load a single cell

into the injector structure region, and nuclear fluorescence of Hoechst was obtained by excitation

with a mercury source (BH2-RFL-T3, Olympus, Japan) filtered by an ET-DAPI filter set

(Chroma Technology Corp., Bellows Falls, VT, USA). If fluorescence was observed through the

same filter set, a video of the nuclear fluorescence of the cell was captured at 15fps for 30s using

the CoolSNAP Pro camera and ImagePro Plus software was employed to determine the effects of

photobleaching over the duration of the cell lysis experiments. If the nucleus exhibited no

VGEN = Regular voltage of function generator

VOUT = Final output voltage

Position A – Output BNC (front)

Position B – Modulation In BNC (back)

72

fluorescence, the flow was adjusted to steer the cell through the funnel in the injector structure

and another cell was loaded. Following Hoechst imaging, the cell was allowed to pass through

the injector structure, and another cell was loaded. Calcein was excited and monitored using the

same light source with a FITC-EGFP filter set (Chroma Technology Corp., Bellows Falls, VT,

USA). A video was captured of the cytoplasmic fluorescence at 15fps for 180s after which the

cell was flushed through the microchannel. While this experiment was intended to determine the

effects of photobleaching, it also serves as a control case in which cells were observed in the

absence of an electric field.

Electrical lysis of plasma membrane

The effects of electrical lysis of the plasma membrane, without hindrance to the nuclear

membrane was investigated by loading a single cell into the injector structure, and positioning it

in place at the funnel, described in Chapter 3 Section 3.2.1. Still images were collected of the

cell, and its diameter was determined using ImagePro Plus. A fluorescent image of the nuclear

stain was obtained and after changing filters to FITC-EGFP, a video of the cytoplasmic

fluorescence was recorded at 15fps for 4-5s, with the function generator turned on 0.6s after

recording had begun. The pulse duration was set to 100µs while the output voltage was 32V and

the remained on for the entire duration of the recording process. Immediately after the function

generator was turned off, the nuclear fluorescence was again imaged to determine the integrity of

the nucleus following the application of an electric field. This process was repeated for both 3T3

and 9L cells using 3 cells per microfluidic chip for 3 different chips, totalling 9 cells for each cell

line.

Additionally, a small number of cells from each cell line were subjected to lower (4 –

20V) and higher voltages (36 – 40V) to observe the effect on both the plasma membrane and

nucleus.

4.2.3 Data analysis

While the fluorescent images and videos obtained for the photobleaching of Hoechst offer a

qualitative perspective, a more quantitative analysis was required to determine the change of

intensity as a function of time. The videos were decompiled into individual frames using a video

processing utility, VirtualDub (VirtualDub.org, USA), and at frames corresponding to a 3s

73

interval the profile of the fluorescent intensity was plotted as a function of position along the y-

axis. Since the funnel of the injector structure allows cell confinement and restricts diffusion of

cellular content laterally along the x and z axes of the channel, the fluorescent profile along the

y-axis of the channel was investigated as illustrated in Fig. 4.5. ImageJ (National Institute of

Health, USA) was used to perform a background subtraction on each image and to extract the

intensity profile. This was repeated for the photobleaching effects of calcein, with the fluorescent

intensity profile extracted frames corresponding to 10s intervals.

Figure 4.5: Illustration of direction of fluorescent profile analysis

The images obtained of the nuclear staining before and after electrical lysis were

analysed in a manner similar to that described previously, however only at two time points, 0s

and 6s. Meanwhile, the analysis of the diffusion of cytoplasmic staining from within the cell was

performed similarly, with y-profiles obtained at time intervals of 0.2s and the FWHM of each

profile was determined as a function of time, permitting extraction of the diffusion rate of each

cell to be obtained.

74

4.3 Results and Discussion

4.3.1 Flow control

The fluidic control system, based on Bernoulli‟s principle, significantly reduced the flow rate in

the microfluidic channel and the ability to stop cells already in motion. Cell velocities between 1-

2 ± 0.5µm·s-1

were regularly achieved, corresponding to flow rates of 0.7-1.4 pl·s-1

, while

manipulation of the translation allowed cells to be stopped with better than 10µm accuracy

within the vicinity of the 140µm wide electrodes.

4.3.2 Hoechst and calcein staining and photobleaching

Hoechst 33342

Throughout all experiments, 9 out of every 10 cells exhibited Hoechst stain related fluorescence,

showing that the stain intercalates well with DNA when using the protocol described. Fig. 4.6 (a)

shows a number of nuclei exhibiting fluorescence due to Hoechst staining. The time period over

which the Hoechst DNA stain was excited during electrical lysis experiments ranged from 1-20s,

thus by observing the effects of photobleaching for a 30s period, it can be inferred whether the

FWHM changes seen after lysis attributed to the electric field enabled diffusion or

photobleaching.

Figure 4.6: a) Hoechst staining of nuclei in cell reservoir; b) Single cell Hoechst stain depicting line along which

fluorescent intensity profile

10µm

5µm

(a)

(b)

Line of

fluorescent

profile

75

Figure 4.7: a) Hoechst fluorescent intensity profile as a function of position for various time points; b) Area under

each intensity peak as a function of time

While there appears to be movement of the nucleus along the y-axis of the channel (Fig.4.7 (a)),

there is no significant photobleaching, as evidenced by a slope of -0.06364 ± 0.2664 s-1

for a line

of best fit to the data presented in Fig. 4.7 (b) as the 95% confidence interval contains zero. Thus

it can be concluded that for the time periods for which the Hoechst stain is excited during lysis

experiments (1-20s), photobleaching effects are negligible.

Calcein AM

The staining of the cytoplasm exhibited slightly lower efficacy compared to the nuclear staining,

with approximately 8 in 10 exhibiting calcein fluorescence (Fig. 4.8 (a)). This may have been

due to inadequate hydrolysis of the dye molecules by intracellular esterases. The extended time

duration over which photobleaching of calcein was observed was required since this dye was

excited for up to 90s during set-up and electrical lysis experiments.

(a) (b)

76

(a) (b)

Figure 4.8: a) Calcein staining of cytoplasm in cell reservoir; b) Single cell calcein stain depicting line along which

fluorescent intensity profile

Figure 4.9: a) Calcein fluorescent intensity profile as a function of position for various time points; b) Area under

each intensity peak as a function of time

The fluorescent intensity profiles of calcein at 60s intervals exhibit a change in peak intensity

(Fig. 4.9 (a)), resulting in a change of the total fluorescent intensity, confirmed in Fig. 4.9 (b). A

line of best fit yields a slope of -0.4179 ± 0.09572 s-1

, supporting photobleaching; however, for

20µm

20µm

Line of

fluorescent

profile

(a)

(b)

(a) (b)

77

the duration of electrical lysis experiments, these effects were considered insignificant as the

overall change of intensity over a 180s interval is only 2.9% as extracted from Fig. 4.9 (b).

4.3.3 Electrical lysis of 3T3 and 9L plasma membranes

3T3 cells

The average diameter of 3T3 cells was determined to be 10 ± 2µm and results indicated that at an

applied voltage of 32V, with 100µs pulse duration, producing an electric field in the range 4.507

– 4.532kVcm-1

, electrical lysis of the plasma membrane was evident. While video of the

cytoplasmic fluorescence provided visual evidence of this as shown in Fig. 4.10, an analysis of

the fluorescent intensity profile offered quantification of the process.

Figure 4.10: a) Fluorescent image of cytoplasm, a) before lysis, 0s; b) after lysis, 4s

Fluorescent intensity profiles extracted from videos of the electrical lysis process at 0.2s

intervals for 3 different cells from the same microfluidic chip is shown in Fig. 4.11 (left). Each

cell exhibits a decrease of maximum intensity and a broadening of the peak, both as a function of

time, and represents the diffusion of the calcium ions from the cytoplasm along the y-axis of the

microfluidic channel. Meanwhile, the corresponding change of the nuclei of each cell, as

represented by the Hoechst stain, from the start to finish of lysis can be seen in Fig. 4.11 (right).

The shift of the Hoechst intensity profile in some plots along the position axis is attributed to

10µm 10µm

(a) (b)

78

motion of the nuclei along the y-axis of the microfluidic channel following lysis. Compared to

the Hoechst stain, the calcein stain appears to exhibit weaker fluorescence, highlighted by the

difference in peak intensities between the two. While this was not evident in cells in all

microfluidic chips, this outcome can be attributed to poor hydrolysis of calcein AM by

intracellular esterases.

Figure 4.11: Fluorescent intensity profile as a function of position for 3 different 3T3 cells on the same chip

showing cytoplasmic (left column) and nucleic (right column) changes over time between t = 0s and t = 6s

Calcein Hoechst

Cell 1

Cell 2

Cell 3

79

Figure 4.12: Plot of FWHM vs. Time, showing diffusion region and start of electric field application

Extracting the FWHM of each profile as a function of time is presented in Fig. 4.12,

showing first a region of no change (A), where there is no electric field applied, to a region of

linear increase representing cytoplasmic diffusion (B), and a saturation region where there is no

further change (C). This general behaviour was observed in all cells for all plots of FWHM as a

function of time on each of the chips interrogated.

Since it is difficult to determine at what exact time point that lysis occurs, it is safe to

assume that it is between the onset of the electric field application and the first rising point of the

slope, which was 200 ± 100 ms for the 9 cells that were investigated. The diffusion rate of

calcein was determined by the slope of a linear regression on the region denoted B. This is

directly related to the formation of non resealable pores on the plasma membrane as discussed in

Chapter 2 Section 2.1.2. Meanwhile, the saturation point, where there is no further diffusion of

calcein, occurs 1.7 ± 0.2s after the electric field was turned on. Table 4.1 summarizes the

diffusion rates of each cell in each of the microfluidic chips investigated for this project.

A

B

C

Start point of electric field

80

Property

Chip 1 Chip 2 Chip 3

Cell 1 Cell 2 Cell 3 Cell1 Cell 2 Cell 3 Cell 1 Cell 2 Cell 3

Diffusion

Rate

(µm·s-1

)

5.712 ±

0.7979

6.062 ±

0.9183

5.364 ±

0.8837

4.061 ±

0.1350

3.665 ±

0.1560

4.522 ±

0.3022

5.316±

0.4791

5.790±

0.6033

5.703±

0.6786

Total

Fluorescent

Intensity of

Hoechst

Stain before

lysis

508.8 478.1 702.5 645.3 711.2 703.0 700.5 708.2 703.9

Total

Fluorescent

Intensity of

Hoechst

Stain after

lysis

498.8 463.1 681.9 633.1 690.8 689.9 689.6 692.0 681.5

% Change in

Hoechst

Fluorescence

2.0 3.1 2.9 1.9 2.9 1.9 1.6 2.3 3.2

Table 4.1: Summary of diffusion rates and fluorescent intensity data for nine 3T3 cells

The average diffusion rate was 5.133 ± 0.277 µm·s-1

, while the FWHM became

asymptotic at 20.23 ± 2.15 µm·s-1

after the saturation point was reached. This supports the

hypothesis that within the experimental time interval, cells undergo significant rupturing of the

plasma membrane due to electrical lysis, creating permanent pores that are not re-sealable. As a

result, the calcein was expelled and diffused along the y-axis of the fluidic channel although

there was no continuous flow present, until a saturation point was reached. Meanwhile, the

average loss of Hoechst fluorescent intensity was 2.4% between the start of the electrical lysis

81

process and the end of the observed saturation region. This suggests that while the plasma

membrane was ruptured, evident by the expulsion of calcein from the cell, the nuclei remained

largely intact with no diffusion of the Hoechst stain observed.

9L cell

The disease model 9L cells had an average diameter of 8.5 ± 2µm and exhibited signs of

membrane lysis when a voltage of 32V, with 100µs pulse duration, producing an electric field in

the range 4.507 – 4.532kVcm-1

. Fig. 4.13 (left) shows the results of the fluorescent intensity

profiles for 3 different cells on a single microfluidic device via analysis of video frames

extracted at 0.2s intervals.

82

Figure 4.13: Fluorescent intensity profile as a function of position for 3 different 9L cells on the same chip showing

cytoplasmic (left column) and nucleic (right column) changes over time between t = 0s and t = 6s

Calcein Hoechst

Cell 1

Cell 2

Cell 3

83

The plots indicate that while the profiles of the fluorescent intensity of calcein in the

cytoplasm exhibit peak broadening and a decrease in maximum intensity, both as a function of

time, the general Gaussian appearance of the profiles remains intact. This is possibly due to

incomplete poration of the plasma membrane, resulting in higher retention of calcein than that

observed in 3T3 cells. Fig. 4.13 (right) shows that over the duration of electrical lysis, the

fluorescent intensity profiles of Hoechst in the nuclei maintain their shape, with miniscule shifts

along the position axis, representing the movement of the nuclei along the y-axis of the channel.

The FWHM of each profile at 0.2s intervals (Fig. 4.14) shows an initial region, before

application of the electric field, A, region of diffusion, B, and saturation region, C. Fluorescent

intensity profiles and FWHM plots of the nine 9L cells investigated on 3 different microfluidic

chips were all observed to adhere to a similar pattern.

Figure 4.14: Plot of FWHM vs. Time, showing diffusion region and start of electric field application

While it is again difficult to determine the exact time of lysis, Fig. 4.14 suggests that lysis

occurs fractionally earlier in 9L cells than was observed for 3T3 cells, which was about 150 ±

100 ms after the application of the electric field for the 9 cells that were investigated. The

diffusion rate of calcein was determined by the same methods used for 3T3 analysis and was

found to reach saturation 2.3 ± 0.2s after application of the electric field. Table 4.2 summarizes

B

A

Start point of electric field

C

84

the diffusion rates of each cell in each of the microfluidic chips investigated for this project.

Property

Chip 1 Chip 2 Chip 3

Cell 1 Cell 2 Cell 3 Cell1 Cell 2 Cell 3 Cell 1 Cell 2 Cell 3

Diffusion

Rate

(µm·s-1

)

4.964±

0.532

4.155 ±

0.572

4.022 ±

0.544

4.268 ±

0.645

4.502 ±

0.568

4.195 ±

0.497

4.226±

0.475

3.829±

0.435

4.162±

0.674

Total

Fluorescent

Intensity of

Hoechst

Stain before

lysis

818.1 785.3 793.5 750.5 780.8 744.8 707.1 748.1 740.3

Total

Fluorescent

Intensity of

Hoechst

Stain after

lysis

797.4 777.5 780.9 731.8 763.8 719.3 682.6 722.4 726.8

% Change in

Hoechst

Fluorescence

2.5 1.0 1.6 2.5 2.2 3.4 3.4 3.4 1.8

Table 4.2: Summary of diffusion rates and fluorescent intensity data for nine 9L cells

The average diffusion rate was 4.258 ± 0.403 µm·s-1

, while the FWHM became

asymptotic at 18.89 ± 1.82 µm·s-1

after the saturation point was reached. This further supports

the hypothesis that within the experimental time interval, non resealable pores are created on the

plasma membrane, evident by the expulsion of calcein from the cell under zero flow rates. The

average loss of Hoechst fluorescent intensity was 2.4% between the start of the electrical lysis

85

process and the end of the observed saturation region, giving further evidence of nuclear

membrane intactness.

4.3.4 Variation of diffusion rate due to electric field

Observations and analysis of 3T3 cells under the influence of electric fields expected to be too

low to induce lysis, and fields higher than that used for all other work in this thesis, were

performed in the same manner as described above for a fixed field strength. The values of the

mean electric field were obtained from simulations in Chapter 2, given in Table 2.1, while the

diffusion rates were averaged for 3 cells at each applied electric field. Fig. 4.15 illustrates that at

low field strengths, there appears to be no diffusion of calcein, while the small amounts shown

may be attributed to electroporation. At higher field strengths, the diffusion rate increases

linearly with the applied electric field; however, due to limitations of the electronic set up, fields

in excess of 10kV·cm-1

were not investigated. It should be expected that at fields at and above

this value, the entire cell, with a large majority of its organelles, including the nucleus, would be

lysed, leading to a saturation point in the diffusion rate.

Figure 4.15: Dependency of diffusion rate of calcein on mean electric field strength for 3T3 cells

86

4.4 Conclusion

Both models demonstrated the expulsion of calcein labelled molecules from the cell upon

application of the electric field, and since there was no flow present in the fluidic channel during

lysis, the diffusion of calcein along the y-axis of the channel could be attributed to a pressure

gradient existing between the inner and outer regions of the cell. Comparison of Fig 4.11 and

Fig. 4.13, along with experimental data for other microfluidic chips, suggest that while 3T3 cells

appear to expel a significant amount of calcein, evident by the loss defined peaks while the

FWHM increases, calcein in 9L cells maintain a distinct Gaussian profile, suggesting that some

of the calcein is still held within the lysed cell. This is most likely due to inadequate pore

creation on the surface of the plasma membrane. Intactness of the nuclear membrane was evident

in both types of cells due to the retention of > 95% of the Hoechst stain within their nuclei.

87

Chapter 5 Summary and Future Work

5.1 Summary

The goal of this thesis was to develop a microfluidic device with integrated electrodes, capable

of performing selective electrical lysis of the plasma membrane of single cells, while leaving the

nuclear membrane intact. An understanding of the mechanism of irreversible electroporation and

the electrical requirements to achieve lysis of only the plasma membrane provided the basis to

achieve this goal.

Due to their variation in size and membrane charging time, the electric field required to

surpass a threshold transmembrane potential or transorganelle potential which mediates lysis, is

significantly different for plasma membranes and those of intracellular organelles, with the latter

requiring a much high electric field to induce lysis. While this concept has been theoretically

investigated [28], there is limited experimental evidence, especially with respect to single cells.

Microfluidic devices afford the opportunity to investigate single cells, while restricting the

aberrant diffusion of cell contents, thus enabling the ability to determine whether selective lysis

is effective.

Performing electrical lysis of single cells within a microfluidic device has previously

been investigated [28, 51]; however, other systems lack the ability to separate the electrode from

either the extracellular media or the cell itself, which often results in thermal or mechanical

damage to cells coming into contact with electrodes. Additionally, due to the geometrical and

electrical limitations of these devices, cells are usually lysed in an uncontrolled manner, with no

regard for selectivity, and cell contents are diffused throughout microchannels due to lack of

cellular confinement, making downstream analysis difficult. The microfluidic device that we

have presented, addresses many of the shortcomings of previous systems, while providing

evidence of selective lysis of the plasma membrane and negating thermal damage to the cell.

88

5.2 Contributions and perspectives

5.2.1 Microfluidic device fabrication for single cell electrical lysis

The current available microfluidic techniques to perform single cell electrical lysis can be

improved by geometric considerations and positioning of electrodes. In the fabrication of the

microfluidic device used in this project, the following contributions were made:

1. Improvement on UV photolithography of SU-8 using a plastic photo mask to

produce aspect ratios of 1:3 and greater, comparable to published reports [80, 102].

The use of glycerol as an index matching and gap compensating media,

previously demonstrated by other groups, coupled with high quality photo masks,

enabled minimum feature widths of 5.5 ± 1.5µm with 3% magnification from

mask to SU-8. However, feature widths can be significantly minimized by using

quartz-chrome masks, which are typically manufactured by laser etching, and

does not require any compensating media.

2. Development of geometrical design which allows integrated electrodes. By taking

advantage of the improved width resolution, a microfluidic chip was designed to

enable electrodes to be offset in the x-axis from fluidic channels and buffered by

PDMS walls to prevent electrode – cell interactions.

3. Integration of In microelectrodes in close proximity to fluidic channel. While Siegel

et al [87] demonstrated the ability to integrate molten solder into PDMS to fabricate

electromagnets, this method was never investigated for microelectrodes for electrical

lysis. Due to the tediousness and time required to work with molten solder at ~

2000C, alternative electrode material should be explored to speed up fabrication time

of the microfluidic system.

89

5.2.2 Selective electrical lysis of plasma membrane of single cells

Theoretical models have shown that with the application of either a DC or AC electric field,

having specific electrical parameters as discussed in Chapter 2, it is possible to lyse the plasma

membrane of a cell, while the membranes of intracellular organelles remain intact.

Experimentally, this theory has not been thoroughly investigated, especially in microfluidics,

while other lysis methods have demonstrated this ability. The following contributions and

observations were made through in vitro studies in this project:

1. Demonstrated the ability to perform selective electrical lysis of the plasma membrane

of single 3T3 and 9L cells, supported by pre and post lysis analysis of the

fluorescently labelled cytoplasm and nuclei. The novel approach of integrating

microelectrodes in close proximity to cells, without coming into contact with cells,

allowed the plasma membrane to be electrically lysed, with visual evidence of the

cytoplasm exiting the cell, while the nuclei remained intact throughout the process.

However, further labelling of other intracellular organelles of interest and upstream

biochemical analysis would be necessary to further refine and validate the lysis

parameters.

2. As a result of the confinement provided by the injector structure, and the theorized

uniform pore creation on the surface of the plasma membrane, relatively low

diffusion rates were achieved, which is desirable for downstream analysis by capillary

electrophoresis in combination with LIF.

5.3 Future Work

5.3.1 Engineering and fabrication aspects

While this project demonstrated the ability to perform selective lysis of the plasma membrane,

the injector structure is incapable of efficiently separating the cytoplasm from the nuclei. The

narrow funnel of the injector structure tapers to a 5.5µm channel; however, a typical nuclei is

~2µm, thus under the influence of flow, would be inseparable from the cytoplasm and other

nuclei. Micro pillars etched into cured SU-8 by excimer laser ablation [103] is one option to

fabricate a sieve like region within the injector structure as an alternative to having an expensive

90

quartz-chrome mask. This would allow the cytoplasm to be separated from the nuclei, and

depending on the spacing of the pillars, can also separate smaller organelles, such as the

mitochondria, from the cytoplasm.

A new photo mask design is necessary to create a new microfluidic device which will

permit high throughput analysis by parallelizing the selective lysis process, similar to parallel

systems demonstrated by Munce et al [24]. Prior to designing a new mask, an estimation of the

minimum distance between fluidic channels, and the effect of electric fields on parallel systems

would be necessary, possibly using COMSOL simulations. The design shown in Fig. 5.2 shows a

parallel system; however, the distances between channels have not been optimized as constraints

provided by LIF detection system will also have to be considered.

The use of In molten solder, while yielding high electrically conductive microelectrodes,

is often tedious and time consuming due to the injection process. In instances where the electrode

filling channels are not properly silanized, the solder has been observed to not fill the channel

entirely, while excess solder on the PDMS surface can sometimes result in darkening of the

PDMS. Additionally, solid solder particulates have been observed to contaminate the cell

reservoir, leading to clogging of the fluidic channel. Upon the completion of this project, a liquid

metal alloy, eutectic GaIn, with melting point ~ 15.50C, was found in the literature [104] to form

stable structures in microchannels, and should be further investigated as an alternative electrode

material. The alloy reportedly fills channels without the need for pre-treatment and can be

removed and re-used in other electrodes, while filled channels can be stored at 40C.

5.3.2 Integration of components

The flow control system used in this project was sufficient to allow a randomized, single cell to

be selected from the cell reservoir; however, optimization of the optical tweezers available as

part of this program, could allow a specific cell to be selected and transported to the injector

structure. Currently, the force produced by the tweezers is not adequate to move a cell over a

large distance, with cells becoming dislodged from the optical trap when coming into contact

with channel walls or other cells.

With the parallelization of the injector structures and electrodes, optical tweezers would

again be inadequate to quickly load multiple cells, thus the flow system can again be employed,

or alternatively, the use of syringe pumps capable of producing the necessary flow rates.

91

However, loading multiple cells flowing along a channel into separate injector structures would

be difficult to control by flow alone, thus optical waveguides, as shown in Fig. 5.2, also

developed as part of this program, would be required to load cells into injector structures for

selective lysis.

Figure 5.1: Parallelized system with multiple electrode filling ports

The information content available through fluorescent analysis, while sufficient to give

evidence of selective lysis, is limited when greater detail is required. Capillary electrophoresis

would provide additional biochemical information from lysed cells and, with new microfluidic

designs and possibly the use of microvalves, can be integrated along with the current lysis

system. Channel lengths and widths will need to be optimized to produce acceptable

electrophoretic resolution as the narrow 5.5µm channel following the injector structure would

result in poor separation of analytes, thus poor resolution. Initial work performed by Pelzer [105]

on the development of a fibre array for LIF detection, specifically for parallel channels, have

shown promising results.

200µm

92

5.3.3 Selective lysis by AC electric field

An electric field generated by a DC source was able to selectively lyse the plasma membrane;

however, as discussed in Chapter 2, to sequentially lyse the membrane, nucleus and other

intracellular organelles, AC fields of varying applied frequencies would be required.

Additionally, an AC field with long pulse duration is capable of generating heat and possibly

inducing denaturation of organelles and lipids if required for future lysis steps.

5.3.4 Multiple fluorescent staining

Evidence of cytoplasmic diffusion and nucleic intactness, following selective membrane lysis,

was available by observing calcein and Hoechst fluorescence for the cytoplasm and nucleus

respectively. However, to determine the effects of the lysing electric field on other intracellular

organelles, such as the mitochondria, additional fluorescent labelling would be required, while

staining of the plasma membrane would enable observation of the membrane integrity post lysis.

Initial triple staining of the cytoplasm with calcein, nucleus with Hoechst and mitochondria with

Mitotracker Red (Sigma-Aldrich Ltd., Oakville, ON, Canada) has shown promising results (Fig

5.2 (a)), while staining of the plasma membrane with Rhodamine B chloride-R18 (Sigma-

Aldrich Ltd., Oakville, ON, Canada) instead of the mitochondria can be improved as the stain

appeared to enter the cytoplasm of most cells (Fig. 5.3 (b)).

Figure 5.2: (a) Staining of nucleus with Hoechst and mitochondria with Mitotracker Red; (b) Triple staining of

cytoplasm (Calcein AM), nucleus (Hoechst) and plasma membrane (R18)

(a) (b)

3µm 8µm

10µm 10µm

(a) (b)

93

Bibliography

[1] D. Cohen, J. Dickerson, C. Whitmore et al., “Chemical Cytometry: Fluorescence-Based

Single-Cell Analysis,” Annual Review of Analytical Chemistry, 1( ), pp. 165-190, 2008.

[2] C. P. Hickman, L. Roberts, and A. Larson. "The Eukaryotic Cell," April 26, 2010;

http://hyperphysics.phy-astr.gsu.edu/hbase/biology/cell.html.

[3] J. C. Kendrew, G. Bodo, H. M. Dintzis et al., “A Three-Dimensional Model of the

Myoglobin Molecule Obtained by X-Ray Analysis,” Nature, 181(4610), pp. 662-666,

1958.

[4] K. Tanaka, H. Waki, Y. Ido et al., “Protein and polymer analyses up to m/z 100 000 by

laser ionization time-of-flight mass spectrometry,” Rapid Commun. Mass Spectrom, 2(8),

pp. 151–153, 1988.

[5] K. Hu, H. Ahmadzadeh, and S. Krylov, “Asymmetry between sister cells in a cancer cell

line revealed by chemical cytometry,” Analytical Chemistry, 76(13), pp. 3864-3866,

2004.

[6] D. Di Carlo, and L. Lee, “Dynamic single-cell analysis for quantitative biology,”

Analytical Chemistry, 78(23), pp. 7918, 2006.

[7] G. Brady, “Expression profiling of single mammalian cells-small is beautiful,” Yeast,

17(3), pp. 211-217, 2000.

[8] J. Dick, “Breast cancer stem cells revealed,” Proceedings of the National Academy of

Sciences, 100(7), pp. 3547, 2003.

[9] A. Schmid, H. Kortmann, P. Dittrich et al., “Chemical and biological single cell

analysis,” Current Opinion in Biotechnology, 1( ), pp. 165-190, 2008.

[10] F. Kamme, R. Salunga, J. Yu et al., “Single-cell microarray analysis in hippocampus

CA1: demonstration and validation of cellular heterogeneity,” Journal of Neuroscience,

23(9), pp. 3607, 2003.

[11] L. P. Lee, and D. Di Carlo, "Single Cell Analysis for Quantitative Systems Biology,"

Single Cell Analysis - Technologies and Applications, D. Anselmetti, ed., pp. 135-158,

Weinheim, Germany: Wiley-Blackwell, 2009.

[12] C. V. Rao, D. M. Wolf, and A. P. Arkin, “Control, exploitation and tolerance of

intracellular noise,” Nature, 420(6912), pp. 231-237, 2002.

[13] E. A. Arriaga, "Single Cell Heterogeneity," Single Cell Analysis - Technologies and

Applications, D. Anselmetti, ed., pp. 223-234, Weinheim, Germany: Wiley-Blackwell,

2009.

94

[14] H. Yan, B. Zhang, and H. Wu, “Chemical cytometry on microfluidic chips,”

Electrophoresis, 29(9), pp. 1775-1786, 2008.

[15] J. Lichtman, and J. Conchello, “Fluorescence microscopy,” Nature Methods, 2(12), pp.

910, 2005.

[16] G. Helmlinger, M. Endo, N. Ferrara et al., “Growth factors: Formation of endothelial cell

networks,” Nature, 405(6783), pp. 139-141, 2000.

[17] R. Yuste, “Fluorescence microscopy today,” Nature Methods, 2(12), pp. 902-904, 2005.

[18] R. Schultz, T. Nielsen, J. Zavaleta et al., “Hyperspectral imaging: a novel approach for

microscopic analysis,” Cytometry, 43(4), pp. 239-247, 2001.

[19] L. Borland, S. Kottegoda, K. Phillips et al., “Chemical analysis of single cells,” Annual

Review of Analytical Chemistry, 1( ), pp. 191-227, 2008.

[20] X. Huang, M. Quesada, and R. Mathies, “DNA sequencing using capillary array

electrophoresis,” Analytical Chemistry, 64(18), pp. 2149-2154, 1992.

[21] D. Michels, S. Hu, R. Schoenherr et al., “Fully automated two-dimensional capillary

electrophoresis for high sensitivity protein analysis,” Molecular & Cellular Proteomics,

1(1), pp. 69-74, 2002.

[22] G. Whitesides, “The origins and the future of microfluidics,” Nature, 442(27), pp. 368-

373, 2006.

[23] D. Irimia, R. Tompkins, and M. Toner, “Single-cell chemical lysis in picoliter-scale

closed volumes using a microfabricated device,” Analytical Chemistry, 76(20), pp. 6137-

6143, 2004.

[24] N. Munce, J. Li, P. Herman et al., “Microfabricated system for parallel single-cell

capillary electrophoresis,” Analytical Chemistry, 76(17), pp. 4983-4989, 2004.

[25] X. Hu, P. Bessette, J. Qian et al., “Marker-specific sorting of rare cells using

dielectrophoresis,” Proceedings of the National Academy of Sciences, 102(44), pp.

15757, 2005.

[26] M. He, J. Edgar, G. Jeffries et al., “Selective encapsulation of single cells and subcellular

organelles into picoliter-and femtoliter-volume droplets,” Analytical Chemistry, 77(6),

pp. 1539-1544, 2005.

[27] A. Valero, J. Post, J. Nieuwkasteele et al., “Gene transfer and protein dynamics in stem

cells using single cell electroporation in a microfluidic device,” Lab on a Chip, 8(1), pp.

62-67, 2008.

[28] H. Lu, M. Schmidt, and K. Jensen, “A microfluidic electroporation device for cell lysis,”

Lab on a Chip, 5(1), pp. 23-29, 2005.

95

[29] J. Godin, C. Chen, S. Cho et al., “Microfluidics and photonics for Bio-System-on-a-Chip:

A review of advancements in technology towards a microfluidic flow cytometry chip,”

Journal of Biophotonics, 1(5), pp. 355, 2008.

[30] D. Huh, W. Gu, Y. Kamotani et al., “Review: Microfluidics for flow cytometric analysis

of cells and particles,” Physiological Measurement, 26( ), pp. R73-R98, 2005.

[31] W. Huang, F. Ai, Z. Wang et al., “Recent advances in single-cell analysis using capillary

electrophoresis and microfluidic devices,” Journal of Chromatography B, 866(1-2), pp.

104-122, 2008.

[32] B. Huang, H. Wu, D. Bhaya et al., “Counting low-copy number proteins in a single cell,”

Science, 315(5808), pp. 81, 2007.

[33] R. Joshi, and K. Schoenbach, “Electroporation dynamics in biological cells subjected to

ultrafast electrical pulses: a numerical simulation study,” Physical review E, 62(1), pp.

1025-1033, 2000.

[34] J. Weaver, “Electroporation of biological membranes from multicellular to nano scales,”

IEEE Transactions on Dielectrics and Electrical Insulation, 10(5), pp. 754-768, 2003.

[35] S. Ho, and G. Mittal, “Electroporation of cell membranes: a review,” Critical reviews in

biotechnology, 16(4), pp. 349-362, 1996.

[36] H. Tien, and A. Ottova, “The bilayer lipid membrane (BLM) under electrical fields,”

IEEE Transactions on Dielectrics and Electrical Insulation, 10(5), pp. 717-727, 2003.

[37] M. Davidson. "Plasma Membrane," April 7, 2010;

http://micro.magnet.fsu.edu/cells/plasmamembrane/plasmamembrane.html.

[38] J. Weaver, K. Powell, R. Mintzer et al., “The electrical capacitance of bilayer

membranes:: The contribution of transient aqueous pores,” Bioelectrochemistry and

Bioenergetics, 12(3-4), pp. 393-404, 1984.

[39] J. Weaver, and Y. Chizmadzhev, “Theory of electroporation: a review,”

Bioelectrochemistry and Bioenergetics, 41(2), pp. 135-160, 1996.

[40] C. Grosse, and H. Schwan, “Cellular membrane potentials induced by alternating fields,”

Biophysical Journal, 63(6), pp. 1632-1642, 1992.

[41] W. Krassowska, and P. Filev, “Modeling electroporation in a single cell,” Biophysical

Journal, 92(2), pp. 404-417, 2007.

[42] M. Prausnitz, V. Bose, R. Langer et al., “Electroporation of mammalian skin: a

mechanism to enhance transdermal drug delivery,” Proceedings of the National Academy

of Sciences, 90(22), pp. 10504, 1993.

[43] H. Wang, and C. Lu, “Electroporation of mammalian cells in a microfluidic channel with

geometric variation,” Analytical Chemistry, 78(14), pp. 5158-5164, 2006.

96

[44] D. Lee, and Y. Cho, “A continuous electrical cell lysis device using a low dc voltage for

a cell transport and rupture,” Sensors and Actuators B: Chemical, 124(1), pp. 84-89,

2007.

[45] M. Berezovski, T. Mak, and S. Krylov, “Cell lysis inside the capillary facilitated by

transverse diffusion of laminar flow profiles (TDLFP),” Analytical and bioanalytical

chemistry, 387(1), pp. 91-96, 2007.

[46] R. Brown, and J. Audet, “Current techniques for single-cell lysis,” Journal of The Royal

Society Interface, 5(Suppl 2), pp. S131, 2008.

[47] Y. Kim, J. Kang, S. Park et al., “Microfluidic biomechanical device for compressive cell

stimulation and lysis,” Sensors and Actuators B: Chemical, 128(1), pp. 108-116, 2007.

[48] D. Carlo, K. Jeong, and L. Lee, “Reagentless mechanical cell lysis by nanoscale barbs in

microchannels for sample preparation,” Lab on a Chip, 3(4), pp. 287-291, 2003.

[49] H. Zhang, and W. Jin, “Determination of different forms of human interferon-gamma in

single natural killer cells by capillary electrophoresis with on-capillary immunoreaction

and laser-induced fluorescence detection,” Electrophoresis, 25(7-8), pp. 1090-1095,

2004.

[50] M. McClain, C. Culbertson, S. Jacobson et al., “Microfluidic devices for the high-

throughput chemical analysis of cells,” Analytical Chemistry, 75(21), pp. 5646-5655,

2003.

[51] Y. Huang, and B. Rubinsky, “Microfabricated electroporation chip for single cell

membrane permeabilization,” Sensors & Actuators: A. Physical, 89(3), pp. 242-249,

2001.

[52] Y. Nashimoto, Y. Takahashi, T. Yamakawa et al., “Measurement of gene expression

from single adherent cells and spheroids collected using fast electrical lysis,” Analytical

Chemistry, 79(17), pp. 6823-6830, 2007.

[53] S. Lee, and Y. Tai, “A micro cell lysis device,” Sensors & Actuators: A. Physical, 73(1-

2), pp. 74-79, 1999.

[54] L. Jang, and M. Wang, “Microfluidic device for cell capture and impedance

measurement,” Biomedical Microdevices, 9(5), pp. 737-743, 2007.

[55] M. Fox, D. Esveld, A. Valero et al., “Electroporation of cells in microfluidic devices: a

review,” Analytical and bioanalytical chemistry, 385(3), pp. 474-485, 2006.

[56] Dow-Corning, "Information about Dow Corning brand silicone encapsulants," Product

Information, Dow Corning Corporation, 2005, pp. 4-5.

[57] D. Carlo, L. Wu, and L. Lee, “Dynamic single cell culture array,” Lab on a Chip, 6(11),

pp. 1445-1449, 2006.

97

[58] P. Marc, C. Sims, M. Bachman et al., “Fast-lysis cell traps for chemical cytometry,” Lab

on a Chip, 8(5), pp. 710-716, 2008.

[59] M. Xu, J. Li, L. Lilge et al., “F 2-laser patterning of indium tin oxide (ITO) thin film on

glass substrate,” Applied Physics A: Materials Science & Processing, 85(1), pp. 7-10,

2006.

[60] MicroChem, "Processing Guidelines for SU-8 2025, SU-8 2035, SU-8 2050 and SU-8

2075," SU-8 2000 Permanent Epoxy Negative Photoresist, MicroChem Inc., 2007, pp. 1-

5.

[61] R. Jackman, S. Brittain, A. Adams et al., “Design and fabrication of topologically

complex, three-dimensional microstructures,” Science, 280(5372), pp. 2089, 1998.

[62] C. Lin, G. Lee, B. Chang et al., “A new fabrication process for ultra-thick microfluidic

microstructures utilizing SU-8 photoresist,” Journal of Micromechanics and

Microengineering, 12(5), pp. 590-597, 2002.

[63] D. C. Duffy, J. C. McDonald, O. J. A. Schueller et al., “Rapid Prototyping of

Microfluidic Systems in Poly(dimethylsiloxane),” Analytical Chemistry, 70(23), pp.

4974-4984, 1998.

[64] W. Dai, K. Lian, and W. Wang, “A quantitative study on the adhesion property of cured

SU-8 on various metallic surfaces,” Microsystem Technologies, 11(7), pp. 526-534, 2005.

[65] N. Toriello, E. Douglas, and R. Mathies, “Microfluidic device for electric field-driven

single-cell capture and activation,” Langmuir, 19(1532-1538, 2003.

[66] M. Agirregabiria, F. J. Blanco, J. Berganzo et al., “Fabrication of SU-8 multilayer

microstructures based on successive CMOS compatible adhesive bonding and releasing

steps,” Lab on a Chip, 5( ), pp. 545-552, 2005.

[67] A. Mata, A. Fleischman, and S. Roy, “Characterization of polydimethylsiloxane (PDMS)

properties for biomedical micro/nanosystems,” Biomedical Microdevices, 7(4), pp. 281-

293, 2005.

[68] C. McDonald, D. C. Duffy, J. R. Anderson et al., “Fabrication of microfluidic systems in

poly(dimethylsiloxane),” Electrophoresis, 21(1), pp. 27-40, 2000.

[69] H. Becker, and U. Heim, “Hot embossing as a method for the fabrication of polymer high

aspect ratio structures,” Sensors & Actuators: A. Physical, 83(1-3), pp. 130-135, 2000.

[70] I. Sohn, M. Ko, Y. Kim et al., “Direct femtosecond laser lithography for photoresist

patterning,” Optical Engineering, 48(024301, 2009.

[71] J. Anderson, D. Chiu, R. Jackman et al., “Fabrication of topologically complex three-

dimensional microfluidic systems in PDMS by rapid prototyping,” Analytical Chemistry,

72(14), pp. 3158-3164, 2000.

98

[72] Y. Liu, J. Fanguy, J. Bledsoe et al., “Dynamic coating using polyelectrolyte multilayers

for chemical control of electroosmotic flow in capillary electrophoresis microchips,”

Analytical Chemistry, 72(24), pp. 5939-5944, 2000.

[73] C. Lin, G. Lee, Y. Lin et al., “Fabrication of microfluidic systems on soda-lime glass,”

Journal of Micromechanics and Microengineering, 11( ), pp. 726-732, 2001.

[74] K. J. Seu, A. P. Pandey, F. Haque et al., “Effect of Surface Treatment on Diffusion and

Domain Formation in Supported Lipid Bilayers,” Biophysical Journal, 92(7), pp. 2445-

2450, 2007.

[75] E. Conradie, and D. Moore, “SU-8 thick photoresist processing as a functional material

for MEMS applications,” Journal of Micromechanics and Microengineering, 12(4), pp.

368-374, 2002.

[76] A. Del Campo, and C. Greiner, “SU-8: a photoresist for high-aspect-ratio and 3D

submicron lithography,” Journal of Micromechanics and Microengineering, 17(6), pp.

81, 2007.

[77] J. Carlier, S. Arscott, V. Thomy et al., “Integrated microfluidics based on multi-layered

SU-8 for mass spectrometry analysis,” Journal of Micromechanics and

Microengineering, 14( ), pp. 619-624, 2004.

[78] R. L. Barber, M. K. Ghantasala, R. Divan et al., “Study of stress and adhesion strength in

SU-8 resist layers on silicon substrate with different seed layers,” Journal of

Micro/Nanolithography, MEMS and MOEMS, 6(3), pp. 033006-9, 2007.

[79] J. Zhang, K. Tan, G. Hong et al., “Polymerization optimization of SU-8 photoresist and

its applications in microfluidic systems and MEMS,” Journal of Micromechanics and

Microengineering, 11( ), pp. 20-26, 2001.

[80] Y. Chuang, F. Tseng, and W. Lin, “Reduction of diffraction effect of UV exposure on

SU-8 negative thick photoresist by air gap elimination,” Microsystem Technologies, 8(4),

pp. 308-313, 2002.

[81] W. Kang, E. Rabe, S. Kopetz et al., “Novel exposure methods based on reflection and

refraction effects in the field of SU-8 lithography,” Journal of Micromechanics and

Microengineering, 16( ), pp. 821-831, 2006.

[82] F. Schneider, J. Draheim, R. Kamberger et al., “Process and material properties of

polydimethylsiloxane (PDMS) for Optical MEMS,” Sensors and Actuators A: Physical,

151(2), pp. 95-99, 2009.

[83] S. Bhattacharya, a. Datta, J. M. Berg et al., “Studies on surface wettability of

poly(dimethyl) siloxane (PDMS) and glass under oxygen-plasma treatment and

correlation with bond strength,” Journal of Microelectromechanical Systems, 14(3), pp.

590-597, 2005.

99

[84] K. Haubert, T. Drier, and D. Beebe, “PDMS bonding by means of a portable, low-cost

corona system,” Lab on a Chip, 6( ), pp. 1548-1549, 2006.

[85] X. Niu, S. Peng, L. Liu et al., “Characterizing and patterning of PDMS-based conducting

composites,” Advanced Materials, 19( ), pp. 2682–2686, 2007.

[86] N. Probst, and E. Grivei, “Structure and electrical properties of carbon black,” Carbon,

40(2), pp. 201-205, 2002.

[87] A. Siegel, S. Shevkoplyas, D. Weibel et al., “Cofabrication of electromagnets and

microfluidic systems in poly (dimethylsiloxane),” ANGEWANDTE CHEMIE-

INTERNATIONAL EDITION IN ENGLISH-, 45(41), pp. 6877, 2006.

[88] M. Liu, J. Sun, and Q. Chen, “Influences of heating temperature on mechanical properties

of polydimethylsiloxane,” Sensors and Actuators A: Physical, 151(1), pp. 42-45, 2009.

[89] D. Halliday, R. Resnick, and K. Krane, Physics Volume 2, 5 ed., pp. 963-967, New York:

John Wiley & Sons Inc., 2002.

[90] J. McDonald, and G. Whitesides, “Poly (dimethylsiloxane) as a material for fabricating

microfluidic devices,” Acc. Chem. Res, 35(7), pp. 491-499, 2002.

[91] A. Wheeler, W. Throndset, R. Whelan et al., “Microfluidic device for single-cell

analysis,” Analytical Chemistry, 75(14), pp. 3581-3586, 2003.

[92] M. Ozkan, T. Pisanic, J. Scheel et al., “Electro-optical platform for the manipulation of

live cells,” Langmuir, 19(5), pp. 1532-1538, 2001.

[93] J. George III, Y. Ahmad, D. Varghai et al., “Pc 4 photodynamic therapy of U87-derived

human glioma in the nude rat,” Lasers in Surgery and Medicine, 36(5), pp. 383-389,

2005.

[94] A. Ghods, D. Irvin, G. Liu et al., “Spheres isolated from 9L gliosarcoma rat cell line

possess chemoresistant and agressive cancer stem-like cells,” Stem Cells, 25(7), pp.

1645-1653, 2007.

[95] G. Roman, M. Wang, K. Shultz et al., “Sampling and Electrophoretic Analysis of

Segmented Flow Streams Using Virtual Walls in a Microfluidic Device,” Analytical

Chemistry, 80(21), pp. 8231-8238, 2008.

[96] H. Suzuki, and R. Yoneyama, “Integrated microfluidic system with electrochemically

actuated on-chip pumps and valves,” Sensors & Actuators: B. Chemical, 96(1-2), pp. 38-

45, 2003.

[97] J. Hong, V. Studer, G. Hang et al., “A nanoliter-scale nucleic acid processor with parallel

architecture,” Nature biotechnology, 22(4), pp. 435-439, 2004.

[98] H. Benson, University Physics, Revised ed., pp. 293-296, New York: John Wiley & Sons

Inc., 1996.

100

[99] H. Noguchi, G. Gompper, L. Schmid et al., “Dynamics of Fluid Vesicles in Flow through

Structured Microchannels,” Europhysics Letters, 89(2), pp. 28002, 2010.

[100] Invitrogen. "Cell Tracker Compounds," March 25, 2010;

http://www.invitrogen.com/site/us/en/home/Applications.html.

[101] A. Technolgies, "Adding DC Offsets to a Function Generator's Output," Measurement

Tips, 2009, pp. 1-4.

[102] W. Kang, E. Rabe, S. Kopetz et al., “Novel exposure methods based on reflection and

refraction effects in the field of SU-8 lithography,” Journal of Micromechanics and

Microengineering, 16(4), pp. 821, 2006.

[103] M. Ghantasala, J. Hayes, E. Harvey et al., “Patterning, electroplating and removal of SU-

8 moulds by excimer laser micromachining,” Journal of Micromechanics and

Microengineering, 11(133, 2001.

[104] M. Dickey, R. Chiechi, R. Larsen et al., “Eutectic gallium-indium (EGaIn): A liquid

metal alloy for the formation of stable structures in microchannels at room temperature,”

Advanced Functional Materials, 18(7), pp. 1097, 2008.

[105] R. Pelzer, “Laser-induced fluorescence fibre array for microfluidics,” Fachbereich II

Mathematik-Physik-Chemie, University of Applied Science, Berlin, 2009.