Download as pdf or txt
Download as pdf or txt
You are on page 1of 58

Journal of Micromechanics and Microengineering

ACCEPTED MANUSCRIPT

Single Cell Electroporation-Current Trends, Applications and Future


prospects
To cite this article before publication: Srabani Kar et al 2018 J. Micromech. Microeng. in press https://doi.org/10.1088/1361-6439/aae5ae

Manuscript version: Accepted Manuscript


Accepted Manuscript is “the version of the article accepted for publication including all changes made as a result of the peer review process,
and which may also include the addition to the article by IOP Publishing of a header, an article ID, a cover sheet and/or an ‘Accepted
Manuscript’ watermark, but excluding any other editing, typesetting or other changes made by IOP Publishing and/or its licensors”

This Accepted Manuscript is © 2018 IOP Publishing Ltd.

During the embargo period (the 12 month period from the publication of the Version of Record of this article), the Accepted Manuscript is fully
protected by copyright and cannot be reused or reposted elsewhere.
As the Version of Record of this article is going to be / has been published on a subscription basis, this Accepted Manuscript is available for reuse
under a CC BY-NC-ND 3.0 licence after the 12 month embargo period.

After the embargo period, everyone is permitted to use copy and redistribute this article for non-commercial purposes only, provided that they
adhere to all the terms of the licence https://creativecommons.org/licences/by-nc-nd/3.0

Although reasonable endeavours have been taken to obtain all necessary permissions from third parties to include their copyrighted content
within this article, their full citation and copyright line may not be present in this Accepted Manuscript version. Before using any content from this
article, please refer to the Version of Record on IOPscience once published for full citation and copyright details, as permissions will likely be
required. All third party content is fully copyright protected, unless specifically stated otherwise in the figure caption in the Version of Record.

View the article online for updates and enhancements.

This content was downloaded from IP address 157.89.65.129 on 07/10/2018 at 05:04


Page 1 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4 Single-Cell Electroporation: Current Trends, Applications and Future
5 Prospects
6
7
8

pt
9 Srabani Kar 1 , L. Mohan 1 , Koyel Dey 1 , Pallavi Shinde 1 , Hwan-You Chang 2 , Moeto Nagai
10 3
11 , Tuhin Subhra Santra 1, *
12
13 1
Department of Engineering Design, Indian Institute of Technology-Madras, India 600 036

cri
14
15
2
16 Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan 30013
17
18 3
Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi,
19
20 Japan 441-8580

us
21
22
23 Abstract
24
25
26 The ability to deliver foreign molecules into a single living cell with high transfection
27
28
29
an
efficiency and high cell viability is of great interest in cell biology for the applications in
therapeutic development, diagnostics and drug delivery towards personalize medicine. Many
30
31 chemical and physical methods have been developed for cellular delivery, however most of
32
dM
33 these techniques are bulk approach, which are cell-specific and low throughput delivery. On
34 the other hand, electroporation is an efficient and fast method to deliver exogenous
35
36 biomolecules such as DNA, RNA, oligonucleotides into target living cells with the advantages
37
38 of easy operation, controllable electrical parameters and avoidance of toxicity. The rapid
39
40
development of micro/nanofluidic technologies in last two decades, facilitates to focus an
41 intense electric field on the targeted the cell membrane to perform single cell micro-nano-
42
43 electroporation with high throughput intracellular delivery, high transfection efficiency and
pte

44
45 cell viability. This review article will emphasize the basic concept and working mechanism
46
associated with electroporation, single cell electroporation and biomolecular delivery using
47
48 micro/nanoscale electroporation devices, their fabrication, working principles and cellular
49
50 analysis with their advantage, limitation, potential applications and future prospect will also be
51
ce

52 elaborated.
53
54 Keywords: electroporation, single cell micro-nano-electroporation, micro-nano fluidic
55
56 devices, intracellular delivery, transfection efficiency, cell viability.
Ac

57
58
* Corresponding author;
59
60

1
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 2 of 57

1
2
3 E-mail address: tuhin@iitm.ac.in, santra.tuhin@gmail.com (Dr. Tuhin Subhra Santra)
4
5
6 1. Introduction
7
8

pt
9 Treating critical diseases in recent years demands significant research on intracellular delivery
10
11 in the field of medical science and cell biology[1,2]. Efficient and safe intracellular delivery of
12
several kinds of bio-molecules such as large plasmids for gene expression and oligonucleotides
13

cri
14 (antisense DNA or interfering RNA) for protein regulations play a key role in cell biology
15
16 research and understandings, such as mutagenesis and genetic engineering of microorganisms
17
18 [3,4]. Most of the biological compounds of medical interest such as dyes, drugs, DNA, RNA,
19 proteins, peptides, and amino acids are polar and hence are unable to cross the cell membranes
20

us
21 [5]. Though nucleic acid-based therapeutics can have the promising potential, challenges arise
22
23 to deliver highly negative charged biological molecules. In this regard, gene silencing got
24
25
significant attention for transferring small sized interfering RNAs (siRNAs) as specific
26 targeting therapeutic reagents [6,7]. Further progress was assisted with the unique idea of
27
28
29
an
packaging DNA or RNA inside nanoscale viral vector. Though this technique offers very high

30 transducing efficiency, its application in human body is a question in terms of safety concerns
31
[8,9] and encourage non-viral vectors mediated delivery of bio-molecules though being less
32
dM
33 efficient than the former one [10]. The non-viral methods include chemical treatment [11],
34
35 utilization of a biolistic gun [12], ultrasound [13], microinjection [14,15], optoporation or
36
37 photoporation [16–18], microwave [19–21], hydrogel technique[22] etc. However, most of
38 these techniques are used for bulk electroporation and often limited by low efficiency of
39
40 plasmid delivery into different cell type due to toxicity, plasmid degradation, and immune
41
42 response[23,24]. In contrary, electroporation has been often elucidated to be a more convenient
43
pte

44
way to deliver several kinds of biomolecules from nano to macro-size, and many efforts have
45 been devoted to improve the efficiency [25].
46
47
48 In 1756 electroporation was first demonstrated on human and animal skin by applying electrical
49
50
sparks though the phenomenon was not identified as electroporation [26]. Later throughout 18th
51
ce

and 19th centuries, several sets of experiments were conducted on different kind of bio-systems
52
53 such as dead frog, muscle-nerve, red blood cells etc. to understand electrical effects on
54
55 biological materials [27]. It was 1936, maybe for the first time, G.M. McKinley [28] proposed
56 the impact of the electric field itself on cell membrane, beyond its thermal effects. The
Ac

57
58 phenomena were strongly supported by A.L. Hodgkin [29] in 1951 who proposed the
59
60 breakdown of insulating cell membrane “under the influence of the abnormally high potential

2
Page 3 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 difference” which is now named as “irreversible electroporation”. In the context of
4
5 biomolecule-cell interaction studies, reversible electroporation, where the cell membrane
6
7 reseals again after switching off the field, is requisite instead of irreversible electroporation. In
8
the present review, we have stressed more on reversible single cell electroporation (SCEP)

pt
9
10 rather than the irreversible one.
11
12
13 Traditional or conventional electroporation is bulk electroporation (BEP) where millions of

cri
14
15
cells are exposed to an external homogeneous electric field with amplitude in the order of a
16 few kV/cm [30]. The inter-electrode distance varies from millimeter to centimeter range. A
17
18 few hundreds of volts are applied at the electrodes to achieve proper electric field needed to
19
20 accomplish electroporation [31,32]. It is, therefore, a technique to transfect millions of cells

us
21
together. However, studying intracellular behavior of individual cell requires further control
22
23 over cell distribution, and electric field distribution and hence the concept of (single cell
24
25 electroporation) SCEP arises which offers an in-depth characterization of cells. Furthermore,
26
27
28
29
an
heterogeneity in single cells, even if they are originated from same mother cell, has already
been shown to be important in cancer diagnostic, classification, and treatment [25,33]. Also,
30 cross contamination between individual cell contents can be prevented if cells are isolated.
31
32 Furthermore, analyzing cellular omics at bulk population provides average information which
dM
33
34
cannot reveal the details about molecular dynamics at single cell level [25,34]. Despite the fact
35 that several high-resolution strategies exist to detect, photograph, and analyze [35–40], just a
36
37 few method exist for controlling and manipulating the biochemical nature of the contents of
38
39 single cells and subcellular organelles [41,42]. At present, it is difficult, to transfect an
40
individual cell with a gene without transfecting its adjacent neighbor. These challenges were
41
42 readily accepted by the micro-nano fluidic research community around twenty years back, and
43
pte

44 consequently publishing research articles with significant improvement in this field [43,44].
45
46 There are few steps of SCEP: a) trap a single cell from the population, b) apply a highly focused
47 intense electric field amplitude and pulse only to the targeted cell without affecting others. To
48
49 accomplish the first step, researchers had invoked small population of cells instead of dense
50
51 distribution. For example, in 1997 Teruel et al. [45] used only 1 l of the sample to load
ce

52
53 adherent cells into a 10 mm2 surface area. Further research interests grew on the miniaturization
54 of electroporation device giving many additional advantages such as negligible cell damage,
55
56 dosage control capabilities with single-cell resolution, avoiding the use of expensive high-
Ac

57
58 voltage pulse generators and complicated microchamber mounts etc. Several approaches have
59
60
been made to reduce the device size to micron scale and nanoscale [25,44,46] including

3
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 4 of 57

1
2
3 modification of chip structures with varieties of micro/nano-electrode arrangements,
4
5 introducing varieties of electrode materials [46], improving transfection by applying ac
6
7 voltages with different waveforms and frequencies [47], improving the image quality in single
8
cell limit etc. Apart from micro/nanochip-based techniques, solid micro-electrodes, micro-

pt
9
10 capillaries, micro-pipette, nanofountain probe, nanochannel and nanoelectrode based
11
12 techniques also become popular for SCEP [43,48–53]. All those techniques are very special in
13

cri
14 the sense of transfecting a target cell without affecting the adjacent one. Moreover, sub-cellular
15
organelles can be targeted providing a greater control and opportunity to study beyond single
16
17 cell limit [54]. Thus the emerging field of single cell electroporation has great impact on a vast
18
19 area of research from cell biology to biophysics, single cell analysis to therapeutic applications
20

us
21 towards personalize medicine.
22
23 Till now there are a few reviews in single cell electroporation: one by Olofsson et al.[55] in
24
25 2003 on basis of probe types and by Wang et al. [56] in 2010 and Santra et al.[31] in 2013
26
27
28
29
an
presenting the guidance with all important research articles in this topic. Beside those articles
which mainly focuses on SCEP techniques, there are reviews on micro/nano- fluidic devices
30 for electroporation irrespective of bulk or a single cell [57,58]. However, all those reviews
31
32 overlook the progress of theoretical studies which are essentials to frame up the entire progress
dM
33
34
of this field. In this review, we have discussed so far progress of SCEP including proposed
35 devices, their challenges and benefits, applications and future prospectives. First, we started
36
37 with the theoretical understanding of cellular behavior in an external electric field. Then the
38
39 next section discussed the parameters and their impact on cellular uptake. The third section
40
presented the so far studies, recent trends, the comparative analysis of different methods and
41
42 challenges of SCEP. The fourth or the final section presented the drawback and future
43
pte

44 perspective.
45
46
47 2. Theory of electroporation
48
49
50
There are several reviews on the theory of electroporation [59–62] combining so far theoretical
51
ce

understandings in the literature. A number of theoretical models have been presented to explain
52
53 electroporation [63]. In this review, we briefly outline the mostly accepted aqueous pore
54
55 formation model explaining the interaction of externally applied electric fields with cell
56 membrane. In general, the structure of cell membrane is a phospholipid bilayer membrane and
Ac

57
58 is typical ~5 nm in thickness [64]. Each layer of phospholipid contains a polar head and non-
59
60 polar hydrophobic hydrocarbon tails arranged in such a way that the polar head of the two

4
Page 5 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 layers face opposite to each other and hydrophobic tails remain at the middle part of membrane
4
5 structure, making them amphiphilic [64]. In the context of electroporation (EP), this structure
6
7 is of particular importance because of the potential and dipolar electric field distribution across
8
the membrane. The conductivities of the cytoplasm (~0.3 S/m) and extracullular medium (~1.2

pt
9
10 S/m) are much higher than cell membrane (~3×10-7 S/m) [63]. When an external voltage pulse
11
12 is applied, the cell membrane acts as dielectric capacitor. The field is highly intense across the
13
membrane usually in the order of 103-108 V/m depending on the experimental condition

cri
14
15
[48,65]. As a result, a strong electrical force acts on membrane transferring a Maxwell stress
16
17 leading to pore formation and membrane integrity [66–68]. This electric field inducs a position-
18
19 dependent modulation of the membrane potential difference, named as transmembrane
20

us
21 potential (𝑉𝑚 ) which plays the key role in electroporation. The correlation between the
22 transmembrane potential and pore formation was thoroughly investigated by experimental
23
24 measurements and theoretical predictions which showed that pore formation occurs only to the
25
26 membrane area where 𝑉𝑚 is above a critical threshold (~0.2 V-1.0 V) [65,69]. A heuristic
27
28
29
an
overview of pore formation and evolution was presented by Debruin et al. [70] in 1998. The
paper assumed that two kinds of pores exist - hydrophobic or non-conducting pores and
30
31 hydrophilic or conducting pores [70]. Initially the pores are hydrophobic and most of them are
32
dM
33 quickly destroyed by lipid fluctuations. The creation and stability of pores are determined by
34
change of membrane free energy due to pore creation which includes pore edge energy,
35
36 membrane surface energy, steric repulsion of lipid heads lining the pore, and the energy
37
38 induced by transmembrane potential. When hydrophobic pores of radius r > r* (~0.5 nm) are
39
40 created, they convert spontaneously to long-lived hydrophilic pores, which make the membrane
41 permeable. The pore radius r* corresponsds to the size at which it converts from hydrophobic
42
43
pte

to hydrophilic [65]. The value of r* is determined by the meeting-point of membrane free


44
45 energy curves for hydrophobic and hydrophilic pores. The hydrophilic pores within a small
46
47
range of radii just above r* immediately expand to the minimum-energy radius rm to minimize
48 free energy. The abrupt increase of membrane conductance occurs when hydrophilic pores are
49
50 created due to onset of charge trasnport through the pores. The explicit expression of temporal
51
ce

52 evolution of pore denity N is given by ordinary differential equation [64,70]


53
54 𝑑𝑁 2 𝑁 2 (1)
55 = 𝑒 𝛽𝑉𝑚 (1 − 𝑒 −𝑞𝛽𝑉𝑚 ),
56 𝑑𝑡 𝑁0
Ac

57
58
59
60

5
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 6 of 57

1
2
3 where 𝑁0 is the pore density per unit area when 𝑉𝑚 = 0 V, , 𝛽, and 𝑞 are constants. After the
4
5 voltage pulse being applied, the pore density starts to increase, reaches equilibrium pore density
6 2
7 𝑁𝑒𝑞 = 𝑁0 𝑒 𝑞𝛽𝑉𝑚 . After removal of voltage pulse when 𝑉𝑚 drops to zero , the pore density starts
8

pt
9 to decrease. The pores start to shrink rapidly to the minimum-energy radius of 0.8 nm, where
10
11 they persist until resealing takes place [65]. The experimentaly observed long lived permeable
12 state of cells can be explained by the shrinkage of pores to the minimum-energy radius [65].
13

cri
14 While pore shrinkage occurs at few tenths of microseconds, resealing takes from a few seconds
15
16 to minutes depending on experimental parameters [65,71,72]. The temporal evolution of pore
17
18
radius is determined by the rate of change of membrane energy with pore radius i.e., the
19 advection velocity. The schematic of pore creation and evolution is shown in Figure 1. Thus
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

Figure 1. Hypothetical structural rearrangements of a lipid bilayer membrane dur ing


52
53 electroporation.
54
55 the stages of electroporation are composed of the follwing steps: The ‘‘induction step’’
56
Ac

57 corresponds to the charging of cell membrane and nucleation of pores at cell membrane (~a
58
59 few hundreds of nanoseconds), the ‘‘expansion step’’ corresponds to thse pore evolution, the
60

6
Page 7 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 ‘‘stabilization step’’ corresponds to the postpulse shrinkage of pores (~at few tenths of
4
5 microseconds), and the ‘‘resealing step’’ corresponds to the resealing of small pores (a few
6
7 seconds to minute) [65].
8

pt
9 Mathematical model of transmembrane potential - When a spherical cell with non-conducting
10
11 membrane is placed in an externally applied uniform electric field 𝐸𝑎 , the field distribution can
12
13 be obtained by Laplace equations, ∇2 𝜑 = 0, which can be solved analytically giving the so

cri
14
15 called generalized Schwan equation for transmembrane potential
16
17 𝑉𝑚 = 1.5𝑟𝑐𝑒𝑙𝑙 𝐸𝑎 cos 𝜃 (1 − exp(−𝑡/𝜏)), (2)
18
19
20 where 𝑟𝑐𝑒𝑙𝑙 is cell radius and  is angle between the normal to the membrane and the direction

us
21
22
of the field. τ is the charging time of the cell to reach maxima of 𝑉𝑚 . τ depends on the dielectric
23 properties of the membrane, the conductivities of cytoplasm (𝜎𝑖𝑛 ) and external buffer (𝜎𝑜𝑢𝑡 ).
24
25 Considering the membrane to be pure dielectric, the simplified expression of τ is given by
26
27
28
29
an
𝜏 = 𝑟𝑐𝑒𝑙𝑙 𝐶𝑚𝑒𝑚𝑏𝑟𝑛𝑒 (𝜎𝑖𝑛 + 2𝜎𝑜𝑢𝑡 )/(2𝜎𝑖𝑛 𝜎𝑖𝑛 ), (3)

30 where 𝐶𝑚𝑒𝑚𝑏𝑟𝑛𝑒 is membrane capacitance. The value of τ is in the order of few millisecond,
31
32 checked experimentally and calculated theoretically [73]. In order to obtain permeable cell
dM
33
34 membrane, the value of Vm has to be larger than a critical threshold voltage (~0.2 V-1.0 V).
35
36 Here in the context, of SCEP, it is imporatnt to mention that most of the cases the targated
37 single cell does not experience homogeneous electric field. Instead, the recent micro-fluidic
38
39 devices are designed in such a way that the field distribution is preferably intense at the cell
40
41 trapping sites [74–77]. Thus aformentioned Schwan equation is no longer valid and requires
42
43
numerical dervation of field distributions and transmembrane potential.
pte

44
45 During electroporation, an electrical current flows through the cell medium and results in Joule
46
47 heating in the system due to the resistive nature of the medium. As a consequence temperature
48
49 of the system can increase. In a homogeneous planner system of electrical conductivity , the
50
Joule heating rate per unit volume is given by 𝑄 = 𝜎|−∆𝜑|2 due to an applied electrical
51
ce

52 𝜎
potential 𝜑 and the corresponding temperature rise is ∆𝑇 = 𝜌𝐶 |∆𝜑|2 ∆𝑡, when ∆𝑡 is electric
53 𝑝
54
55 pulse duration, 𝜌 is mass density, and 𝐶𝑝 is specific heat [78]. These relations assumes no
56
fringe effect on electrodes and no heat dissipation in-between the pluses. However, the Joule
Ac

57
58 heating can be controlled by the applied voltage and the electrical properties of the system
59
60 including its proper design. The side effect of Joule heating include local burn in the cells and

7
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 8 of 57

1
2
3 hence should be minimal while conducting the electroporation. These side-effect can be
4
5 reduced by applying low field, short pulse and using a low ionic content pulsing buffer.
6
7 Miniaturization of the electroporation device can meet all those requirements and single cell
8
electroporation is the straightforward consequence of device miniaturixzation. The impact of

pt
9
10 temperature on electroporation is described in detail in the next section.
11
12
13 3. Electroporation parameters

cri
14
15
16 The extent of experimental success for electroporation is mathematized by two technical words
17
18 – transfection efficiency and cell viability, obtained by analyzing experimental data. There are
19 a lot of factors playing important roles to achieve electroporation with high transfection
20

us
21 efficiency and high cell viability. A significant improvement in the experimental outcomes is
22
23 only possible by proper optimization of different electroporation protocols. Briefly, we have
24
25
discussed the important experimental parameters and their interlinks.
26
27
28
29
3.1. Electric field distribution
an
30 Electric field distributions play a crucial role to obtain high cell viability because only the part
31 of membrane experience electroporation where the transmembrane voltage is above the
32
dM
33 threshold value. As shown by Kotnik et al. [69,79] intracellular uptake only occurred at the
34
35 poles of the cell. For transmembrane potential high above the threshold value causes
36
37
irreversible pore formation and eventual cell death [80–82]. Thus the concept of localized
38 electroporation came in which cell is partially exposed to external fields to induce local
39
40 membrane deformation. Our previous work has shown to achieve ~90-95% cell viability by
41
42 localized single cell electroporation (LSCEP) [71,83,84].
43
pte

44
45 3.2. Electric pulse
46
47 The electroporation experiments, started with DC voltages, showed distinguished response for
48
49 AC voltages. Several experiments were carried out to reveal the impacts of pulse duration,
50 voltage amplitude and repetition rate on transfection efficiency and cell viability [72]. When
51
ce

52 single pulse was considered millisecond pulse was seen to create larger pore size with higher
53
54 resealing time compare to microsecond pulse [85]. The pore size extends with pulse duration
55
while the pore density increases with field intensity [65].
56
Ac

57
58
59
60

8
Page 9 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 3.3. Electrode material
4
5
6 A metal electrode in electroporation chamber accomplishes transduction of the ionic current
7 into the electrical current [86]. The metal particles with a size of a few hundreds of nanometre
8

pt
9 are released from the electrodes after electric pulses being applied and create local
10
11 enhancements of the electric field leading to the occurrence of electrochemical reactions at the
12
13
electrode/electrolyte interface [87,88]. Though various kinds of metal like aluminum [87], gold

cri
14 [30], silver [89], platinum [53,90] have been used as electrode materials, aluminum is mostly
15
16 used materials as electrodes because of high conductivity, ease of manufacturing and low-cost.
17
18 However, the major drawback of using aluminum is that generated aluminum ions and
19 aluminum oxides are dissolved at both the anode and cathode after electroporation
20

us
21 contaminating the electrolyte causing toxic effects and a conglomeration of cells and aluminum
22
23 complexes and therefore can reduce on cell viability after electroporation being conducted [91–
24
25
93]. One more issue is the direct imaging of cells during electroporation since any metal
26
27
28
29
an
electrode like gold or aluminum will impede the cell-view from microscope objective. It is,
therefore, important to use transparent electrodes instead of metal or design the chip in such a
30 way that the electrodes would not disturb the view. One such approach was using optically
31
transparent indium tin oxide (ITO) electrode amenable to optical inspection of cells during
32
dM
33 electroporation and subsequent effects, facilitating precise measurement of the following
34
35 processes after electric field being applied to the cells [48,77]. However, there are few
36
37 limitations of ITO such as low conductivity, easy degradation in the ionic electrolyte at high
38 voltage, high risk of damage due to Joule heating in the system etc. Researchers also use carbon
39
40 fiber as electrode materials especially in micropipette based electroporation which also shows
41
42 good results [87].
43
pte

44 One more critical concern is the performance of electrode for AC voltage instead of DC. The
45
46 low impedance, highly stable metal electrodes are required, particularly in micropipette
47
48 method, to operate them at high-frequency voltages for recording rapid signals [86]. Though
49
50
Pt and Ag/AgCl electrodes are attractive for high conductivity, Pt electrodes are expensive and
51
ce

Ag/AgCl electrodes must maintain a layer of AgCl, responsible for the reversible redox
52
53 reaction at the electrode surface and stable half-cell potential in electrolytes containing Cl as
54
55 the main anion [86]. It is also important to note that excessive coating of AgCl tends to increase
56
the resistance of the electrode demanding precise optimization on its thickness to minimize the
Ac

57
58 electrode-electrolyte interface impedance [94]. One intermediate approach was coating an Ag
59
60 wire followed by removing a portion of the initial deposit by reversing the voltage bias,

9
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 10 of 57

1
2
3 resulting in electrodes with a lower and more stable electrode impedance [95]. It is also
4
5 important to keep the electrode-electrolyte junction potential to be stable during an experiment
6
7 to avoid offset artifacts.
8

pt
9
10 3.4. Electroporation buffer
11
12 Selection of appropriate buffer solution is a serious and inevitable job for high yield
13

cri
14 electroporation. The choice of the buffer depends on cell type [96,97]. An appropriate buffer
15 with low ionic content can enhance transfection efficiency [96]. Usually, buffer with high ionic
16
17 strength (high conductance) such as phosphate buffered saline (PBS), HEPES buffer, serum-
18
19 free growth media are used in electroporation of mammalian cells [97]. Also, the buffer pH is
20

us
21
as important as buffer compositions to mimic the cytoplasm of the cell [96,97].
22
23
3.4. Temperature
24
25
26 Ambient temperature has a great effect on electroporation and requires to be optimized before
27
28
29
an
and after electroporation in order to control membrane permeabilization, transfection, pore
resealing, and cell viability. Because temperature acts on lipid fluidity; consequently pore
30
31 resealing time is found to be shortened at a higher temperature. Fast resealing decreases
32
dM
33 transfection efficiency and increases cell viability for example cells were found to be permeable
34 for up to 4 h at 4 °C, while at 37 °C the membrane resealing occurs in less than 5 min [98]. In
35
36 addition, the critical voltage for electropermeabilization was found to be strongly dependent
37
38 on temperature, for example, it increases by a factor of 2 with decreasing temperature from
39
40 37C to 5C in Madin-Darby canine kidney epithelial cells [99]. The experimental studies also
41 showed that the rise of temperature can reduce the critical potential for electroporation. The
42
43
pte

critical voltage at 5C was seen as much as two times the voltage needed at 37C [100].
44
45
46 Nowadays the use of polymeric electroporation chip is very popular because of their easy to
47
fabricate, low cost, easy visualization characteristics. Mostly electro-kinetically driven
48
49 microfluidic devices are based on polydimethylsiloxane (PDMS), and hybrid PDMS/Glass.
50
51 However, their low conductivities give rise to localized Joule heating at the microchannel
ce

52
53 leading to a significant temperature rise. For example, a nearly fivefold increase in the
54 maximum buffer temperature has been seen in the PDMS/PDMS chips over the PDMS/Glass
55
56 systems at high potential field strengths [101].
Ac

57
58
59
60

10
Page 11 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 4. Why SCEP?
4
5
6 Compare to traditional bulk electroporation (BEP), where thousands of volts are applied across
7
8 millions of cells with little control over the permeabilization of individual cells, single-cell

pt
9
10
electroporation (SCEP) technique offers acute transfection of individual cells having flexibility
11 on targated cell numbers (from single cell to high throughput) with low-voltage-pulse (a few
12
13 volts) induced highly focused electric field. SCEP techniques also offer manipulation on the

cri
14
15 cell positioning, cell concentration and reduce reagent consumption. BEP undergoes excess
16
heat generation due to Joule heating because of resistance in the systems, local pH variation,
17
18 electric field distortion, metal ion dissolution from metallic electrodes, and irreversible
19
20 electroporation caused by excessive voltage, resulting in low delivery efficiency and low cell

us
21
22 viability. Real-time monitoring of intracellular delivery in SCEP helps to establish optimal
23 electroporation protocols helps to achieve very high transfection efficiency with high cell
24
25 viability. Moreover, delivering well-defined amounts of materials into cells is important for
26
27
28
29
an
various biological studies and therapeutic applications. SCEP allows qualitative and
quantitative dosage control to conduct cell-autonomous studies of protein function, not possible
30 in BEP [51]. Moreover, the contents of individual cells can be extracted and analyzed, critical
31
32 for investigating the distribution of various critical parameters between cells, instead of average
dM
33
34 data analysis from a population of cells in BEP [102,103]. In this context, it is important to
35
mention that even though the cells which are identical in geometry, contents and overall
36
37 behavior, genetically exhibit marked variations in cellular response and gene expression even
38
39 when the population's characteristics are predictable, implying that BEP is not sufficient to
40
41 describe the precise cellular function [104]. Furthermore, the complexity of neurons and
42 neuronal circuits in brain tissue requires the genetic manipulation, labeling, and tracking of
43
pte

44 single cells [105]. In this context, SCEP has been shown to be useful for in vivo fluorescent
45
46 imaging of neuronal morphology, and connectivity[105]. In order to manipulate and analyze
47
48
the cellular machinery, access to its content is often required [106]. The capability to study the
49 properties of intracellular organelles is a challenging task in cell biology and its therapeutics
50
51
ce

studies. In this context SCEP can be a prospective technique yet far from direct application.
52
53 Both batch-type and flow-through designs have been made to face several existing challenges
54 in current electroporation systems.
55
56
Ac

57
58
59
60

11
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 12 of 57

1
2
3 5. Single cell electroporation: Intracellular delivery and analysis
4
5
6 In conventional electroporation technique, an electric field is applied on aggregations of
7
8 millions of cells located between the electrodes, called bulk electroporation. There is no control

pt
9
10
over individual cells in the medium and thus it blocks precise bio-physical studied in single
11 cell limit in addition with adverse side effects like uncontrolled Joule heating, considerable pH
12
13 change of solutions, lack of precise targeting etc.

cri
14
15
16
With the recent advancement of micro/nano-technologies, micro-needle injection offers precise
17 control over single cell drug/gene delivery but demands high skilled practitioners, special
18
19 equipment, controlled environment. In contrary, single-cell electroporation techniques become
20

us
21 more popular in research community for its simple and convenient working principle to study
22
any kind of cells for understanding bio-physics properties allowing unprecedented spatial and
23
24 temporal control on cargo delivery. It was first proposed by Huang et al. in 1999 who designed
25
26 a microchip having facility to capture and transfect individual cells [107,108]. Here we will
27
28
29
an
briefly discuss the proposed devices and their performances for SCEP. So far demonstrated
methodologies of single cell electroporation platforms can be divided into seven categories:
30
31
32 5.1. Micropipette based single cell electroporation and configurations
dM
33
34
35
While a flexible highly localized single cell electroporation technique is considered with
36 multiplex solution delivery, microfluidic pipette is one of the best approaches. It is of particular
37
38 importance because of its special capabilities to obtain high-resolution spatial control very
39
40 close to targeted single cell or subcellular organelles such as the somatic, dendritic, or axonal
41 region of an adherent neuron, because the exposer can be handled freely in three dimensions
42
43
pte

by using a micromechanical positioner (Figure 2) [52,109]. Though it was initiated with the
44
45 concept of generating blebs across the membrane of targeted single cell or bunch of cells to
46
47
allow bio-molecules to get in, later multipurpose micropipette was combined with electrodes
48 to use them in electroporation [105,106].
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

12
Page 13 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37 Figure 2. (a-c) Schematic view of the circulating liquid tip. (b-f) Photographic images of the
38 microfluidic pipette. (g) Illustration of a microfluidic pipette in a typical experimental setup.
39
40 (h) Working principle of the microfluidic pipette. Pressure difference was created between the
41
42 channels to completely recirculate the solution into the pipette. The volume of the recirculation
43
pte

44
zone (green sphere in panel h) was comparable to the size of the single cell of diameter 10 µm
45 (red sphere in panel h). “Reprinted with the permission from [109]. Copyright (2010) American
46
47 Chemical Society”.
48
49
50
To apply the electric field two strategies of electrode arrangements were proposed by Ainla et
51
ce

al. [106] : one, the electrodes are arranged externally (with respect to the device) (Figure 3A),
52
53 and two, electrodes are fabricated into the pipette enabling solution delivery and targeted field
54
55 to be applied together on one device facilitating the fluidic and electrode probes to be aligned
56
unitedly (Figure 3B) unlike the one by one positioning for the former case. Mostly carbon-fiber
Ac

57
58 microelectrodes are used in this platform.
59
60

13
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 14 of 57

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32 Figure 3. (A,B) Schematic view of the pipette tip with two configurations of electrodes. The
dM
33 electric field is represented by red dotted lines. (C) A schematic working principle of
34
35 representation of a two-channel superfusion pipette, allowing multiplexing between two
36
37 chemical environments:𝐶𝑎2+ for chemical stimulation and Trypan blue for a subsequent
38
39 viability test. “Reproduced from [106] with permission of The Royal Scociety of Chemistry.
40
41 Steinmeyer et al. [105] demonstrated front loaded micropipette to deliver multiple genetic
42
43 vectors and reagents (indicated by different colors in Figure 4 ) into targeted cells within the
pte

44
45 same brain tissue. They achieved high-throughput, efficient, reliable, and combinatorial
46 delivery with single-cell accuracy. The schematic of the step by step process is shown in Figure
47
48 4. They performed multi-colored labeling of adjacent cells with different fluorescent reporter
49
50 plasmids and obtained very high ransfection rate. They were also able to transfect neurons
51
ce

within the same brain tissue with different plasmids encoding different protein isoforms for
52
53 analysis of their effects on linear dendritic spine density.
54
55
56
Ac

57
58
59
60

14
Page 15 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19 Figure 4. Schematic of sequential operations and complete setup. (a) The micropipette was
20

us
21 front-loaded from a multiwell plate containing reagents, (b) before being rapidly transferred
22 and positioned at a fixed location in the field of view of the microscope objective. (c,d) The
23
24 three-axis stage was used to hold the slice bath/manipulation chamber for moving the sample
25
26 and bringing cells into contact with the stationary micropipette, allowing cells to be
27
28
29
an
transfected. (e) The micropipette tip was automatically transferred to a cleaning solution bath
and a rinsing reservoir for washing and finally loading sample from a different well of the
30
31 multiwell plate, allowing the cycle to continue. “Reproduced from [105]. CC BY 4.0”
32
dM
33
34
Ohmachi et al. [110] also utilized this method to deliver a variety of biomolecules such as
35 plasmids, Alexa Fluor 488, and exciton-controlled hybridization-sensitive fluorescent
36
37 oligonucleotide (ECHO) RNA probes into individual living E. gracilis cells showing very high
38
39 transfection efficiency and cell viability. This technique was also used to deliver siRNA into
40
GFP-expressing Golgi and Purkinje cells in cerebellar cell cultures [111] and into adult sensory
41
42 neurons [112] in primary cultures. Also, oligonucleotides and plasmid DNA were reported to
43
pte

44 be delivered in neurons of organotypic rat hippocampus and mouse cortex slice cultures with
45
46 transfection efficiency being as high as ~80% [113]. Small oligonucleotides were found to
47 enter the cell immediately during pulse application while large plasmids were detected to
48
49 remain localized at the cell membrane for more than 10 min before they enter the cell. This
50
51 method of SCEP was found to serve no effect on the electrophysiology of transgene-expressing
ce

52
53
cells. Expression of several GFP-tagged neuronal proteins demonstrates that the method is
54 beneficial to analyze subcellular targeting and/or signaling of transgenic proteins in
55
56 physiologically intact neurons [113]. In vivo electroporation of Xenopus laevis tadpole brains
Ac

57
58 were also transfected with DNA, dextrans, morpholinos and their combinations by using
59
modified patch electrodes and common electrophysiological equipment [114] and micropipette
60

15
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 16 of 57

1
2
3 [115]. In vivo -targeted electroporation was performed on single tectal neurons within the
4
5 albino Xenopus laevis tadpole using a pipette electrode filled with a solution of the delivery
6
7 molecules and with a tip diameter much smaller than the width of the target cell [116]. A similar
8
method was utilized for longitudinal imaging of synaptic structure and function in the adult

pt
9
10 mouse neocortex in vivo [117].
11
12
13 To obtain acute control over electroporation area, Iwata et al. [118] et al. proposed nanopipette

cri
14
15
based non-contact approach for SCEP where the probe and cell surface distance can be
16 controlled by using a scanning ion conductance microscope (SICM) allowing low-invasive
17
18 electroporation of a single cell. The schematic diagram is shown in
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35 Figure 5. An electrode, acting as an inner electrode, is inserted within the nanopipette and outer
36
37 electrode is fabricated at its outer edge. When an external voltage is applied between those
38
39 electrodes, a local-focused electric field is created at the edge/tip of the nanopipette transfecting
40
the targeted single cell. The nanopipette is positioned in the vicinity of the target cell by
41
42 detecting the ion current using the SICM technique and the SICM unit is put on a sample stage
43
pte

44 of an inverted optical microscope for direct observation of cell manipulation and experiments
45
46 were conducted on Hela cell to deliver potassium iodide (PI) dye [118]. Though there are a lot
47 of works using micropipettes, the major disadvantage is the fact that the tip has to be repeatedly
48
49 repositioned and aligned in a sequential manner to target different regions requiring
50
51 considerable skill and expertise.
ce

52
53
54
55
56
Ac

57
58
59
60

16
Page 17 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19 Figure 5. Nanopipette for single-cell electroporation. A local electric field is formed at the
20

us
21 edge of the nanopipette by applying an electrical voltage between the outer electrode and
22 inside electrode of the nanopipette. “Reproduced from [118]. Copyright (2014) The Japan
23
24 Society of Applied Physics”.
25
26
27
28
29
5.2. Patch-clamp technique
an
30 The patch-clamp technique facilitates to record ionic currents flowing either through single ion
31
32 channel or through whole cell membrane [119]. There are two operational configurations - cell-
dM
33
attached mode and whole-cell mode [119]. The whole-cell mode is mostly utilized for single
34
35 cell electroporation [82,120,121]. Here we discuss the approach presented by Ryttsén et al.
36
37 [120] who used patch-clamp in combination with microelectrodes and fluorescence
38
39 microscopy to facilitate the recording, labeling and genetic manipulation of single cells. The
40 schematic of the technique is shown in Figure 6. A few micron-sized carbon fiber
41
42 microelectrodes, controlled by high-graduation micromanipulators, were enclosed in a plastic
43
pte

44 tip filled with electrolyte which is in contact with a metal (silver) wire to apply the voltage.
45
46
The targeted cell/cells were patch clamped and the electrodes are positioned on opposite sides
47 of the cell (Figure 6). The electrode tips were positioned 2–5 m from the outer cell surface at
48
49 an angle of 0–20°, and 160–180° with respect to the object plane, and 90° with respect to the
50
51 patch pipette. This method enables probing of membrane properties, important for studying
ce

52
53 membrane dynamics, throughout the electroporation in the targeted single cell by measuring
54 the current response during and after the exposer of pulsed external voltage. This technique
55
56 was also used for transfecting neurons to deliver plasmid DNA, leading to stable transgene
Ac

57
58 expression [120,121]. Rae et al. [122] also used the modified patch-clamp technique to
59
electroporate single cell and deliver mostly several kinds of DNAs.
60

17
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 18 of 57

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52 Figure 6. Schematic drawing of the experimental setup. (a) A patch-clamped cell in a buffer
53
54
solution containing the solutes to be introduced into the cell and the electrodes were arranged
55 in a linear fashion across the cell opposing each other, at a 90° angle with respect to the patch
56
Ac

57 pipette. (b) While a voltage pulse was applied, pores were formed allowing intracellular
58
59
60

18
Page 19 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 delivery. (c) After switching off the pulse, the solutes are present in the electroporated cell.
4
5 “Reprinted with permission from [120]”.
6
7
8

pt
9
10 Kitamura et al. [121] (2008) performed patch-clamp recordings from single neurons in vivo
11
12 which was visualized by two-photon microscopy. A patch electrode was used to capture
13
negative image of the targeted cell by perfusing the surrounding extracellular space with a

cri
14
15 fluorescent dye. The same electrode was then placed on the cell to allow the formation of a
16
17 gigaseal. Thus the method was shown to be useful for studying transgene expression after
18
19 single-cell electroporation of plasmid DNA into identified cell types, without the need to pre-
20

us
21
label neuronal populations in vivo [121].
22
23
5.3. Micro/Nano- electrode based devices
24
25
26 Among many transfection techniques to deliver biomolecules into cells, the micro-electrode
27
28
29
an
based devices are very popular for the minimal cellular disturbance and their enough size for
single cell analysis. Micro-pattern electrodes with reduced inter-electrode distance permit a
30
31 considerable reduction in operating voltage to reach the same electric field strength (V cm−1)
32
dM
33 and have become one of the main concepts of micro-electroporation systems in the last decade.
34 This kind of device was first proposed by Lundqvist et al. [46] in 1998. Two carbon fiber solid
35
36 microelectrodes with diameter ~5 µm were placed on two side of a selective adherent
37
38 progenitor cell by using high graduation micromanipulators. The schematic setup is shown in
39
Figure 7. A gap of 2-5 micron was maintained between the electrode tip and cell surface to
40
41 avoid electrochemical transformation of proteins and other membrane-associated structure.
42
43 Since the size of the electrodes were much less than average cell dimension and could be placed
pte

44
45 precisely (in steps of 0.2 micron) by a micromanipulator, the high spatial resolution was
46 achieved. Thus this method facilitated to select a particular cell from a randomly distributed
47
48 cell patterns cultured on a planner substrate to apply high voltage without affecting adjacent
49
50 cells. Two factors are important for successful electroporation: one, the cell should be isolated
51
ce

52
from adjacent cell and two, the electric field should be well focused near the targeted cell
53 membrane which requires cell trapping on specific positions of the device.
54
55
56
Ac

57
58
59
60

19
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 20 of 57

1
2
3
4
5 i. iii.
6
7
8

pt
9
10
11
12
13

cri
14
15
16 ii.
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31 Figure 7. i. Schematic setup for single-cell electroporation. ii. Selective electroporation of
32 single cell. (A and C) Bright-field images of all cells. The arrow shows the targeted one. (B
dM
33
34 and D) Fluorescence showing electroporation is localized preferentially to targeted cells. iii.
35
36 Time sequencing of electroporation with three 1-ms pulses of ~0.5 V. “Reprinted from [46],
37
38
Copyright (1998), with permission from National Academy of Sciences, U.S.A”.
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

20
Page 21 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 In 2015, Bürgel et al. [123] presented a microchip where sets of facing Pt electrodes of
4
5 thickness 200 nm were desigend across a microfluidic channel, as shown in
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
Figure 8. The microsystem is combined with electrical impedance spectroscopy (EIS) offering
30
31 a label-free analysis to probe the type, size, and dielectric properties of cells. In EIS, a pair of
32
dM
33 electrodes is used to apply an AC voltage resulting in a current to flow between them which is
34
35 sensitive to the cell passing between the electrodes. The current change is being measured to
36 determine the dielectric properties of the cell. The electrodes were temporarily switched on
37
38 during the passage of a single cell.
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

21
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 22 of 57

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26 Figure 8. Schematic micro-chip design and working principle. “Reprinted with permission
27
28
29
from [123]”.
an
30
31
32
dM
33 Zhang et al. [124] designed co-planer microelectrodes, fabricated in the side wall of micro-
34
35 cavity geometry on the microchannel of the microfluidic chip, can induce electroporation
36 around the contact area of cells and cavity resulting high-yield electro-fusion in Myoblast cells.
37
38 A theoretical analysis was carried out along with experiments to understand pore distribution
39
40 for different configurations of electrode micro-cavity structures. He et al. [30] fabricated a
41
42
MEMS-based micro electroporation chip containing three-dimensional (3D) gold electrodes to
43
pte

determine the optimal physical parameters for the uptake of biomolecules in HeLa cells.
44
45
46 In our previous works [71,125] we demonstrated a unique approach for microelectrode based
47
48
single cell nano-electroporation. In this configuration, transparent ITO electrodes were used
49 instead of metallic one allowing direct imaging. The device structure is shown in Figure 9. The
50
51
ce

electrodes were covered with a dielectric passivation layer of SiO2 to prevent direct touch of
52
53 electrodes to the cells so that entire cell surface does not experience the intense electric field;
54 instead area of permeabilization can be controlled in addition with the facility of cell-selective
55
56 electroporation and eventually improved viability [126]. Furthermore, it also avoids bubble
Ac

57
58 generation, reduces chemical reaction, and control over Joule heating [72]. To construct sub-
59
60 micron range electroporation region the final ITO electrodes and SiO2 layer were cut by using

22
Page 23 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 focus-ion-beam technology. The final chip allows biomolecules to be delivered through the
4
5 holes in the sub-micro-channel from the bottom of the device facilitating the user to keep the
6
7 molecules and dye solution separate from cell suspension with cell viability ~60-80 %. In the
8
next work, we modified the electrode structure from rectangular edge to triangular edge

pt
9
10 [74,77,127] to achieve ultra-localized the electric field with higher field strength resulting in
11
12 an improved cell viability (~90-95%) for plasmid delivery.
13

cri
14
15
16
17 (a)
18
19
20

us
21
22
23
24
25
26
27
28
29
(b)
an
30
31
32
dM
33
34
(c)
35
36
37
38
39
40
41
42
43
pte

Figure 9. (a) Schematic representation of localized single cell nano-electroporation chip,


44
45 where an intense electric field affects a small region of the cell membrane to permeate drug
46
47
from outside to inside of the cell. “Reprinted with permission from [48]. Copyright (2013)
48 American Institute of Physics”. (b) Upper left Figure: Optical microscope image of patterned
49
50 ITO electrode (20X). Lower left Figure: the SEM image of ITO electrode with micro
51
ce

52 channel. Upper right Figure: Working principle of electroporation and intracellular delivery.
53
“Reprinted with permission from [125]. Copyright (2012) Springer Publisher Ltd”. (c)
54
55 Scanning electron microscopy (SEM) image of nano-electrode tip with 60 nm electrode gap
56
and 40 nm tip diameter. “ [2014] IEEE. Reprinted with permission from [77]”.
Ac

57
58
59
60

23
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 24 of 57

1
2
3 In the previously described micro/nano- devices, the cell membrane is electroporated only
4
5 across the fraction of cell surface which is perpendicular to the electric field distribution.
6
7 Consequently, the only fraction of cell surface get electroporated and limits the total
8
electroporation area of a cell. To address this limitation, Zheng et al. [128] designed pinch flow

pt
9
10 technique based microfluidic device which can control cell rotation allowing to expose the
11
12 perpendicular electric field all over the cell surface. The device consists of a three/two-inlet
13

cri
14 microfluidic channel decorated with a pair of micro-electrodes (inter-electrode distance ~ 410
15
microns). The corresponding schematic diagram is shown in
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37 Figure 10.
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

24
Page 25 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 Figure 10. The schematic of cell rotation based EP technique with two configurations.
4
5 “Reprinted with permission from [128]. Copyright (2016) Springer Publisher Ltd”.
6
7
8

pt
9
10 Electroporation buffer or pulsing buffer solution was infused from top and bottom (only
11
12 bottom) channels and cell carrier buffer was infused from middle (top) channel for three (two)
13

cri
14 inlet device. Different flow rate ratios were examined to control the cell positioning, rotation,
15
and stabilization.
16
17
18 5.4. Flow-through microfluidic microhole array
19
20

us
21
In 2001, Huang et al. [107] for the first time demonstrated microfluidic device for SCEP which
22 was indeed a flow-through a device having a microhole to capture single cell. The chip
23
24 consisted of three fabricated silicon chips as the top layer, middle layer and bottom layer, glued
25
26 together by high dielectric strength adhesive to hold them together in saline in the presence of
27
28
29
an
high electric field applied by using two translucent polysilicon electrodes. The chip had the
facility to capture a cell into micron size hole where the cell would experience the dense electric
30
31 field. However, the chip had little control over cell and drug dosage. Later, the same group
32
dM
33 [129] designed another automated micro-electroporation system where electroporation in
34 individual cells can be controlled precisely by measuring real-time electrical currents that flow
35
36 through the cell. The schematic of chip structure is shown in
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

25
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 26 of 57

1
2
3 Figure 11. The chip had two chambers – the top chamber where cell solution is loaded and
4
5 allowed to flow, and the bottom chamber where drug solution is loaded (
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
Figure 11a). The upper chamber contained a micro-channel containing a micro-hole having
30
31
width same order as cell (
32
dM
33
34
35
36
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54 Figure 11b). The micro-hole acted as the electroporation site where the cell was captured by
55
56 pressure difference. The cell captured at micro-hole was exposed to a controlled electric field
Ac

57
58 to deliver the desired genetic molecules and then released by pressure difference to be replaced
59 by the next cell. Around ~100% gene transfer rate had been reported. To avail the visual
60

26
Page 27 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 observation of the cell manipulation process the central region of the chip is designed to be
4
5 transparent.
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31 Figure 11. (a) Concept of flow-through micro-electroporation chip; (b) Upper figure: the
32
dM
33 working principle of trapping and subsequent electroporation. Lower Figure: Sketch of the
34
35 layout of micro-hole, micro-channel and integrated electrodes. “Reprinted with permission
36
37
from [129]”.
38
39 Essentially at the same year of 2003, Khine et al. [89] developed a PDMS chip to selectively
40
41 immobilize and locally electroporate single cells. This chip did not require any chip attached
42
43
electrodes unlike previously discussed micro/nano- electrode based techniques or glass
pte

44 pipettes, unlike micro-pipette based electroporation techniques. The electrodes were placed at
45
46 a distance from the cell eliminating toxic effect to the cells arising from Joule heating.
47
48 Moreover, this device not only allows parallel single cell electroporation but also facilitate
49
direct imaging of cells due to transparent PDMS and monitor the current change during
50
51
ce

electroporation. Figure 12 shows the chip layout, displaying wide inlet and outlet channels
52
53 along with attached syringes for cell trapping by applying negative pressure. Experiments were
54
55 conducted on Hela cell to deliver Trypan blue showing characteristic ‘jumps’ in current
56 corresponding to drops of cell resistance for an applied voltage <1 volt which indicate
Ac

57
58 successful delivery.
59
60
(c)
27
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 28 of 57

1
2
3
4 (a) (b)
5
6
7
8

pt
9
10
11
12
13

cri
14
15 Figure 12. (a) The image of chip layout. The wide channels are for cell input and output. An
16
17 electrode was connected to one of these main channels. The other channels were used to
18 provide negative pressure for cell trapping. (b) A schematic of the cross-section of the chip
19
20 and working principle. (c) An image showing trapped cell in the channel. The cell extends

us
21
22 into the channel leaving a tail-like structure in the channel. “Reproduced from [89] with
23
24
permission of The Royal Society of Chemistry”.
25
26
27
28
29
an
In 2007, Valero et al. [130] demonstrated silicon-glass microfluidic device capable of trapping
30 and subsequently electroporating at most nine cells together in parallel. The principle of
31
32 operation and the schematic device structure is shown in Figure 13. The device contained two
dM
33
34 parallel microchannels (one have with ~50 m and the other one ~20 m) connected by nine
35
36 micro-holes (width ~4 m) acting as trapping sites. The arrangements of the platinum
37
38
electrodes are shown in Figure 13a which allow selective addressing of each microhole to
39 achieve control over cell selective electroporation. The cells flowed through the upper wider
40
41 channel and other channel was used to create under pressure to trap the cells at the microholes.
42
43 Once the cells have been trapped, the pressure was stopped and top and bottom reservoirs were
pte

44
loaded with DNA construct which had to be delivered into the cells.
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

28
Page 29 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
Figure 13. a. Working principle of cell trapping and electroporation. b. Microfluidic chip
20 layout, zoom-in on trapped single cell. “Reproduced from [130] with permission of The

us
21
22 Royal Society of Chemistry”.
23
24
25
26
27
28
29
an
5.5. Micro/nano-channel based microfluidic electroporation devices

30 A micro/nanofluidic based single-cell analysis platform offers to control precise amounts of


31
delivery of various transfection agents into living cells. Nano-channel based device was
32
dM
33 introduced by Ly James Lee’s group [53,75,76,131] in 2011. It consists of two microchannels
34
35 connected by a nanochannel integrated with optical tweezers to manipulate cell position close
36
37 to the nanochannel in one microchannel, as shown in Figure 14. The transfection agent was
38 loaded in the other microchannel. An intense electric field was delivered over a very small area
39
40 on the cell membrane across the nanochannel, allowing a precise dose of biomolecules to be
41
42 electrophoretically driven through the nanochannel, the cell membrane and into the cell
43
pte

44
cytoplasm, without affecting cell viability. The applied voltage duration and number of pulses
45 were adjusted to control the dose. The method could create a very large pore or several large
46
47 pores in the cell membrane adjacent to the nanochannel thus reduced the affected area and
48
49 provided a spatial resolution of 200 nm. Further experiments were carried out showing the
50 precise delivery ability and intracellular RNA analysis ability of molecular beacons without
51
ce

52 cell lysis or fixation [53]. In addition, a precise dosage of genes or other types of stimulus could
53
54 be controlled into individual cells before molecular beacons-based living cell interrogation
55
56 which is a very important feature for rare cell analysis. The platform also could handle a cell
Ac

57 population from single to ≈102 cells and provided greater flexibility to play with cell number.
58
59 High transfection efficiency was found for large genes which are indeed a challenge for
60

29
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 30 of 57

1
2
3 conventional EP techniques. Also, it offered greater flexibility than PCR and
4
5 immunocytochemistry-based methods. Also, OSKM plasmid of induced pluripotent stem cell
6
7 (iPSC) was injected into mouse embryonic fibroblasts cells showing high delivery dosage in a
8
much more uniform manner compared with similar gene transfection carried out by the

pt
9
10 conventional bulk electroporation (BEP) method [76].
11
12
13

cri
14 (a)
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
(b)
an
30
31
32
dM
33
34
35
Figure 14. (a) Optical image of the micro-nano channel. . “Reproduced from Ref. [53] CC BY
36
37 4.0”. (b) Design of cover lid and entire package system to encapsulate NEP chip. (I) Gold-
38
39 coated DNA nanowire (SEM). (II) Imprinting with 300 nanochannels. (III) NEP device with
40
41 MNM array in the central area. (IV) Device package case. (V) Ready for cell and gene loading.
42 (VI) Multi-location spinning disc. “Reprinted with permission from [76]. Copyright (2014)
43
pte

44 John Wiley & Sons Ltd”.


45
46
47
48 5.6. Field effect transistor-based devices for SCEP
49
50 In 2013, Jokilaakso et al.[132] utilized nanowire and nanoribbon channel field effect transistor
51
ce

52 for single cell electroporation. They used silicon nanowires with diameters in the range 50-100
53
54
nm or nanoribbon with width ~2 m as transistor channel allowing them to obtain highly
55 localized electric field at the nanometre scale above the device surface. The schematic of the
56
Ac

57 device is shown in
58
59
60

30
Page 31 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19 Figure 15. In this device magnetic beads were used to move the cells into the desired position
20
and thus overcomes the need for any trapping center; any transparent substrate required for

us
21
22 optical tweezer; and high voltage with low medium conductivity in the dielectrophoresis-based
23
24 trapping of cells. The device surfaces were passivated using an organosilane monolayer to
25
26 prevent unwanted adhesion with the cell. In this study, pulsed voltages at 10 MHz repetition
27
28
29
an
rate and amplitudes up to 900 mVpp were applied between the shorted source-drain and the
back gate of the transistor. They also confirmed that the cell only at the top of the transistor
30
31 was lysed and the cells immediately adjacent were remained unaffected, thus enabling single-
32
dM
33
cell transfection. 100 % cells were seen to be lysed at 900 mVpp voltage for 1 s.
34
35
36
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Figure 15. (a,b) The schematic device structure of FET. (c,d) The SEM image of the grouping
Ac

57 of five nanowires in (c) and nanoribbon in (d). (e) The transistors array with both nanowires
58
59
60

31
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 32 of 57

1
2
3 and nanoribbons. “Reproduced from [132] with permission of The Royal Society of
4
5 Chemistry”.
6
7
8

pt
9
10
11
5.7.Nanofountain probe SCEP
12
13 The schematic of the nanofountain probe SCEP set up is shown in Figure 16. The device

cri
14
15
contains twelve cantilevered probes with an average aperture size ~750 nm (Figure 16a) which
16 connected to two independent microreservoirs through sealed microchannels. The molecular
17
18 solution to be delivered was stored in the on-chip microreservoirs to load into the
19
20 microchannels by using capillarity or an externally applied pressure. A nanomanipulator or

us
21 AFM was used to control the position of interest of the targeted cell in an extremely small step
22
23 size of 40 nm. The onset of contact of the probe-tip on targeted area of a specific cell was
24
25 detected either by optical observations of the cell morphology or by monitoring the change of
26
27
28
29
an
electrical resistance due to the sealing between probe tip and cell. After the contact being
established, an electric pulse was applied between Ag/AgCl wire and conducting coverslip
30 acting as electrodes as shown in Figure 16. This technique of SCEP using nanofountain probe
31
32 has recently (2018) been utilized by Yang et al.[133] for generation of monoclonal cell lines.
dM
33
In this approach, arrays of cell colonies (with each colony has ~10-20 cells) were patterned by
34
35 microstamping. An indivisual cell of each colony was transfected with a plasmid encoding GFP
36
37 and resistance to the antibiotic Zeocin, by using nanofountain probe method. When the colony
38
39 of GFP-positive cells proliferated from the initial transfected cell, the stable clonal cell lines
40 were selected using Zeocin. Thereafter GFP-expressing colonies were harvested as clonal cell
41
42 lines with stable transfection. Thus this method conferred a significant advantage over slower
43
pte

44 and labor-intensive dilution procedures.


45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

32
Page 33 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
Figure 16. (a) Working principle of nanofountain probe. (b) Schematic of the package for
single cell electroporation. “Reprinted with permission from [51]. Copyright (2013)
30 Americal Chemical Society.
31
32
dM
33
34
35
36 5.8.Parallel single cell electroporation
37
38 The bulk or individual electroporation techniques of cells are limited either by limited
39
40 selectivity for bulk or low throughput for individual electroporation. So far demonstrated
41 techniques have few issues. For example, microelectrode EP technique offers high
42
43
pte

throughput electroporation allowing single cell selectivity through individual addressing of the
44
45 microelectrodes. Indeed, this is not a direct method of cell selection. Microstructures of
46
electrodes were also designed on microfluidic devices to apply focused electric field on the
47
48 particular trapped cell without affecting the surroundings, also resulting in high throughput
49
50 [130,134]. Micropipette based technique, though allow to select a particular cell of
51
ce

52 experimentalist’s choice from a population and allow different drugs to be injected into
53 different cells with accurate dosage control, which none of the other techniques allow for, is a
54
55 very slow approach to porate cells one by one and requires a skilled user and is, generally, low
56
Ac

57 throughput.
58
59
60

33
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 34 of 57

1
2
3 Beside those mechanisms in situ parallel single cell electroporation technique was first
4
5 demonstrated by Valley et al. [135] in 2009 which allows an array of single cells to be
6
7 transfected simultaneously with controlling over selectivity on each individual cells. The
8
device was integrated with optical tweezers to control parallel single cell movement. The

pt
9
10 schematic device layout and the cell-selective electroporation is shown in Figure 17.
11
12
13 (a)

cri
14 (c)
15
16
17
18
19
20

us
21 (b)
22
23
24
25
26
27
28
29
an
30
31
32
dM
33 Figure 17. (a) Schematic overall device layout. (b) Schematic cross-section of the device
34
35 showing experimental setup and mechanism of electroporation. (c) Optical patterns cause
36
37
electric field concentration across illuminated cells resulting in selective electroporation.
38 “Reproduced from [135] with permission of The Royal Society of Chemistry”.
39
40
41
42
43
pte

In 2010 Fei et al. [136] demonstrated micronozzle array structures to improve non-uniform
44
45 electric field distribution on a cell population, which is indeed parallel single cell
46
47 electroporation, though not mentioned by the author, allowing manipulation of cell
48
49 distributions and controlled electroporation of bunch of cells. The device consists of a pair of
50 cross channels (~500 µm width and depth), one act as top and other as the bottom, connected
51
ce

52 by a center hole as shown in Figure 18. The top channel intersects with a 1 cm diameter
53
54 reservoir located at the center of the device where microporous membranes can be fixed.
55
Femtosecond pulsed laser ablation technique was adapted to form a pattern of
56
Ac

57 micropore/micronozzle array with pore radius ~0.4 µm on nanoporous polyethylene


58
59 terephthalate (PET) membraned coated with 2-3 µm thick gelatin, acting as cell binding
60

34
Page 35 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 membrane. The device was fabricated by using a poly(methyl methacrylate) (PMMA). A 45
4
5 µm thick PMMA film enclosing the bottom channel was used at the back side of the device
6
7 allowing top access via reservoirs at the ends. The shape of the pore could be tuned from micro-
8
nozzle to straight microchannel by changing the laser energy. Experiments were performed on

pt
9
10 mouse embryonic stem cells to transfect with plasmids gWiz SEAP as reporter genes, and the
11
12 transfection of on the 100 × 100 array was evaluated 24 h after electroporation. The
13

cri
14 micronozzle structures were found to transfect almost doubled over that with microchannels
15
because the electric field is more concentrated at the small-end of the micronozzle, resulting in
16
17 better-localized electroporation resulting ~75% cell survival.
18
19
20 (a)

us
21
22
23
24
25
26
27
28
29 (b)
an
30
31
32
dM
33
34
35
36
37
38
39 Figure 18. (a) Schematic of disk structure and working principle. (b) Phase contrast (left)
40
41
and fluorescent image (right) of cells trapped in 10 × 10 micronozzle array. “Reprinted with
42 permission from [136]. Copyright (2010) American Chemical Society”.
43
pte

44
45
46
47 At the essentially the same year 2010, Wang et al. [137] independently demonstrated single-
48
49 cell electroporation chip that can locally electroporate array of cells with an operating voltage
50
51 as low as ~1 Volt. The chip consists of one bottom metal electrode on the top of a substrate, a
ce

52
53 sandwiched dielectric polymer layer with thickness ~10 µm patterned with an array of
54 microwells, and a conducting top lead as another electrode. The distance between these two
55
56 electrodes was 60 µm, the diameter of the microwell was ~10 µm, nearly same to average cell
Ac

57
58 diameter. The schematic configuration of the device is shown in
59
60

35
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 36 of 57

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19 Figure 19. When an AC voltage was applied to the electrodes, the microwell arrays create non-
20

us
21 uniform electric fields to generate positive dielectrophoresis forces to capture one single-cell
22 at each microwell. After cell capturing, the AC voltage was shifted to a DC pulse voltage to
23
24 generate an intense electric field across the membranes of the captured cells leading to
25
26 electroporation. The field distribution, shown in Fig. 19 , showed the strong electric field was
27
28
29
an
generated on the cell membrane in contact with the microwell, and decay quickly upwards,
allowing localized electroporation at relatively low voltage. Human lymphoma cells in an 8 ×
30
31 11 array were successfully electroporated with ~100% electroporation yield. This technique
32
dM
33 was able to avoid bubble generation due to water electrolysis and Joule heating caused by the
34
high field strength implementing both DEP trapping and cell electroporation with the same
35
36 device.
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57 Figure 19. Schematic device configuration of electroporation in the non-uniform distribution


58
59 of electric field.
60

36
Page 37 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6 High throughput “3D Nanochannel electroporation chip” (NEP) was introduced by Chang et
7
8 al. [138] in 2016 which can parallely electroporate ~40,000 cells per cm2 of the chip. This

pt
9 system consists of four parts, as shown in Figure 20, one 3D electroporation chip, a support
10
11 platform, PDMS spacer, a pair of electrodes. The top of 3D chip is a PDMS chamber to load
12
13 cells to be electroporated. The molecules to be delivered are filled into the bottom chamber,

cri
14
15
separated from the top chamber by the 3D chip (Figure 20). During cell loading, a vacuum was
16 applied underneath the 3D chip to drive the cells towards the nanopores for alignment and
17
18 enhancing the contact with the pores. High voltage pulses were applied between the top and
19
20 the bottom electrode. Voltages with different amplitudes (15-140 V), pulse widths (5,10, and

us
21
30 ms) and number of pulses were varied to study the impact on delivery efficiency. The device
22
23 showed around >90% of cell viability and ~90% of transfection efficiency when only the cells
24
25 sited on the nanopores are counted. This approach also offers precise control over dosage with
26
27
28
29
an
high throughput. The results of NEP were compared with microchannel electroporation chip
(MEP) with pore diameter of ~5 m. The device set up was same as for NEP, the only
30 differences are the micro-scale pore size and using magnetic micro-beads for cell alignment in
31
32 the MEP system [139]. In their next work, they modified the NEP chip with micro-cap array
dM
33
34 to align the cells on nanochannel more precisely [140]. They achieved a high-throughput
35 massively parallel delivery of genetic cargo (microRNA, plasmids) into mouse primary
36
37 cardiomyocytes with transfection efficiency ~ 90-100% in a controllable fashion.
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

37
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 38 of 57

1
2
3
4 (a)
5
6
7
8

pt
9
10
11
12
13

cri
14 (b)
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
Figure 20. (a) The schematic set up of 3D electroporation chip. (b) The schematic of

30
microchannel – nanochannel connection and SEM micrograph of the chip. “Reproduced
31 from [138] with permission of The Royal Society of Chemistry.
32
dM
33
34 Guo et al. [141] demonstated micro-electrode array chip having facility of cell positioning and
35
36
real time impedance monitoring for cell selective parallel electroporation at single cell level.
37 They patterened array of quadrupole-electrodes unit (named positioning electrodes) for cell
38
39 positioning and pair of planer electrodes (named center electrodes) located at the centers of
40
41 each quadrupole-electrode unit for performing electroporation (Figure 21). Based on negative
42 dielectroforesis by using the positions electrodes the cells were trapped and positioned onto the
43
pte

44 center electrodes. Finally, the cells were electroporated by applying voltage to the center
45
46 electrodes. The effiency of the selective electroporation was reported to be higher than 90%.
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

38
Page 39 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
Figure 21. (a) Schematic diagram of electrical electrical arrangement for the center
16
17 electrodes. (b) nDEP assisted cell positioning on the center electrodes forming arrays. (c)
18
19 Results of selective electroporation in array dyed by Propidium iodide. “Reprinted with
20

us
21 permission from [141]. CC BY 4.0”.
22
23
24
25
26 6. Cell positioning in SCEP
27
28
29
an
In order to conduct single cell electroporation, cell positioning is the foremost required step
30
31
because only target cell has to experience the intense electric field required to break down the
32 bilipid membrane. Many methods of cell positioning have been demonstrated such as creating
dM
33
34 pressure difference at trap centers, optically-induced-dielectrophoresis (ODEP), electrically
35
36 induced dielectrophoresis, and micro-fluidic structures [142]. Dielectrophoresis (DEP) is a
37
movement of polarized particle or cell due to translational force by a non-uniform electric field.
38
39 It is one of the most versatile methods to control cell movement and positioning due to its
40
41 capability to integrate with in-situ cellular measurements. Also, the cells under DEP-based
42
43 manipulation could maintain good viability [142]. Here is the table listing the methods adapted
pte

44 by SCEP researchers for cell positioning.


45
46
47 Year/ Group Trapping configuration
48
49
50
1. Huang et al. Applying a negative pressure to pull towards and captured
51
ce

[107,129,143] by a microhole
52
53 (2000,2001,2003).
54
55
56
Ac

57
58
59
60

39
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 40 of 57

1
2
3 2. Khine et al. [89] (2005), A negative pressure had been applied to pull cell towards
4
5 Valero el al. [130] trapping channel or microhole structures.
6
7 (2007).
8

pt
9
3. 2008; Ionescu- Zanetti Cells are trapped in the capillary channel by slight
10
11 et al. [144] transient negative pressure
12
13

cri
14 4. Valley et al. [135] Optically induced dielectrophoretic (DEP) force (so-called
15
16
(2009), Boukany et al. optical tweezers)
17 [75] (2011), Valley et
18
19 al. [135] (2009).
20

us
21
22
5. Fei et al. [136] (2010), A vacuum was used to trap the cells
23 Chang et al. [138] on the support membrane. (PSCEP)
24
25 (2016).
26
27
28
29
6. Wang et al.
an
[137] Electrically assisted DEP force
(2010), Bertani et al.
30
31 [145] (2015), Wang et
32
dM
33 al. [137] (2010).
34
35 7. Jokilaakso et al. [132] Magnetic beads
36
37 (2013).
38
39
40
41
42
43 7. Applications
pte

44
45 Precise control on drug/gene delivery with minimal external applied voltage irrespective of cell
46
47 and cargo types make SCEP an attractive platform for biomedical and therapeutic research.
48
49 SCEP is widely applicable to investigate cell to cell variation with their intracellular
50
51
characterization [55]. The selective cell to cell fusion can be achieved with this technique which
ce

52 might be useful for single-cell cloning and in-vitro fertilization [146]. Due to advantage of
53
54 micro/nano- fabrication and miniaturization of electrodes at nanoscale level, it is possible for
55
56 selective organelles manipulation within a cell [147]. SCEP is useful to record action potential
Ac

57
and transmembrane potential from electroactive cells and tissue, which is potentially applicable
58
59 for biomedical research such as electrophysiological study during cell development and
60

40
Page 41 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 pharmaceutical screening [90]. Most exciting achievement is to maintain cell viability for long
4
5 time after performance of electroporation, enabling real-time study of interaction between
6
7 impermeable molecules in the living cell. Several studies have been conducted in last two
8
decades including intracellular delivery of RNA, DNA, proteins and their complexes. In recent

pt
9
10 years, researchers extensively focus on their direct biomedical applications such as adoptive
11
12 immunotherapy, gene/RNA-based therapies, cell reprogramming, and intracellular bio-
13

cri
14 interrogation of living cells etc. Here we have discussed recent progress in this field. The
15
uptake of small molecules (e.g., dyes), and RNA has been successfully tested on a variety of
16
17 cell types, including human cells, hamster cell, yeast, and bacteria [53,75,131]. Also, plasmid
18
19 DNA or macromolecules like quantum dots have been successfully delivered in vitro [75] as
20

us
21 well as in vitro on a living tadpole brain on single cell level [114]. Nanochannel electroporation
22 device has been used to analyze a different kind of the RNAs such as messenger RNA(mRNA),
23
24 micro RNA (miRNA) in single cell [53].
25
26
27
28
29
an
Mutations of the GATA2 gene have been associated with leukemia. Bertani et al. [145] used
3D NEP chip to drive GATA2 molecular beacons into human immortalized myclogeneous
30 leukemia cell via electroporation. The molecular beacon opens up and binds to GATA2 present
31
32 within cell, detected via fluorescence. Thus it shows a greater prospect to study the
dM
33
34
hematopoietic stem cells important for leukemia disease analysis and drug discovery in future.
35 Electroporation technique is highly applicable for transdermal drug delivery, where insulin-
36
37 loaded nanocarriers are delivered into the blood for diabetes treatment [148]. However, for
38
39 diabetes diagnosis, the glucose concentration variations are needed to detect under ambient
40
condition. Thus it is necessary to characterize beta cells accurately at single cell level, which
41
42 produce insulin [149]. Moreover, transdermal drug delivery by electroporation is by far not
43
pte

44 limited to insulin.
45
46
SCEP technique has some drawback. As for example, when fabricated electrodes are dipped
47
48 far apart in the media, the current flows through the entire solution resulting in Joule heating
49
50 and chemical cross contaminations due to electrode reactions at the electrode-solution interface
51
ce

52 and the formation of an electric double layer at the surface of the electrodes leading to reduction
53 of applied electric field strength. Although whole-cell patch-clamp methods [6, 7] overcome
54
55 many of the electrophysiological problems of sharp electrode recording, they are laborious and
56
Ac

57 invasive. Therefore, whole-cell patch clamping has a low efficiency and limited reliability for
58
59
sequentially obtaining physiological and morphological data from multiple cells.
60

41
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 42 of 57

1
2
3 8. Future prospects
4
5
6 Single cell electroporation techniques were introduced in the past two decades for biological
7
8 and therapeutic research. It enables to study cell-to-cell variation in a population; to investigate

pt
9
10
and manipulate intra-cellular chemistry of cells. Despite all new approaches,
11 their delivery performance and reliability have not yet been approved to exhibit their full
12
13 potential to biological researchers and users. This can be assigned to several drawbacks

cri
14
15 associated with the current SCEP systems: (a) relatively complicated operational steps for
16
direct use in biological fields; (b) each configuration has its own advantages and limitations
17
18 regarding the cost and performance; and (c) inadequate database for clinically important
19
20 molecular probes and cells to verify the relevance.

us
21
22
The commercialization of the aforementioned new approaches demands easy-to-use strategies
23
24 along with their excellent performance and reliability. In this regards, user-friendly high-level
25
26 automation is highly encouraged and expected by the end users in this twenty-first century.
27
28
29
an
Furthermore, the whole operating system should be well integrated into one single system to
utilize the full potential of SCEP. For example, in situ imaging of single cell analysis, which is
30
31 a well-established technique, has to be well integrated with electroporation device for gene
32
dM
33 transfection. High-throughput parallel single cell electroporations approach with high
34
35
uniformity can have interesting future prospects for studying gene delivery and subsequent
36 protein expressions [135,136,150].
37
38
39 So far demonstrated SCEP devices utilize Lab-on-a-Chip or bio-micro electro-mechanical
40
41
systems (Bio-MEMS) technologies for device fabrication. The most common concerns of those
42 devices are that they require many expensive and complicated equipment. Furthermore, the
43
pte

44 outputs are mostly stochastic increasing the number of trials and repetition of the same
45
46 expensive steps and lot of chemicals. Therefore, optimization only expanse a lot of intellectual
47
effort besides the equipment and steps. Further concerns rely on unwanted leakage, non-
48
49 repeatability of operational outcome, post-delivery unwanted issues such as channel blockage
50
51 by cells, corrosion of device surfaces and electrodes due to chemical reaction etc. These issues
ce

52
53 have to be handled in a serious manner to envision future ready-to-use SCEP systems. So far
54 demonstrated SCEP devices, the utilization of micro/nano-fluidic and Lab-on-a-chip
55
56 technology integrated with micro total analysis systems (μ-TAS) can provide high throughput
Ac

57
58 single cell data with minimum reagent consumption [151]. The bioinformatics technique can
59
60

42
Page 43 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 employ to study this large ensemble of SCEP data to find answers of unresolved questions in
4
5 past centuries.
6
7
8 In the light of recent progress, in particular, the high throughput parallel single cell

pt
9 electroporations techniques, we can say that we are not very far from our envision of utilizing
10
11 those devices for human resources. We need to face few more challenges. For example, most
12
13 of the newly fabricated devices use RNA, DNA plasmid for demonstrating the device

cri
14
15
performance, basically testing delivery efficiency and cell viability. But, there are very few
16 studies to utilize those devices for understanding the cytosol interaction with molecules of
17
18 medical interests which are very useful for disease diagnostic and therapeutic applications.
19
20 Such utilization demands extensive collaborative efforts among engineers, biologists,

us
21
physicists, chemists, and medical practitioners. At the present stage of progress in this
22
23 interdisciplinary field, more attention should be dedicated on the collective efforts for
24
25 collecting a database of important molecules in parallel with improving the device
26
27
28
29
quick concept demonstration.
an
performances and using them for studying meaningful intracellular delivery instead of just

30
31 9. Conclusions
32
dM
33
34 This review has discussed the evolution of the different techniques for single cell
35
36 electroporation from its origins to its future prospects for cellular therapy and drug discovery.
37
There are several approaches presented in more than a few hundreds scientific papers to study
38
39 electro-physio properties from cellular to sub-cellular level. The basic need of this method is
40
41 to extend the electro-cellular research in more controlled and precise way down to nanometer
42
43 scale and analyse sub-cellular biological structures by utilizing the advance bio-
pte

44 nanoengineering techniques. The attempts of applying nanoscience and nanoengineering have


45
46 already an impact and, proven its importance and still, a research area needed to be improved
47
48 more. However, methods like micro-pipette based electroporation techniques permit
49
50
simultaneous delivery of mutiple reagents into individual cell with high transfection efficiency
51
ce

and viability. The nanopiller elctroporation help to record intracelluar action potentials, that
52
53 play key role in nervous system and cellular process. Smart designs of integrated nanofluidic
54
55 channels offer to study ion channel dynamics throughout the electroporation by measuring
56 current. The recents progress of high throughput parallel single cell delivery can have high
Ac

57
58 impact on biological science towards personalized medicine. The advent of micro-total analysis
59
60 systems integrated with Lab-ob-a-Chip devices will magnify its impact in future. Hopefully,

43
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 44 of 57

1
2
3 this review has presented sufficiently detailed description of the state-of-the-art to inspire the
4
5 readers and propose new solutions to analyse subcellular biological insights that remain to be
6
7 overcome.
8

pt
9
10 Acknowledgement
11
12 The authors greatly appreciate for the financial support from Science and Engineering Research
13

cri
14 Boards (SERB), Department of Science and Technology (DST), Government of India, under
15
16 grant number ECR/2016/001945. We acknowledge all the publishers to obtain the copyright
17 permission.
18
19
20 Conflicts of Interest

us
21
22
23 The authors declared no conflicts of interest.
24
25
26 References
27
28
29
an
30
31 [1] Phalon C, Rao D D and Nemunaitis J 2010 Potential use of RNA interference in
32
dM
cancer therapy Expert Rev. Mol. Med. 12 e26
33
34
35 [2] Pecot C V, Calin G A, Coleman R L, Lopez-Berestein G and Sood A K 2011 RNA
36
37 interference in the clinic: challenges and future directions Nat. Rev. Cancer 11 59–67
38
39 [3] Lam J K W, Chow M Y T, Zhang Y and Leung S W S 2015 siRNA versus miRNA as
40
41 therapeutics for gene silencing Mol. Ther. - Nucleic Acids 4 e252-
42
43
pte

44
[4] Uprichard S L 2005 The therapeutic potential of RNA interference FEBS Lett. 579
45 5996–6007
46
47
48 [5] Thassu D, Deleers M and Pathak Y V. 2007 Nanoparticulate Drug Delivery Systems
49
50
(Taylor & Francis)
51
ce

52 [6] Duan L, Yan Y, Liu J, Wang B, Li P, Hu Q and Chen W 2016 Target delivery of
53
54 small interfering RNAs with Vitamin E-coupled nanoparticles for treating hepatitis C
55
56
Sci. Rep. 6 24867
Ac

57
58 [7] Lam J K W, Chow M Y T, Zhang Y and Leung S W S 2015 siRNA versus miRNA as
59
60 therapeutics for gene silencing Mol. Ther. - Nucleic Acids 4 e252

44
Page 45 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 [8] Sun J Y, Anand-Jawa V, Chatterjee S and Wong K K 2003 Immune responses to
4
5 adeno-associated virus and its recombinant vectors Gene Ther. 10 964–76
6
7
8 [9] Lehrman S 1999 Virus treatment questioned after gene therapy death. Nature 401

pt
9 517–8
10
11
12 [10] Tong H J, Shi Q, Fernandes J C, Liu L, Dai K R and Zhang X L 2009 Progress and
13
prospects of chitosan and its derivatives as non-viral gene vectors in gene therapy

cri
14
15 Curr. Gene Ther. 9 495–502
16
17
18 [11] Kingston R E, Chen C A and Rose J K 2003 Calcium Phosphate Transfection Curr.
19
20 Protoc. Mol. Biol. 63 9.1.1-9.1.11

us
21
22 [12] Xia J, Martinez A, Daniell H and Ebert S N 2011 Evaluation of biolistic gene transfer
23
24 methods in vivo using non-invasive bioluminescent imaging techniques BMC
25
26 Biotechnol. 11 62
27
28
29
[13]
an
Yoon C S and Park J H 2010 Ultrasound-mediated gene delivery Expert Opin. Drug
30 Deliv. 7 321–30
31
32
dM
33
[14] Truong H H, de Sonneville J, Ghotra V P S, Xiong J, Price L, Hogendoorn P C W,
34 Spaink H H, van de Water B and Danen E H J 2012 Automated microinjection of cell-
35
36 polymer suspensions in 3D ECM scaffolds for high-throughput quantitative cancer
37
38 invasion screens Biomaterials 33 181–8
39
40 [15] Pepperkok R, Schneider C, Philipson L and Ansorge W 1988 Single cell assay with an
41
42 automated capillary microinjection system Exp. Cell Res. 178 369–76
43
pte

44
45
[16] Funahashi K, Sawai S, Santra T S, Shibata T and Nagai M 2017 Optical transfection
46 system using pulse laser for massively parallel localized intracellular delivery 2017
47
48 International Symposium on Micro-NanoMechatronics and Human Science (MHS) pp
49
50 1–3
51
ce

52 [17] Santra T S, Wu T-H and Chiou P Y 2015 Photothermal microfluidics (IET USA Inc.)
53
54
55 [18] Xiong R, Raemdonck K, Peynshaert K, Lentacker I, De Cock I, Demeester J, De
56
Smedt S C, Skirtach A G and Braeckmans K 2014 Comparison of gold nanoparticle
Ac

57
58 mediated photoporation: Vapor nanobubbles outperform direct heating for delivering
59
60 macromolecules in live cells ACS Nano 8 6288–96

45
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 46 of 57

1
2
3 [19] Jiang R, Meng L, Sun L, He X, Liang X, Zhang J and Zhang Z 2016 Microwave
4
5 ablation-assisted liver gene transfection in rats Int. J. Hyperth. 32 666–72
6
7
8 [20] Nguyen T H P, Shamis Y, Croft R J, Wood A, McIntosh R L, Crawford R J and

pt
9 Ivanova E P 2015 18 GHz electromagnetic field induces permeability of Gram-positive
10
11 cocci Sci. Rep. 5 10980
12
13
[21] Doran T J, Lu P J, Vanier G S, Collins M J, Wu B and Lu Q L 2009 Microwave

cri
14
15 irradiation enhances gene and oligonucleotide delivery and induces effective exon
16
17 skipping in myoblasts Gene Ther. 16 119–26
18
19
20 [22] Seidlits S K, Gower R M, Shepard J A and Shea L D 2013 Hydrogels for lentiviral

us
21 gene delivery Expert Opin. Drug Deliv. 10 499–509
22
23
24 [23] Luo D and Saltzman W M 2000 Synthetic DNA delivery systems Nat. Biotechnol. 18
25
26 33–7
27
28
29
[24]
an
Wong I Y, Almquist B D and Melosh N A 2010 Dynamic actuation using nano-bio
30 interfaces Mater. Today 13 14–22
31
32
dM
33
[25] Santra T S and Tseng F-G 2016 Essentials for single cell analysis
34
35 [26] Nollet A Recherches Sur Les Causes Particulieres Des Phenomenes Electriques
36
37
38 [27] Ivorra A. R B 2010 Irreversible Electroporation (Springer, Berlin, Heidelberg) pp 1–
39 21
40
41
42 [28] Yu C and Peng R-Y 2017 Biological effects and mechanisms of shortwave radiation:
43
pte

44 a review Mil. Med. Res. 4 24


45
46 [29] Hodgkin A L 1951 The ionic basic of electrical activity in nurve and muscle Biol. Rev.
47
48 26 339–409
49
50
[30] He H, Chang D C and Lee Y K 2007 Using a micro electroporation chip to determine
51
ce

52 the optimal physical parameters in the uptake of biomolecules in HeLa cells


53
54 Bioelectrochemistry 70 363–8
55
56
[31] Santra T S and Tseng F G 2013 Recent trends on micro/nanofluidic single cell
Ac

57
58 electroporation Micromachines 4 333–56
59
60

46
Page 47 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 [32] Santra T, Wang and P-C and Tseng F-G 2013 Advances in Micro/Nano
4
5 Electromechanical Systems and Fabrication Technologies
6
7
8 [33] Qian M, Wang D C, Chen H and Cheng Y 2017 Detection of single cell heterogeneity

pt
9 in cancer Semin. Cell Dev. Biol. 64 143–9
10
11
12 [34] Tseng F-G and Santra T S 2016 Single Cell Analysis in Biotechnology and System
13
Biology (MDPI)

cri
14
15
16 [35] Ilic B, Czaplewski D, Zalalutdinov M, Craighead H G, Neuzil P, Campagnolo C, Batt
17
18 C, Ilic B, Czaplewski D, Zalalutdinov M and Craighead H G 2001 Single cell
19
20 detection with micromechanical oscillators Single cell detection with micromechanical

us
21 oscillators J. Vac. Sci. Technol. B 19 2825–8
22
23
24 [36] Yasukawa T, Kaya T and Matsue T 2000 Characterization and Imaging of Single
25
26 Cells with Scanning Electrochemical Microscopy Electroanalysis 12 653–9
27
28
29
[37]
an
Heyn C, Ronald J A, Mackenzie L T, Macdonald I C, Chambers A F, Rutt B K and
30 Foster P J 2006 In Vivo Magnetic Resonance Imaging of Single Cells in Mouse Brain
31
32 with Optical Validation Magn. Reson. Med. 29 23–9
dM
33
34 [38] Hinds K A, Hill J M, Shapiro E M, Laukkanen M O, Silva A C, Combs C A, Varney
35
36 T R, Balaban R S, Koretsky A P and Dunbar C E 2003 Highly efficient endosomal
37
38 labeling of progenitor and stem cells with large magnetic particles allows magnetic
39
resonance imaging of single cells Blood 102 867–72
40
41
42 [39] DeLong E F, Wickham G S and Pace N R 1989 Phylogenetic stains: ribosomal RNA-
43
pte

44 based probes for the identification of single cells Science (80-. ). 243 1360 LP-1363
45
46 [40] Duan R, Zuo X, Wang S, Quan X, Chen D, Chen Z, Jiang L, Fan C and Xia F 2013
47
48 Lab in a Tube: Ultrasensitive Detection of MicroRNAs at the Single- Cell Level and in
49
50 Breast Cancer Patients Using Quadratic Isothermal Ampli fi cation JACS 135 4604–7
51
ce

52 [41] Park H, Kim D and Yun K S 2010 Single-cell manipulation on microfluidic chip by
53
54 dielectrophoretic actuation and impedance detection Sensors Actuators, B Chem. 150
55
56 167–73
Ac

57
58 [42] Wang J, Wang M and Jang L 2010 Effects of electrode geometry and cell location on
59
60 single-cell impedance measurement Biosens. Bioelectron. 25 1271–6

47
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 48 of 57

1
2
3 [43] Santra T S and Tseng F-G 2014 Micro/Nanofluidic devices for single cell analysis
4
5 Micromachines 5 154–7
6
7
8 [44] Tseng F-G and Santra T S 2015 Micro/Nano Fluidic Devices for Single Cell Analysis

pt
9
10 [45] Teruel M N and Meyer T 1997 Electroporation-induced formation of individual
11
12 calcium entry sites in the cell body and processes of adherent cells Biophys. J. 73
13
1785–96

cri
14
15
16 [46] Lundqvist J a, Sahlin F, Aberg M a, Strömberg a, Eriksson P S and Orwar O 1998
17
18 Altering the biochemical state of individual cultured cells and organelles with
19
20 ultramicroelectrodes. Proc. Natl. Acad. Sci. U. S. A. 95 10356–60

us
21
22 [47] Chang D C 1989 Poration Using October 56 641–52
23
24
25 [48] Santra T S, Wang P C, Chang H Y and Tseng F G 2013 Tuning nano electric field to
26
27
28
29
Phys. Lett. 103
an
affect restrictive membrane area on localized single cell nano-electroporation Appl.

30
31 [49] Santra T, Chen S, Chang C and Chen T 2012 Electrically Mediated Gene Delivery
32
dM
33
and Their Diffusion Mechanism on Localized Single Cell Using Ito Microelectrode
34 Based Transparent Chip µTAS - 16th Int. Conf. Miniaturized Syst. Chem. Life Sci. 1
35
36 962–4
37
38
39
[50] Santra T S, Kar S, Borana J, Wang P-C and Tseng F-G 2014 Nanolocalized single cell
40 membrane nanoelectroporation IEEE Nanotechnol. Mag. 30–4
41
42
43 [51] Kang W, Yavari F, Minary-jolandan M, Giraldo-vela J P, Sa A, Mcnaughton R L,
pte

44
45
Parpoil V and Espinosa H D 2013 Nanofountain Probe Electroporation (NFP-E) of
46 Single Cells Nano Lett. 13 2448–57
47
48
49 [52] Ainla A, Jeffries G D M, Brune R, Orwar O and Jesorka A 2012 A multifunctional
50
51
pipette Lab Chip 12 1255
ce

52
53 [53] Zhao X, Huang X, Wang X, Wu Y, Eisfeld A K, Schwind S, Gallego-Perez D,
54
55 Boukany P E, Marcucci G I and Lee L J 2015 Nanochannel Electroporation as a
56
Platform for Living Cell Interrogation in Acute Myeloid Leukemia Adv. Sci. 2 1–5
Ac

57
58
59 [54] Ding X, Stewart M P, Sharei A, Weaver J C, Langer R S and Jensen K F 2017 High-
60

48
Page 49 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 throughput nuclear delivery and rapid expression of DNA via mechanical and
4
5 electrical cell-membrane disruption Nat. Biomed. Eng. 1
6
7
8 [55] Olafsson J, Nolkrantz K, Ryttsen F, Lambie B A, Weber S G and Orwar O 2003

pt
9 Single-cell electroporation Curr. Opin. Biotechnol. 14 29–34
10
11
12 [56] Wang M, Orwar O, Olofsson J and Weber S G 2010 Single-cell electroporation Anal.
13
Bioanal. Chem. 397 3235–48

cri
14
15
16 [57] Chang L, Li L, Shi J, Sheng Y, Lu W, Gallego-Perez D and Lee L J 2016 Micro-
17
18 /nanoscale electroporation Lab Chip 16 4047–62
19
20
[58] Fox M B, Esveld D C, Valero A, Luttge R, Mastwijk H C, Bartels P V., Van Den

us
21
22 Berg A and Boom R M 2006 Electroporation of cells in microfluidic devices: A review
23
24 Anal. Bioanal. Chem. 385 474–85
25
26
27
28
29
[59]
an
Chen C, Smye S W, Robinson M P and Evans J A 2006 Membrane electroporation
theories: A review Med. Biol. Eng. Comput. 44 5–14
30
31 [60] Tsong T Y 1991 Electroporation of cell membranes. Minireview. Biophys. J. 60 297–
32
dM
33
306
34
35 [61] Weaver J C 2003 Electroporation of Biological Membranes from Multicellular to
36
37 Nano Scales IEEE Trans. Dielectr. Electr. Insul. 10 754–68
38
39 [62] Weaver J C and Chizmadzhev Y A 1996 Theory of electroporation: A review
40
41 Bioelectrochemistry Bioenerg. 41 135–60
42
43
pte

44 [63] Pavlin M, Kotnik T, Miklavčič D, Kramar P and Maček Lebar A 2008 Advances in
45 Planar Lipid Bilayers and Liposomes Academic Press vol 6, ed A L Liu (Academic
46
47 Press) pp 165–226
48
49
50 [64] Neu J C and Krassowska W 1999 Asymptotic model of electroporation Phys. Rev. E -
51
ce

Stat. Physics, Plasmas, Fluids, Relat. Interdiscip. Top. 59 3471–82


52
53
54 [65] Krassowska W and Filev P D 2007 Modeling electroporation in a single cell Biophys.
55
56 J. 92 404–17
Ac

57
58 [66] Lewis T J 2003 A Model for Bilayer Membrane Electroporation Based on Resultant
59
60 Electromechanical Stress IEEE Trans. Dielectr. Electr. Inulation 10 769–77

49
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 50 of 57

1
2
3 [67] Zimmermann U, Pilwat G, Beckers F and Riemann F 1976 Effects of external
4
5 electrical fields on cell membranes Bioelectrochemistry Bioenerg. 3 58–83
6
7
8 [68] Crowley J M 1973 Electrical Breakdown of Bimolecular Lipid Membranes as an

pt
9 Electromechanical Instability Biophys. J. 13 711–24
10
11
12 [69] Kotnik T, Pucihar G and Miklavčič D 2010 Induced transmembrane voltage and its
13
correlation with electroporation- mediated molecular transport J. Membr. Biol. 236 3–

cri
14
15 13
16
17
18 [70] Debruin K A and Krassowska W 1998 Electroporation and shock-induced
19
20 transmembrane potential in a cardiac fiber during defibrillation strength shocks Ann.

us
21 Biomed. Eng. 26 584–96
22
23
24 [71] Santra T S, Chiu C, Agarwal N, Ganatathi A, Wang P and Tseng F 2013 Nanofocused
25
26 Electric Field for Localized Single Cell Nanoelectroporation with Membrane
27
28
29
an
Reversibility IEEE NEMS vol 1 pp 961–4

30 [72] Santra T S, Chang H and Wang P 2014 Impact of pulse duration on localized single-
31
32 cell nano-electroporation Analyst 139 6249–58
dM
33
34 [73] Teissie J, Golzio M and Rols M P 2005 Mechanisms of cell membrane
35
36 electropermeabilization: A minireview of our present (lack of ?) knowledge Biochim.
37
38 Biophys. Acta - Gen. Subj. 1724 270–80
39
40 [74] Santra T S, Wang P-C and Tseng F-G 2013 Nano-intensified electric field for multi-
41
42 localized single cell electroporation 17th International Conference on Miniatnrized
43
pte

44 Systems for Chemistry and Life Sciences 27-31 October 2013, Freiburg, Germany pp
45
1036–8
46
47
48 [75] Boukany P E, Morss A, Liao W C, Henslee B, Jung H, Zhang X, Yu B, Wang X, Wu
49
50 Y, Li L, Gao K, Hu X, Zhao X, Hemminger O, Lu W, Lafyatis G P and Lee L J 2011
51
ce

Nanochannel electroporation delivers precise amounts of biomolecules into living cells


52
53 Nat. Nanotechnol. 6 747–54
54
55
56 [76] Gao K, Li L, He L, Hinkle K, Wu Y, Ma J, Chang L, Zhao X, Perez D G, Eckardt S,
Ac

57
McLaughlin J, Liu B, Farson D F and Lee L J 2014 Design of a microchannel-
58
59 nanochannel-microchannel array based nanoelectroporation system for precise gene
60

50
Page 51 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 transfection Small 10 1015–23
4
5
6 [77] Santra T S, Lee C W, Kar S, Borana J, Wang P C and Tseng F G 2014 Nanolocalized
7
8 single cell membrane nanoelectroporation 9th IEEE International Conference on

pt
9 Nano/Micro Engineered and Molecular Systems, IEEE-NEMS 2014 vol 2 pp 285–8
10
11
12 [78] Rubinsky B 2010 Irreversible Electroporation ed J H Nagel (Springer)
13

cri
14
[79] Kotnik T, Kramar P, Pucihar G, Miklavčič D and Tarek M 2012 Cell membrane
15
16 electroporation - Part 1: The phenomenon IEEE Electr. Insul. Mag. 28 14–23
17
18
19 [80] Freeman S A, Wang M A and Weaver J C 1994 Theory of electroporation of planar
20
bilayer membranes: predictions of the aqueous area, change in capacitance, and pore-

us
21
22 pore separation Biophys. J. 67 42–56
23
24
25 [81] Joshi R P and Schoenbach K H 2000 Electroporation dynamics in biological cells
26
27
28
29
an
subjected to ultrafast electrical pulses: A numerical simulation study Phys. Rev. E -
Stat. Physics, Plasmas, Fluids, Relat. Interdiscip. Top. 62 1025–33
30
31 [82] O’Neill R J and Tung L 1991 Cell-attached patch clamp study of the
32
dM
33
electropermeabilization of amphibian cardiac cells Biophys. J. 59 1028–39
34
35 [83] Santra T S, Lee C W, Kar S, Borana J, Wang P C and Tseng F G 2014 Nanolocalized
36
37 single cell membrane nanoelectroporation 9th IEEE International Conference on
38
39
Nano/Micro Engineered and Molecular Systems, IEEE-NEMS 2014 pp 285–8
40
41 [84] Santra T and Tseng P 2013 Nano-Intensified Electric Field for Multi-Localized Sin
42
43 Gle Cell Electroporation 17th Int. Conf. Miniaturized Syst. Chem. Life Sci. 1036–8
pte

44
45 [85] Saulis G and Saulė R 2012 Size of the pores created by an electric pulse: Microsecond
46
47 vs millisecond pulses Biochim. Biophys. Acta - Biomembr. 1818 3032–9
48
49
50 [86] Zhao Y, Inayat S, Dikin D A, Singer J H, Ruoff R S and Troy J B 2008 Patch clamp
51
ce

technique: Review of the current state of the art and potential contributions from
52
53 nanoengineering Proc. Inst. Mech. Eng. Part N J. Nanoeng. Nanosyst. 222 1–11
54
55
56 [87] Chang C C, Mao M, Liu Y, Wu M, Vo-Dinh T and Yuan F 2016 Improvement in
Ac

57 Electrotransfection of Cells Using Carbon-Based Electrodes Cell. Mol. Bioeng. 9 538–


58
59 45
60

51
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 52 of 57

1
2
3 [88] Chafai D E, Mehle A, Tilmatine A, Maouche B and Miklavčič D 2015 Assessment of
4
5 the electrochemical effects of pulsed electric fields in a biological cell suspension
6
7 Bioelectrochemistry 106 249–57
8

pt
9 [89] Khine M, Lau A, Ionescu-Zanetti C, Seo J and Lee L P 2005 A single cell
10
11 electroporation chip Lab Chip 5 38
12
13
[90] Xie C, Lin Z, Hanson L, Cui Y and Cui B 2012 Intracellular recording of action

cri
14
15 potentials by nanopillar electroporation Nat. Nanotechnol. 7 185
16
17
18 [91] Exley C and Derek Birchall J 1992 The cellular toxicity of aluminium J. Theor. Biol.
19
20 159 83–98

us
21
22 [92] Kim J A, Cho K, Shin M S, Lee W G, Jung N, Chung C and Chang J K 2008 A novel
23
24 electroporation method using a capillary and wire-type electrode Biosens. Bioelectron.
25
26 23 1353–60
27
28
29
[93]
an
Loomis-Husselbee J W, Cullen P J, Irvine R F and Dawson A P 1991 Electroporation
30 can cause artefacts due to solubilization of cations from the electrode plates.
31
32 Aluminum ions enhance conversion of inositol 1,3,4,5-tetrakisphosphate into inositol
dM
33 1,4,5-trisphosphate in electroporated L1210 cells. Biochem. J. 277 883–5
34
35
36 [94] Geddes L A, Baker L E and Moore A G 1969 Optimum electrolytic chloriding of
37
38 silver electrodes Med. Biol. Eng. 7 49–56
39
40 [95] Grubbs D S and Worley D S 1983 New technique for reducing the impedance of
41
42 silver-silver chloride electrodes Med. Biol. Eng. Comput. 21 232–4
43
pte

44
45
[96] Li S 2006 Optimizing Electrotransfection of Mammalian Cells In Vitro Cold Spring
46 Harb. Protoc. 2006 pdb.prot4449
47
48
49 [97] Potter H and Heller R 2003 Transfection by Electroporation Curr. Protoc. Mol. Biol.
50
51
CHAPTER Unit-9.3
ce

52
53 [98] Rols M-P, Delteil C, Serin G and Teissié J 1994 [1] M.-P. Rols, C. Delteil, G. Serin,
54
55 and J. Teissié, Nucleic Acids Res. 22, 540 (1994).Temperature effects on
56
electrotransfection of mammalian cells Nucleic Acids Res. 22 540
Ac

57
58
59 [99] Meglič S H, Levičnik E, Luengo E, Raso J and Miklavčič D 2016 The Effect of
60

52
Page 53 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 Temperature on Protein Extraction by Electroporation and on Bacterial Viability BT -
4
5 1st World Congress on Electroporation and Pulsed Electric Fields in Biology,
6
7 Medicine and Food & Environmental Technologies ed T Jarm and P Kramar
8
(Singapore: Springer Singapore) pp 175–8

pt
9
10
11 [100] Diaz-Rivera R and Rubinsky B 2017 The effect of temperature on the critical electric
12
13 field for electroporation: A single cell electroporation device study vol 1

cri
14
15 [101] Erickson D, Sinton D and Li D 2003 Joule heating and heat transfer in
16
17 poly(dimethylsiloxane) microfluidic systems Lab Chip 3 141–9
18
19
20 [102] Di Carlo D, Aghdam N and Lee L P 2006 Single-cell enzyme concentrations, kinetics,

us
21 and inhibition analysis using high-density hydrodynamic cell isolation arrays Anal.
22
23 Chem. 78 4925–30
24
25
26 [103] Rao C V., Wolf D M and Arkin A P 2002 Control, exploitation and tolerance of
27
28
29
intracellular noise Nature 420 231–7
an
30 [104] Ko M S H, Nakauchi H and Takahashi N 1990 expression results from changes in the
31
32 number of transcriptionally active templates 9 2835–42
dM
33
34 [105] Steinmeyer J D and Yanik M F 2012 High-throughput single-cell manipulation in
35
36 brain tissue PLoS One 7 1–10
37
38
39
[106] Ainla A, Xu S, Sanchez N, Jeffries G D M and Jesorka A 2012 Single-cell
40 electroporation using a multifunctional pipette Lab Chip 12 4605–9
41
42
43 [107] Huang Y and Rubinsky B 2001 Microfabricated electroporation chip for single cell
pte

44
45
membrane permeabilization Sensors Actuators, A Phys. 89 242–9
46
47 [108] Huang Y and Rubinsky B 1999 Micro-Electroporation: Improving the Efficiency and
48
49 Understanding of Electrical Permeabilization of Cells Biomed. Microdevices 2 145–50
50
51
ce

[109] Ainla A, Jansson E T, Stepanyants N, Orwar O and Jesorka A 2010 A microfluidic


52
53 pipette for single-cell pharmacology Anal. Chem. 82 4529–36
54
55
56 [110] Ohmachi M, Fujiwara Y, Muramatsu S, Yamada K, Iwata O, Suzuki K and Wang D
Ac

57 O 2016 A modified single-cell electroporation method for molecule delivery into a


58
59 motile protist, Euglena gracilis J. Microbiol. Methods 130 106–11
60

53
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 54 of 57

1
2
3 [111] Tanaka M, Yanagawa Y and Hirashima N 2009 Transfer of small interfering RNA by
4
5 single-cell electroporation in cerebellar cell cultures J. Neurosci. Methods 178 80–6
6
7
8 [112] Boudes M, Pieraut S, Valmier J, Carroll P and Scamps F 2008 Single-cell

pt
9 electroporation of adult sensory neurons for gene screening with RNA interference
10
11 mechanism J. Neurosci. Methods 170 204–11
12
13
[113] Rathenberg J, Nevian T and Witzemann V 2003 High-efficiency transfection of

cri
14
15 individual neurons using modified electrophysiology techniques J. Neurosci. Methods
16
17 126 91–8
18
19
20 [114] Bestman J E, Ewald R C, Chiu S-L and Cline H T 2006 In vivo single-cell

us
21 electroporation for transfer of DNA and macromolecules Nat. Protoc. 1 1267–72
22
23
24 [115] Hewapathirane D S and Haas K 2008 Single Cell Electroporation in vivo within the
25
26 Intact Developing Brain J. Vis. Exp. 222 4–6
27
28
29
an
[116] XF L and K H 2011 Single-cell electroporation in Xenopus Cold Spring Harb Protoc
30 9 pii:pdb.top065607
31
32
dM
33
[117] Pagès S, Cane M, Randall J, Capello L and Holtmaat A 2015 Corrigendum: Single
34 cell electroporation for longitudinal imaging of synaptic structure and function in the
35
36 adult mouse neocortex in vivo Front. Neuroanat. 9 1–12
37
38
39
[118] Iwata F, Yamazaki K, Ishizaki K and Ushiki T 2014 Local electroporation of a single
40 cell using a scanning ion conductance microscope Jpn. J. Appl. Phys. 036701 036701
41
42
43 [119] Molleman A 2003 Patch Clamping: An Introductory Guide to Patch Clamp
pte

44
45
Electrophysiology (John Wiley & Sons, Ltd)
46
47 [120] Ryttsén F, Farre C, Brennan C, Weber S G, Nolkrantz K, Jardemark K, Chiu D T and
48
49 Orwar O 2000 Characterization of single-cell electroporation by using patch-clamp
50
51
and fluorescence microscopy Biophys. J. 79 1993–2001
ce

52
53 [121] Kitamura K, Judkewitz B, Kano M, Denk W and Häusser M 2008 Targeted patch-
54
55 clamp recordings and single-cell electroporation of unlabeled neurons in vivo Nat.
56
Methods 5 61–7
Ac

57
58
59 [122] Rae J L and Levis R A 2002 Single-cell electroporation Pflugers Arch. 443 664–70
60

54
Page 55 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 [123] Bürgel S C, Escobedo C, Haandbæk N and Hierlemann A 2015 On-chip
4
5 electroporation and impedance spectroscopy of single-cells Sensors Actuators, B
6
7 Chem. 210 82–90
8

pt
9 [124] Zhang X, Hu N, Chen X, Fan T, Wang Z, Zheng X, Yang J and Qian S 2017
10
11 Controllable cell electroporation using microcavity electrodes Sensors Actuators, B
12
13 Chem. 240 434–42

cri
14
15 [125] Chen S C, Santra T S, Chang C J, Chen T J, Wang P C and Tseng F G 2012 Delivery
16
17 of molecules into cells using localized single cell electroporation on ITO micro-
18
19 electrode based transparent chip Biomed. Microdevices 14 811–7
20

us
21 [126] Santra T S, Chen C-W, Chang H-Y and Tseng F-G 2016 Dielectric passivation layer
22
23 as a substratum on localized single-cell electroporation RSC Adv. 6 10979–86
24
25
26 [127] Santra T S, Borana J, Wang P and Tseng F 2014 Nanoelectroporation and controllable
27
28
29
an
intracellular delivery into localized single cell with high transfection and cell viability
MEMS 2014, San Francisco, CA, USA, January 26 - 30, 2014 pp 865–8
30
31
32 [128] Zheng M, Shan J W, Lin H, Shreiber D I and Zahn J D 2016 Hydrodynamically
dM
33 controlled cell rotation in an electroporation microchip to circumferentially deliver
34
35 molecules into single cells Microfluid. Nanofluidics 20 1–12
36
37
38 [129] Young H and Boris R 2003 Flow-through micro-electroporation chip for high
39
efficiency single-cell genetic manipulation Sensors Actuators, A Phys. 104 205–12
40
41
42 [130] Valero A, Post J N, van Nieuwkasteele J W, ter Braak P M, Kruijer W and van den
43
pte

44 Berg A 2008 Gene transfer and protein dynamics in stem cells using single cell
45
electroporation in a microfluidic device Lab Chip 8 62–7
46
47
48 [131] Boukany P E, Wu Y, Zhao X, Kwak K J, Glazer P J, Leong K and Lee L J 2014
49
50 Nonendocytic delivery of lipoplex nanoparticles into living cells using nanochannel
51
ce

electroporation Adv. Healthc. Mater. 3 682–9


52
53
54 [132] Jokilaakso N, Salm E, Chen A, Millet L, Guevara C D, Dorvel B, Reddy B, Karlstrom
55
56 A E, Chen Y, Ji H, Chen Y, Sooryakumar R and Bashir R 2013 Ultra-localized single
Ac

57
cell electroporation using silicon nanowires Lab Chip 13 336–9
58
59
60 [133] Yang R, Lemaître V, Huang C, Haddadi A, McNaughton R and Espinosa H D 2018

55
AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1 Page 56 of 57

1
2
3 Monoclonal Cell Line Generation and CRISPR/Cas9 Manipulation via Single-Cell
4
5 Electroporation Small 14 1702495
6
7
8 [134] Khine M, Ionescu-Zanetti C, Blatz A, Wang L-P and Lee L P 2007 Single-cell

pt
9 electroporation arrays with real-time monitoring and feedback control Lab Chip 7 457
10
11
12 [135] Valley J K, Neale S, Hsu H-Y, Ohta A T, Jamshidi A and Wu M C 2009 Parallel
13
single-cell light-induced electroporation and dielectrophoretic manipulation Lab Chip

cri
14
15 9 1714
16
17
18 [136] Fei Z, Hu X, Choi H W, Wang S, Farson D and Lee L J 2010 Micronozzle array
19
20 enhanced sandwich electroporation of embryonic stem cells Anal. Chem. 82 353–8

us
21
22 [137] Wang J, Yang S, Wang C, Wu Q and Wang Z 2010 A DEP-Assisted Single-Cell
23
24 Electroporation Chip with Low Operation Voltage IEEE SENSORS 2010 Conference
25
26 vol 1 pp 2097–100
27
28
29
an
[138] Chang L, Bertani P, Gallego-Perez D, Yang Z, Chen F, Chiang C, Malkoc V, Kuang
30 T, Gao K, Lee L J and Lu W 2016 3D nanochannel electroporation for high-
31
32 throughput cell transfection with high uniformity and dosage control Nanoscale 8 243–
dM
33 52
34
35
36 [139] Chang L, Howdyshell M, Liao W C, Chiang C L, Gallego-Perez D, Yang Z, Lu W,
37
38 Byrd J C, Muthusamy N, Lee L J and Sooryakumar R 2015 Magnetic tweezers-based
39
3D microchannel electroporation for high-throughput gene transfection in living cells
40
41 Small 11 1818–28
42
43
pte

44 [140] Chang L, Gallego-Perez D, Chiang C L, Bertani P, Kuang T, Sheng Y, Chen F, Chen


45
Z, Shi J, Yang H, Huang X, Malkoc V, Lu W and Lee L J 2016 Controllable Large-
46
47 Scale Transfection of Primary Mammalian Cardiomyocytes on a Nanochannel Array
48
49 Platform Small 12 5971–80
50
51
ce

[141] Guo X and Zhu R 2016 Controllable in-situ cell electroporation with cell positioning
52
53 and impedance monitoring using micro electrode array Sci. Rep. 6 31392
54
55
56 [142] Zheng Y, Nguyen J, Wei Y and Sun Y 2013 Recent advances in microfluidic
Ac

57
techniques for single-cell biophysical characterization Lab Chip 13 2464
58
59
60 [143] Huang Y and Rubinsky B 2000 Micro-Electroporation Improving the Ef ® ciency and

56
Page 57 of 57 AUTHOR SUBMITTED MANUSCRIPT - JMM-103716.R1

1
2
3 Understanding of Electrical Permeabilization of Cells - Huang, Rubinsky - 2000.pdf
4
5 145–50
6
7
8 [144] Ionescu-Zanetti C, Blatz A and Khine M 2008 Electrophoresis-assisted single-cell

pt
9 electroporation for efficient intracellular delivery Biomed. Microdevices 10 113–6
10
11
12 [145] Bertani P, Lu W, Chang L, Gallego-Perez D, Lee L J, Chiang C and Muthusamy N
13
2015 Bosch etching for the creation of a 3D nanoelectroporation system for high

cri
14
15 throughput gene delivery J. Vac. Sci. Technol. B 33 06F903
16
17
18 [146] Strömberg a, Ryttsén F, Chiu D T, Davidson M, Eriksson P S, Wilson C F, Orwar O
19
20 and Zare R N 2000 Manipulating the genetic identity and biochemical surface

us
21 properties of individual cells with electric-field-induced fusion. Proc. Natl. Acad. Sci.
22
23 U. S. A. 97 7–11
24
25
26 [147] Andersson H and Van Den Berg A 2004 Microtechnologies and nanotechnologies for
27
28
29
an
single-cell analysis Curr. Opin. Biotechnol. 15 44–9

30 [148] Rastogi R, Anand S and Koul V 2010 Electroporation of polymeric nanoparticles: An


31
32 alternative technique for transdermal delivery of insulin Drug Dev. Ind. Pharm. 36
dM
33 1303–11
34
35
36 [149] Kim K and Lee W G 2017 Electroporation for nanomedicine: a review J. Mater.
37
38 Chem. B 5 2726–38
39
40 [150] Chang L, Hu J, Chen F, Chen Z, Shi J, Yang Z, Li Y and Lee L J 2016 Nanoscale bio-
41
42 platforms for living cell interrogation: current status and future perspectives Nanoscale
43
pte

44 8 3181–206
45
46 [151] Neužil P, Campos C D M, Wong C C, Soon J B W, Reboud J and Manz A 2014 From
47
48 chip-in-a-lab to lab-on-a-chip: Towards a single handheld electronic system for
49
50 multiple application-specific lab-on-a-chip (ASLOC) Lab Chip 14 2168–76
51
ce

52
53
54
55
56
Ac

57
58
59
60

57

You might also like