You are on page 1of 14

Nanotechnology

PAPER You may also like


- Anomalous mobility of a driven active
Advanced nanostructures for cell membrane particle in a steady laminar flow
F Cecconi, A Puglisi, A Sarracino et al.
poration - Local time of diffusion with stochastic
resetting
Arnab Pal, Rakesh Chatterjee, Shlomi
To cite this article: Apresio K Fajrial and Xiaoyun Ding 2019 Nanotechnology 30 264002 Reuveni et al.

- On characterising assemblages in
Einstein–Podolsky–Rosen scenarios
Vinicius P Rossi, Matty J Hoban and Ana
Belén Sainz
View the article online for updates and enhancements.

This content was downloaded from IP address 175.159.124.104 on 02/12/2022 at 03:06


Nanotechnology

Nanotechnology 30 (2019) 264002 (13pp) https://doi.org/10.1088/1361-6528/ab096b

Advanced nanostructures for cell membrane


poration
Apresio K Fajrial and Xiaoyun Ding
Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309 United
States of America

E-mail: xiaoyun.ding@colorado.edu

Received 1 November 2018, revised 21 January 2019


Accepted for publication 22 February 2019
Published 12 April 2019

Abstract
Nanostructured devices are able to foster the technology for cell membrane poration. With the
size smaller than a cell, nanostructures allow efficient poration on the cell membrane. Emerging
nanostructures with various physical transduction have been demonstrated to accommodate
effective intracellular delivery. Aside from improving poration and intracellular delivery
performance, nanostructured devices also allow for the discovery of novel physiochemical
phenomena and the biological response of the cell. This article provides a brief introduction to
the principles of nanostructured devices for cell poration and outlines the intracellular delivery
capability of the technology. In the future, we envision more exploration on new nanostructure
designs and creative applications in biomedical fields.
Keywords: cell membrane disruption, cell poration, intracellular delivery, nanostructure

(Some figures may appear in colour only in the online journal)

1. Introduction gradient across the cell. The local electric field gradient
around the plasma membrane drives the water molecule thus
The capability to access the inner compartment of mammalian increasing the chance for water penetration that at the same
cells is important for biomedical applications. One such time produces pores on the membrane [7]. In 1982, Neumann
example, such as increasing the permeability of the cell et al demonstrated that electroporation is able to transfer
membrane, allows the delivery of chemicals or genetic agents genes into mouse fibroblast cells and it has been popularly
into the cell which is the stepping stone in manipulating cell used for transfecting genes to other types of cell ever since
behavior. The introduction of foreign materials into cells [8]. The molecular transport on the porated membrane can be
could induce various cell responses such as modulation of facilitated by electrical drift for large molecules or diffusion
gene expression and differentiation of specific cell [1–5]. for small molecules [9]. Unfortunately, the common bulk
Besides cargo delivery, intentionally forming pores in the cell process of electroporation is not capable to produce uniform
membrane enables the harvesting of the intracellular comp- electric field influence to all the treated cells. The size and the
onent. For decades, researchers have collected protein and distribution of the pores on the membrane, thus, are not
nucleic acid from the cell by breaking the cell membrane [6]. homogeneous and may not allow the specific substance to be
This process, popularly known as cell lysis, is conducted by delivered [10].
exposing the cell to chemical agents, either enzymes, deter- Aside from chemical agent and electrical field, many
gent, or by applying mechanical disruption. However, dete- other physical methods have been demonstrated to break the
riorating the whole cell membrane will ultimately kill the cell. cell membrane efficiently. [11]. Ultrasound acoustic cavita-
Various technology has been developed to ensure the tion shock wave has been able to accommodate DNA trans-
formation of pores in the cell membrane without compro- fection into mammalian cells [12–14]. Moreover, shear stress
mising the cell viability. Electroporation is one of the most from the oscillating bubble produced by the acoustic wave is
successfully commercialized methods for creating temporary able to compromise the cell membrane integrity. Beside
pores in the cell membrane and for delivering cargo into the acoustic energy, mechanical force to break the cell membrane
cell. This method utilizes the electric field to cause a charge can be introduced by fluid flow. A high-speed jet flow of

0957-4484/19/264002+13$33.00 1 © 2019 IOP Publishing Ltd Printed in the UK


Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

solution with cargo can penetrate the cell seamlessly in vivo


[15]. Likewise, shear stress from micromechanical structure
also allows a high transfection rate [16]. Similarly to the
acoustic shock wave, the laser can generate highly localized
energy which wounds the cell membrane [17, 18]. Although
the throughput is far less than the ultrasonic device, the pre-
cision of the laser allows a specific cell to be targeted with
high efficiency [19, 20].
The emerge of nanotechnology has shed light on new cell
membrane poration technology. With the size of up to a few
hundred nanometers, nanotechnology has the perfect scale to
manipulate the cell and its organelles, the dimensions of
which are in the order of a few microns. By using a sub-
micron structure, the cytosol is accessible for interrogation
with minimal disruption to the cell. In drug delivery studies,
nanotechnology gains huge fame especially due to its role in
enhancing the efficacy and specific targeting of the drug into
the target site [21]. Additionally, the power of nanotechnol-
Figure 1. Device operation in nanostructure mechanical poration. (a)
ogy is able to alleviate specific limitations of former cell Seeding of the cell on the nanostructure. Reprinted by permission
poration technology by providing precision and accuracy, from Macmillan Publishers Ltd: Nature Communications [31],
reducing the adverse effects, and increasing yield and effi- Copyright (2014). (b) Spatially controlling the nanostructure. [32]
ciency of the method. 2015 Scientific Reports with permission of Springer. CC BY 4.0.
In this article, we provide a summary of recent technol-
ogy for cell membrane poration that utilizes nanostructures.
Specifically, we discuss the advanced nanostructures that are
implemented as a platform for cell membrane poration. better protein extraction performance compared to chemical
Therefore, nanoparticles that may be consumed by the cell, cell lysis [24]. Random formation of the sharp tip such as the
such as quantum dots, silica nanoparticle, and metal nano- hierarchical silicon nanospikes and nanowires also allow
particle, are not covered in the discussion. We describe sev- high-throughput protein extraction and at the same time iso-
eral poration mechanisms of the nanostructure and categorize lating the genetic materials of the target cells [25–27].
it based on the transduction principle that determines the Although the main reason for improvement for lysate
poration mechanism. We examine the performance for cargo extraction using sharp nanostructure is not clearly understood,
delivery of the various nanostructure-based cell membrane we assume that the nanostructure acts as a fine grater for the
poration technologies. Finally, we will also provide insight cell. In comparison to chemical lysis employing detergent to
into the new opportunities of nanotechnology for cell mem- compromise the cell membrane, mechanical nanostructure
brane poration and molecular cell biology applications. combined with the driving force from the fluid flow ruptures
the plasma membrane through strong shear stress owing to the
sharp nanoscale tips. In addition, incorporating detergent or
2. Mechanical poration other chemicals for lysis may cause denaturation of the pro-
tein and other biomolecules. The absence of any chemical in
Mechanical poration is the most straightforward mechanism the mechanical lysis procedure preserves the biochemical
for breaking the cell membrane. In general, the mechanical content of the cells. This reason might also explain the higher
poration is analogous to a surgeon’s blade cutting patient extraction yield of the nanostructured devices compared to
tissue. The mechanical forces cause enough stress on the cell chemical lysis.
to generate discontinuity to the plasma membrane. In a For cell lysis, the mode of operation is usually by flowing
mammalian cell, lipid bilayer composing the plasma mem- the cell through the structure. Often this type of mode of
brane is typically around 10 nanometers thick and might vary operation integrates the nanostructure in a microfluidic
depending on the chemical content of the layer [22]. There- channel where the cell is flown. However, for the cell pora-
fore, a mechanical structure with a larger feature than the lipid tion, the mode of operation needs to ensure it is minimally
bilayer thickness is supposedly capable to breaking the invasive to the cell and maintains the cell viability. Figure 1
plasma membrane. represents the mode of operation for the cell poration using
Nanostructures have been proven to enhance cell mem- mechanical nanostructure. Devices harnessing mechanical
brane disruption. For example, the nanoscale barb developed energy for cell poration are usually operated by seeding the
by Di Carlo et al efficiently lyses cells and improves the cell right on the nanostructured materials or by controlling the
accessibilities of the protein content [23]. The barbs have a nanostructure itself. High aspect ratio nanostructures, such as
really sharp tip with a radius of curvature smaller than 25 nm. vertically aligned nanowires, are capable of direct penetration
Similar to the nanoscale barb, arrays of silicon pillar with a to the cell membrane as the cell spreads over the substrate
sharp tip, called nanoblade, can rapidly lyse the cell with [28]. Whereas, a single nanostructure like a nanopipette and a

2
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

nanoneedle, fabricated by modifying AFM tip, can be spa- indentation of more than 1mm is sufficient to puncture the
tially manipulated to puncture the cell membrane [29, 30]. plasma membrane of human epidermal melanocyte [29, 38].
In the first mode of operation of the cell poration device, A smaller diameter nanoneedle with straight side wall
i.e. seeding the cell on the substrate depicted in figure 1(a), requires less force to penetrate the cell membrane in com-
the penetration of nanostructure is mainly driven by the parison to the larger and tapered one [38]. Combining arrays
properties and the types of the cell itself. Puncturing the of diamond nanoneedles with a diameter around 300nm and
plasma membrane of the stiffer cell requires less strain and centrifugation with speed 300–400rpm, Wang et al demon-
tension compared to the softer cell. Experimental observation strated that the system could efficiently poke fibroblast, and
shows that lipid membrane could fail when the tension is even primary neuron cells [39]. The calculated force gener-
1–10 mN m−1 and the rupture strain is 1%–5% [33–35]. ated by their system is approximately 2nN per needle.
Using mechanical model, Xie et al calculated the critical According to Angle et al the penetration forces required to
tension of membrane failure of 5.6 mN m−1 and varying puncture the cell membrane depends on the probe sharpness,
rupture strain: 0.7% for a stiff cell; 2% for a regular cell; and not the properties of the cellular architecture [40]. Moreover,
6% for a soft cell [28]. The critical tension value does not for low force operation, sharp nanostructure design with a tip
assure membrane penetration, it only indicates a threshold with a size of less than 250nm is suggested. A variety of
value above which penetration of the nanowire will start single nanostructure has been conceived with multiple func-
occurring. The mechanisms of how the vertical nanowire tionalities such as femtoliter liquid injection, molecule
breaks the cell membrane are described in two ways: impaling delivery, organelle probing, and electrical signal recording
and adhesion. The impaling mechanism specifies that the [41–44].
penetration occurs when the rounded cell initially contacts the Using only mechanical force for cell poration is expected
nanowire and the membrane is deformed by the gravitational to not induce obnoxious cell responses due to biochemical
force. Therefore, the cell does not adhere well to the substrate. activation which commonly occurs when using chemical
On the other hand, the adhesion-mediated mechanism affirms agents or the viral vector [45, 46]. Also, specific control over
that the cell membrane deformation occurs over a longer number and size of pores is possible. As long as the nanos-
period of time compared to the impaling mechanism. The tructure is able to be fabricated, the desired output of how the
binding of the cell membrane onto the substrate induces a structure breaks the cell membrane can be designed. On the
local force between the nanowire and the membrane. The other hand, the fabrication issue remains the challenge of the
former mechanism is suitable to explain the membrane widespread adoption of the mechanical nanostructure for cell
penetration for cells that are typically grown in suspensions membrane poration. Until now, the nanofabrication process is
such as B cell and T cell of the lymphocyte family. However, usually conducted in a clean room which does not come
the impaling mechanism also suggests that higher aspect ratio cheap. In addition to costly fabrication, the mechanical
and a smaller diameter nanowire is vital to break the cell nanostructure design might need to be customized specifically
membrane compared to adhesion-mediated cell membrane for each cell type, not a kind of one-size-fits-all technology.
disruption. Shalek et al demonstrate experimentally that The effectiveness of the nanostructure for cell poration highly
longer and a sharper nanowire with a height of 2–3 mm and a depends on the suitability of the cell dimension with respect
diameter less than 150 nm is required to ensure effective to the structure geometry. For example, too large deformation
penetration into mouse B and T cells [36]. Unfortunately, this experienced by the cell can lead to harmful mechan-
long and sharp nanowire will reduce the viability of adherent otransduction or even DNA damage due to nuclear defor-
cells which is likely caused by nuclear penetration. From their mation [47–51]. Regardless of its complex design and
result, obviously, the size and geometry of the nanostructure fabrication method, the nanostructure still provides a pro-
also define the poration performance of the device. Extensive mising future for cell poration technology.
study has shown that the nanoneedle with a cylindrical shape
and diameter of less than 400 nm can penetrate non-inva-
sively into human epidermal melanocyte, HEK293 cells, and 3. Electroporation
breast cancer-derived MCF-7 cells [37]. By increasing the
diameter up to 800 nm, the nanoneedle starts to compromise Infusing electric field via the nanostructure contributes several
the viability of all the aforementioned cells. major advantages for cell electroporation. The commonly
Figure 1(b) portrays the second mode of operation which used bulk electroporation requires a high voltage in the order
penetration is caused by controlling the nanostructure. When of several kilovolts and creates random pore formation on the
manipulating the nanostructure, more parameters beside the surface of the cell membrane. The high electric field may
cell properties and nanostructure characteristics need to be lower the cell viability when exposing the electric field to a
considered. In general, the critical driving factors which larger area of the cell, it may cause an ionic imbalance in the
determine the effectiveness of membrane poration using intracellular environment that could lead to cell death [52].
controllable nanostructure is how the nanostructure is being The pH around the cathode of the electroporation system is
moved. The tension generated by the movement of the also prone to change which would be toxic to the cell [53]. In
nanostructure on the cell membrane is determined by the addition, some electro-sensitive particles for intracellular
force imposed to the membrane. A nanoneedle with a dia- delivery such as quantum dots or negatively charged mole-
meter of 200–300nm, a force at around 1–2nN and cules could aggregate and dysfunction when transferred using

3
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

Figure 2. Design of nanoelectroporation device. (a) Single nanochannel electroporation device. Reprinted by permission from Springer
Nature Customer Service Centre GmbH: Nature, Nature Nanotechnology [65], Copyright 2011. (b) Arrays of nanochannel with a bulk
electrode (left) and nanochannel electrodes (right). Reprinted with permission from [66], Copyright (2013) American Chemical Society. [67]
2017 Scientific Reports with permission of Springer. CC BY 4.0.

bulk electroporation [54, 55]. In contrast, the nanostructure duration of the electric pulse. The ability to precisely control
mediates a localized electric field which causes gentler dis- the amount of molecules delivered into the cell is clearly
ruption yet effective [56]. When coupled with the nanos- unachievable using bulk electroporation.
tructure, such as carbon nanotubes or nanoelectrode gap, the Nanostructure arrays for electroporation can be con-
required electrical input could decrease significantly due to stituted by bulk electrode or nanoelectrodes. Figure 2(b)
the electrical field enhancement on the tip of the nanostructure shows these different setups. The first setup operates by
[57–60]. The low applied voltage in the nanostructure-medi- exploiting the nanostructure to help enhance the electric field
ated electroporation prevents bubble formation which indi- induced by the bulk electrode that is positioned across the
cates minimal electrochemical reaction that may compromise nanochannel [65, 66, 68]. Clearly, the electroporation setup is
cell viability [61]. Additionally, a high-throughput cell pro- similar to the bulk electroporation. Nevertheless, the presence
cessing device using electroporation mechanism can be rea- of the nanochannel could improve the field localization when
lized with low energy consumption [62]. the electrical field is applied across the cell. The molecular
Harnessing the nanostructure for electroporation can delivery into the cell is enhanced by electrophoresis thus,
uncover a novel mechanism for the cell permeabilization and controlling the presence of electrical field could modulate the
cargo delivery transport. The molecular transport process in amount of the delivered molecule. The second setup utilizes
bulk electroporation, which permeabilizes the cell membrane the nanostructure itself as the nanoelectrode [67, 69, 70].
randomly on both poles facing cathode and anode, resembles Using the later design, selective poration of a specific cell in
the diffusion phenomenon through the long-live pores the same population is possible by only firing certain
[9, 63, 64]. The diffusion process exhibits a gradual increase nanoelectrodes. Beside temporal selectivity, the nanoelec-
of the concentration of the molecule inside the cell. On the trode could also act as a sensing instrument for intracellular
other hand, the nanochannel electroporation device as in signal recording. The nanometer-sized pores generated by the
figure 2(a) delivers the molecule instantaneously into the nanoelectrode increase the signal quality of the recorded cell
cytosolic space within 30 ms [65]. For comparison, bulk action potential which enables real-time detection of electrical
electroporation takes 150 s to achieve the same delivery signal alteration due to drug intake [71].
amount which is three orders of magnitude slower than the The nanostructure-enabled electroporation provides
nanochannel electroporation. The rapid molecule delivery is localized permeabilization of the cell membrane with high
theoretically described as a result of field enhanced particle intracellular delivery efficacy and minimal disruption even
acceleration inside the channel. The nanochannel electro- though the throughput is still limited. In order to ensure the
poration also allows precise dosage control by varying the localized electroporation to take effect, the cell should be in

4
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

close contact and tightly sealed to the nanochannel or the is able to slice the membrane in a ‘cat-door’ shape [82]. This
nanoelectrode. Current nanostructure-mediated electropora- unique opening shape allows large cargo to be passed through
tion technology involves adhesion of the cell to the nanos- without causing too much loss of the cell membrane and
tructure in order to fulfill this requirement. Consequently, the cytosol content thus preserving the cell’s health.
system is not suitable for batch processing. For single cell Arrays of plasmonic nanostructures, moreover, could
application, close contact between the cell and the structure generate pores on a large cell population on the substrate.
could be achieved by trapping the cell using optical or Figure 3 presents various substrates that have been realized
acoustic tweezer and locating the cell close to the nanos- including an array of gold nanoparticles, tipless metallic
tructure [72, 73]. nanostructures, and sharp-tip metallic nanostructures [80,
Another challenge of this technology is the complicated 86–88]. In addition, by resembling the nanochannel electro-
fabrication. The nanochannel electroporation device by Lee poration device, the plasmonic nanotube substrate supports
et al exploit the DNA combing and imprinting method which manipulation of the type of the chemicals that need to be
is a recently developed method developed by themselves [74]. delivered after poration [89]. Spatial control of the poration is
The nanostraw electroporation device by Melosh et al com- achievable by controlling the position of the laser beam and
bine atomic layer deposition method on membrane template the amount of the laser fluence. The sharp tip plasmonic
followed by reactive ion etching to expose the hollow nano- substrates by Wu et al (figure 3(a)) and Saklayen et al
wire [75]. The device development requires advance knowl- (figure 3(c)) require around 55 mJ cm−2, even though the type
edge in nanofabrication especially for incorporating new of materials they employ is different; the former adopts a thin
materials in complex geometry [70]. Until now, there are only film of titanium on silicon while the later utilizes gold pyr-
a handful of research groups that are working on nanoelec- amidal nanostructure [86, 88]. On the other hand, tipless
trode devices for cell study due to a limited expertise in the plasmonic nanostructures such as gold nanoparticle layers in
topic. figure 3(b) require 150 J cm−2 for effective cell poration
which is several orders of magnitude larger than its sharp tip
counterpart [87]. As a side note, high laser energy may be
4. Optical and plasmonic poration
phototoxic and harmful for the cell viability. Nonetheless, the
plasmon-based cell poration device is minimally invasive to
Conductive materials illuminated by electromagnetic waves
the cell considering the rapid membrane disruption involved
that match its resonance frequency could generate a plas-
during the poration process. In spite of that, short wavelength
monic effect which in turn produces enormous energy in the
spectrum such as UV has been widely known as a major
form of heat [76]. Metallic nanostructure immersed in water,
cause of DNA damage in the cell. Therefore, a safer device
for example, when irradiated with a laser, produces an
could be developed by using an infrared laser to avoid any
explosive bubble due to superheated water [77]. The energy
damage to the living cell. Reducing the laser energy via short
that either comes from the thermal heating or the bubble
pulse laser instead of continuous illumination may inflict less
explosion has been used to enhance membrane permeabili-
zation of the cell [78]. The heat generation in the plasmonic cell damage even though it could also compromise poration
nanostructure highly depends on the shape and geometry of effectivity.
the nanostructure [79]. The local near-field enhancement of The utilization of laser setup might hurdle the widespread
the plasmonic effect could be increased by tuning the implementation of the nanoplasmonic device for cell poration.
morphology of the nanostructure [80]. The integration Optical setup with a coherent laser source is complicated and
between optical laser system and metallic nanostructure is rather unaffordable in general. The laser needs to scan the
able to promote highly effective cell membrane poration [81]. plasmonic substrate which usually occurs slowly depending
Similar to the mechanical nanostructure, the plasmonic on the intended laser fluence. Wu et al reported that this
nanostructure that is utilized for cell membrane poration can limited the delivery efficiency of their device [86]. The tem-
be in a single structure as well as an arrayed fashion. Chiou poral delay of the plasmon activation will allow the cell to
group develop a single nanostructure called photothermal heal before the cargo can be delivered. For efficient energy
nanoblade which can cut the cell membrane and load various transfer, the laser excitation wavelength has to be well
cargo into a single mammalian cell, ranging from mitochon- adjusted according to the nanostructure morphology [79].
dria to bacteria [82–85]. The nanostructure is fabricated by Therefore, a methodical design of the size, shape, and geo-
depositing a thin layer of noble metal onto the surface of the metry of the nanostructure has to be done prior to fabrication
micropipette. Illuminating the metal with a continuous-wave because out of resonance excitation would not produce
laser would produce the vapor bubble that bolsters the plasmon effect. Despite these challenges, the nanoplasmonic
breaking of the cell membrane. The process of vapor bubble poration device can be integrated with other application such
generation to cell membrane disruption occurs in less than as the biosensor. Surface plasmon resonance has been inten-
200 ns. This type of plasmonic nanostructure is able to create sively developed for sensing and quantization of assorted
a large window in the cell membrane and at the same time biomolecules [90]. In addition, the plasmon excitation might
maintains cell viability by conserving the cell structure and also be generated using electrically pumped nanolaser that
allowing rapid reseal of the cell membrane [86]. The nanos- can be monolithically combined with the nanostructure sub-
tructure is intentionally fabricated in a crescent shape so that it strate [91–93]. These technologies will definitely improve

5
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

functions as a trap for unwanted substances. Particles that are


unable to escape will follow a degradation procedure by the
lysosome. From this perspective, the ability to flee the vesi-
cular trap is the rate-determining step for particle uptake
during endocytosis. In contrast, direct cytosol access allows
cargo from the extracellular environment to bypass the
endosomal pathway. The creation of pores by the nanos-
tructure could facilitate particle transport through diffusion or
other physicochemical approaches which is not particularly
dependent on how the cell responds to the particle. Therefore,
the cell poration allows delivery of cargo beyond genetic
materials such as protein, artificial nanoparticles, organelles,
or even bacteria. Delivering those substances into the cell may
allow stem cell reprogramming, organelle labeling, mea-
surement of the intracellular environment, and tracking the
intracellular dynamics [95–98].
The delivery efficiency using the nanostructure is dif-
ferent for each technology based on the mode of operation,
type of cell, the geometry of the structure, and the type of
cargo. The majority of the single nanostructure can success-
fully deliver cargo due to precise control of the structure into
the cell, or by moving the cell to the nanostructure. Earlier
versions of this technology employed micromanipulator to
steer the nanostructure [30, 41, 82, 83, 85, 99]. Later on, the
nanostructure is often fabricated along the AFM probe,
therefore, the positioning of the structure can achieve
nanoscale resolution using the piezoelectric actuator
[29, 37, 43, 56]. For the same purpose, manipulating the cell
position using optical tweezers has also been demonstrated
[65]. Even though single nanostructure manipulation can
deliver cargo effectively, this process limits the study into a
single cell. Scaling up the cargo delivery process of the single
nanostructure may be assisted by automating the nanos-
tructure manipulation using three-dimensional stage con-
troller. The automatic stage apparatus, called CellBee, is able
to handle up to 10 cells min−1 [100].
A more favorable approach to increase the cargo delivery
Figure 3. Plasmonic nanostructure device for membrane poration. (a) throughput is by applying arrays of the nanostructure and
Crescent-shaped metallic nanostructure. Reprinted by permission interfacing them with a population of cells. A summary of the
from Springer Nature Customer Service Centre GmbH: Nature,
Nature Methods [86] Copyright 2015. (b) Gold nanoparticle layer. delivery performance of nanostructure arrays is listed in
[87] John Wiley & Sons. © 2016 WILEY‐VCH Verlag GmbH & table 1. In a special case, the nanostructure arrays may per-
Co. KGaA, Weinheim. (c) Pyramidal sharp-tip gold plasmonic form intracellular delivery in the tissue-level domain such as
substrate. Reprinted with permission from [88]. Copyright (2017) plasmid DNA delivery to promote neovascularization of
American Chemical Society. Scale bar, 10 μm muscle tissue [101, 102]. Various types of nanostructures
have demonstrated successful cargo delivery into living cells
mainstream adoption of the nanoplasmonic method for cell and each of them has their own superiority over the others.
membrane poration in the future. Nanostructure mechanical poration requires no other sup-
porting instrument such as the signal generator or laser setup.
However, mechanical poration is usually less effective in
5. Discussion delivering nucleic acid compared to electrical-based poration
technology due to the lack of electrical driving force of the
The cell poration could provide direct access into the intra- charged molecule. For electrical poration, the dosage of the
cellular compartment of the cell. Naturally, cellular uptake delivered particle may be controlled through the amount of
proceeds through the endocytic pathway which is mediated electric pulse. Yet, close contact of the cell and the nanos-
by copious proteins and biomolecules [94]. The chemical tructure is required in order for the process to take effect. This
substances captured with the aid of endocytosis is then con- is a huge challenge for the suspension-grown cell. Plasmonic-
signed into the vesicle. The majority of foreign particles are based nanostructure poration allows larger pores to be created
perceived as a threat to the cell, therefore, the vesicle without adverse effect to the cell’s health. But the number of

6
Nanotechnology 30 (2019) 264002
Table 1. Nanostructure arrays for intracellular delivery.

Delivery performance:
Delivery efficiency
(%D)
Transduction
No. Type of nanostructure arrays principle Type of cell Type of cargo Cell viability (%V) Remarks
1 Vertically aligned silicon nanowire Mechanical Dendritic cells DNA %D<95% Effective for various immune cells
[36, 103]
Diameter<150 nm B cells RNA %V<95%
Height=1–3 μm T cells Protein
Macrophages
NK Cells
NIH3T3
Hippocampal
neuron

2 Diamond nanoneedle array Mechanical NIH3T3 EthD-1 %D: EthD-1<80% Implemented using common centrifugation system
[39, 104]
Diameter=300–500 nm A549 Dextran %D: 3k Dextran<60%
Height=4.5–7.5 μm Hippocampal QD %D: 20 nm QD<60%
neuron
Antibody %D: Antibody<35.5%
7

DNA %D: DNA<45%


%V<92%

3 Microfabricated silicon nanoneedle Mechanical HeLa Dextran %D: 70k Higher delivery performance by oscillating the nanostructure
arrays with tapered wall [105] or Dextran<40%
straight wall [32, 106]
Diameter=23–200 nm NIH3T3 Protein %D: Cre
enzyme<40%
Height=12–25 μm DU145 DNA %D: DNA<34%
%V<90%

4 Vertically aligned carbon nanofiber Mechanical CHO-K1 shRNA %D: shRNA<89% Co-delivery of multiple genes
arrays for RNAi [107, 108]
Diameter=100 nm DNA %D: 2 DNAs<76.7%
Height=10–17 μm

5 Carbon nanosyringe array [109] Mechanical NIH3T3 DNA %D<34% Hollow tubes of the nanosyringe allows cargo loading prior

A K Fajrial and X Ding


to cell poration
Diameter=50 nm %V<85%
Height=40–160 nm

6 Silicon hollow nanoneedle Mechanical NIH3T3 Dextran %D<70% Requiring saponin for delivery
array [110]
Nanotechnology 30 (2019) 264002
Table 1. (Continued.)

Delivery performance:
Delivery efficiency
(%D)
Transduction
No. Type of nanostructure arrays principle Type of cell Type of cargo Cell viability (%V) Remarks
Diameter=250–500 nm HEK293 DNA
Height=5 μm

7 Nanostraw [75] Mechanical CHO Ion %D: Co2+<70% Highly dense structures at around 107 straws/cm2
Diameter=100 nm HeLa Dye %D: Alexa
Fluor<40%
Height=1 μm DNA %D: DNA<10%

8 ZnO Nanowire in micro- Mechanical MCF-7 Molecular %D<59% Pressure driven microchannel utilizing PDMS membrane to
channel [111] beacon move cell to the nanowire
Diameter=37 nm %V<83%
Height=530 nm

9 Silicon nanochannel electroporation Electrical H9C2 Nucleotide %D: ODN<73% Precise control over delivered cargo at single cell level
[68, 112]
Pore size=650 nm NK-92 DNA %D: DNA<74%
8

Mouse embryo- %D: OSKM<10%


nic fibroblast
%V<90%

10 Aluminum nanospike electropora- Electrical HeLa Propidium %D<93% Low voltage operation without any bubble generation
tion [61] iodide (PI)
%V<93%

11 Nanostraw electroporation [66] Electrical CHO PI %D: PI<95% Dosage control and co-delivery of multiple genes
Diameter=250 nm HEK293 DNA %D: DNA<81%
Height=1.5 μm %D: 2 DNAs<74%
%V<95%

12 Hollow nanoelectrodes [67] Electrical NIH3T3 PI %D<80% Low voltage with spatial control
Diameter=400 nm (outer), %V<98%
250 nm (inner)
Height=1.8 μm

A K Fajrial and X Ding


13 Gold nanoparticle layers [87] Plasmonic HeLa Dextran %D: 4.4k 4-fold transfection efficiency compared to lipofection
Dextran<54%
Roughness=500 nm HUVEC DNA %D: DNA<53%
Mouse embryo- %V<95%
nic fibroblast
Nanotechnology 30 (2019) 264002
Table 1. (Continued.)
Delivery performance:
Delivery efficiency
(%D)
Transduction
No. Type of nanostructure arrays principle Type of cell Type of cargo Cell viability (%V) Remarks
14 Plasmonic pyramid array sub- Plasmonic HeLa Calcein %D: Calcein<95% Handle up to 50 000 cells min−1
strate [81, 88]
Base length=2.4 μm Dextran %D: 10k
Dextran<79%
Height=1.4 μm %D: 70k
Dextran<70%
%D: 150k
Dextran<68%
%D: 500k
Dextran<24%
%D: 2 m
Dextran<16%
%V<98%
9

15 Biopohotonic laser-assisted surgery Plasmonic HeLa Calcein %D: 20 nm Pressure driven cargo delivery system
tool [86] Dextran beads<93%
3 μm diameter of hole arrays with NHDF Protein %D: 200 nm
crescent-shaped Ti nanostructure beads<87%
PB-MDM Polystyrene %D: 500 nm
beads beads<79%
RPTEC Bacteria %D: 1 μm
beads<75%
%D: 2 μm
beads<62%
%D: Bacteria<57.9%
%V<90%
16 Hollow plasmonic gold nano- Plasmonic NIH3T3 PI — Spatial and temporal control over porated cells
tube [89]
Diameter=180 nm (outer), 90 nm
(inner)

A K Fajrial and X Ding


Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

treated cells is limited to the laser scanning speed due to the observation instead of a single cell. Aside from cargo deliv-
typical laser beam spot only allowing a small area to be ery, the nanostructured device also plays a significant role in
exposed. interfacing the biosystem with engineering toolsets
Upon cell poration by the nanostructure, treated cells can [116, 117]. The tiny feature of the nanostructure is non-
be harvested by following common cell culture procedures. invasive for long-term intracellular and extracellular signal
Cells grown in suspension can be removed directly from the probe [118, 119]. The details on the nanostructure for cellular
nanostructured device and restored back in the incubator for electrical recording have been covered elsewhere [120–124].
further use. Treated adherent cells are usually incubated on The nanostructure devices could also assist in developmental
the device for 24 h [103, 113]. After incubation, the cells are biology studies. For example, engineering the nanostructure
detached by trypsin and washed. Subsequently, harvested materials and dimension has been demonstrated to stimulate
cells can be examined by various biological assays. neural cultures, improve cartilage cell proliferation, and
In general, cell membrane poration using the nanos- influence stem cell fate [125–129]. Nevertheless, there is still
tructures can preserve the cell viability of the cell in the range room for improvement of creative nanotechnology for cell
of 75% to 98%. However, delivery efficiency is highly membrane poration and cellular studies.
diverse (see table 1). Small molecules, such as propidium It is important that future research investigates how living
iodide and calcein, have the highest delivery efficiency. This cells interact with nanostructures as limited study has been
condition happens due to two main reasons. First, small done in this area [116, 130, 131]. A serendipitous response of
molecules with small radii will diffuse faster due to the the cell when interacting with the structure may or may not be
transport kinetics. Therefore, small molecules will accumulate desired for the overall performance for cell poration. Cultur-
quickly in the cell resulting in more delivery efficiency. ing the cell in the nanostructured environment can induce
Incorporating an active actuator, such differential pressure various cell response such as a change in proliferation and
chambers may assist the delivery of larger molecules [86]. differentiation [132, 133]. Therefore, understanding cell-
Although less size-dependent diffusion takes the role in the nanostructure interfaces may provide guidance on how to
process, the delivery efficiency for large cargo is still unable optimize current technology to increase delivery efficiency
to match small molecule efficiency. The second reason is the without compromising cell health.
varying degree of membrane repair time depending on the Another challenge is the limitation in nanofabrication
wound size. Larger pores tends to be repaired faster due to which relies mostly on lithographic method. Until now,
Ca2+ signaling while small pore repair takes a longer time and nanofabrication of a specific design is popularly conducted
is thus stay stable for a longer period of time [9]. In the via lithography, either bottom-up or top-down approach.
presence of Ca2+ ion at physiological concentration, the Even though an innovative nanofabrication method has been
plasma membrane usually reseals as quickly as 30 s [114]. investigated, conventional techniques such as photo-
Delivery of small molecules may reach up to 95% efficiency lithography and e-beam lithography are the ones that are
using a variety of nanostructure devices. For large cargo such widely available in academic and industrial settings
as bacteria, the delivery efficiency that has been achieved is [134–136]. Novel research demonstrates that non-lithography
50% by plasmonic nanostructure device which offers rapid techniques are able to fabricate a well-controlled nanos-
membrane poration [86]. An important thing to note is that tructure such as the vertically aligned nanowire [137, 138].
transfection of the cell using naked DNA plasmid is less Recently, plasmonic nanostructure for cell poration can also
likely to succeed. Oftentimes the nucleic acid is coupled with be fabricated using self-assembly of colloidal nano-
liposome to facilitate the transport to the nucleus [115]. particle [139].
Hence, cargo pre-treatment may be needed to achieve high For ex vivo application like immunotherapy, a high-
delivery efficiency. throughput intracellular delivery is of utmost importance.
Current nanostructure devices on average can achieve around
∼105 cells each processing time. In the clinical trial, a pro-
6. Summary and outlook cessing of 108–109 cells is required depending on the dosage
[140]. To achieve this outcome, the viral vector is still a
In this paper, we have reviewed recent nanostructured devices preferred choice for the adoptive immune therapy as a huge
for cell membrane poration. The nanostructures discussed number of engineered cells can be manufactured [141].
here utilize the physical approach to generate temporal dis- However, engineering cells through viral vector tend to be
ruption in the cell membrane. The proof-of-principle works labor intensive and handling viral agent requires some
described here demonstrate that nanostructure allows high degrees of expertise. Viral vector transfection is also limited
delivery efficiency of various cargo into the cell without by the number and size of the gene to package [142].
excessive reduction to cell viability. Nanostructure enables Improving current nanostructured devices may bridge this gap
precision control over how much cargo is delivered, which by offering high-throughput cell processing. Novel nanos-
cells are targeted, and when the intracellular delivery is tructure design or hybrid technology by combining multiple
intended, unlike bulk cell membrane poration method. The physical approaches may be able to unravel improved per-
power of nanostructured device for intracellular delivery formance of the technology. The possibility offered by
makes it possible to uncover novel biological mechanism nanotechnology warrants more exploration for future research
such as the pathogen-host interaction in population-level in cell membrane poration application.

10
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

Acknowledgments [29] Obataya I, Nakamura C, Han, Nakamura N and Miyake J


2005 Nano Lett. 5 27–30
The authors acknowledge support through the startup funds [30] Schrlau M G, Falls E M, Ziober B L and Bau H H 2008
Nanotechnology 19 015101
from the University of Colorado Boulder. [31] Xu A M, Aalipour A, Leal-Ortiz S, Mekhdjian A H, Xie X,
Dunn A R, Garner C C and Melosh N A 2014 Nat.
Commun. 5 3613
[32] Matsumoto D, Sathuluri R R, Kato Y, Silberberg Y R,
ORCID iDs Kawamura R, Iwata F, Kobayashi T and Nakamura C 2015
Sci. Rep. 5 15325
Apresio K Fajrial https://orcid.org/0000-0001-8946-6648 [33] Evans E, Heinrich V, Ludwig F and Rawicz W 2003 Biophys.
Xiaoyun Ding https://orcid.org/0000-0003-4252-9335 J. 85 2342–50
[34] Sen S, Subramanian S and Discher D E 2005 Biophys. J. 89
3203–13
[35] Verma P, Wong I Y and Melosh N A 2010 Biointerphases 5
37–44
References [36] Shalek A K et al 2012 Nano Lett. 12 6498–504
[37] Han S, Nakamura C, Obataya I, Nakamura N and Miyake J
[1] Hawiger J 1999 Curr. Opin. Chem. Biol. 3 89–94 2005 Biochem. Biophys. Res. Commun. 332 633–9
[2] Rosi N L, Giljohann D A, Thaxton C S, Lytton-Jean A K R, [38] Obataya I, Nakamura C, Han S, Nakamura N and Miyake J
Han M S and Mirkin C A 2006 Science 312 1027–30 2005 Biosens. Bioelectron. 20 1652–5
[3] Maia J, Santos T, Aday S, Agasse F, Cortes L, Malva J O, [39] Wang Y et al 2014 Nat. Commun. 5 4466
Bernardino L and Ferreira L 2011 ACS Nano 5 97–106 [40] Angle M R, Wang A, Thomas A, Schaefer A T and
[4] Suh J S, Lee J Y, Choi Y S, Chong P C and Park Y J 2013 Melosh N A 2014 Biophys. J. 107 2091–100
Biomaterials 34 4347–59 [41] Knoblauch M, Hibberd J M, Gray J C and van Bel A J E 1999
[5] Chen A A, Derfus A M, Khetani S R and Bhatia S N 2005 Nat. Biotechnol. 17 906–9
Nucleic Acids Res. 33 e190–190 [42] Kouklin N A, Kim W E, Lazareck A D and Xu J M 2005
[6] Tan S C and Yiap B C 2009 J. Biomed. Biotechnol. 2009 Appl. Phys. Lett. 87 173901
574398 [43] Chen X, Kis A, Zettl A and Bertozzi C R 2007 PNAS 104
[7] Tieleman D P 2004 BMC Biochemistry 5 10 8218–22
[8] Neumann E, Schaefer-Ridder M, Wang Y and [44] Singhal R, Orynbayeva Z, Sundaram R V K, Niu J J,
Hofschneider P H 1982 EMBO J. 1 841–5 Bhattacharyya S, Vitol E A, Schrlau M G, Papazoglou E S,
[9] Weaver J C and Chizmadzhev Y A 1996 Bioelectrochem. Friedman G and Gogotsi Y 2011 Nat. Nanotechnol. 6 57–64
Bioenerg. 41 135–60 [45] Thomas C E, Ehrhardt A and Kay M A 2003 Nat. Rev. Genet.
[10] Esser A T, Smith K C, Gowrishankar T R, Vasilkoski Z and 4 346–58
Weaver J C 2010 Biophys J 98 2506–14 [46] Jin L, Zeng X, Liu M, Deng Y and He N 2014 Theranostics 4
[11] Meacham J M, Durvasula K, Degertekin F L and 240–55
Fedorov A G 2014 J Lab Autom. 19 1–18 [47] Charras G T and Horton M A 2002 Biophys. J. 82 2970–81
[12] Kim H J, Greenleaf J F, Kinnick R R, Bronk J T and [48] Denais C M, Gilbert R M, Isermann P, McGregor A L,
Bolander M E 1996 Hum. Gene Ther. 7 1339–46 te Lindert M, Weigelin B, Davidson P M, Friedl P,
[13] Bao S, Thrall B D and Miller D L 1997 Ultrasound Med. Wolf K and Lammerding J 2016 Science 352 353–8
Biol. 23 953–9 [49] Irianto J et al 2017 Curr. Biol. 27 210–23
[14] Greenleaf W J, Bolander M E, Sarkar G, Goldring M B and [50] Irianto J, Xia Y, Pfeifer C R, Greenberg R A and Discher D E
Greenleaf J F 1998 Ultrasound Med. Biol. 24 587–95 2017 Biophys. J. 112 446–9
[15] Furth P A, Shamay A, Wall R J and Hennighausen L 1992 [51] Bennett R R, Pfeifer C R, Irianto J, Xia Y, Discher D E and
Anal. Biochem. 205 365–8 Lie A J 2017 Biophys. J. 112 2271–9
[16] Sharei A et al 2013 PNAS 110 2082–7 [52] Gehl J Acta Physiologica Scandinavica 177 437–47
[17] Palumbo G, Caruso M, Crescenzi E, Tecce M F, [53] Li Y, Wu M, Zhao D, Wei Z, Zhong W, Wang X,
Roberti G and Colasanti A 1996 J. Photochem. Photobiol. Liang Z and Li Z 2015 Sci. Rep. 5 17817
B: Biology 36 41–6 [54] Golzio M, Teissié J and Rols M-P 2002 PNAS 99 1292–7
[18] Schneckenburger H, Hendinger A, Sailer R, Strauss W S L, [55] Derfus A M, Chan W C W and Bhatia S N Adv. Mater. 16
Lyttek M and Schmitt M 2002 J. Biomed. Opt. 7 410–7 961–6
[19] Uchugonova A, König K, Bueckle R, Isemann A and [56] Kang W, Yavari F, Minary-Jolandan M, Giraldo-Vela J P,
Tempea G 2008 Opt. Express 16 9357–64 Safi A, McNaughton R L, Parpoil V and Espinosa H D 2013
[20] Baumgart J, Humbert L, Boulais É, Lachaine R, Nano Lett. 13 2448–57
Lebrun J-J and Meunier M 2012 Biomaterials 33 2345–50 [57] Shahini M and Yeow J T W 2011 Nanotechnology 22 325705
[21] Farokhzad O C and Langer R 2009 ACS Nano 3 16–20 [58] Shahini M and Yeow J T W 2013 Lab on a Chip 13 2585–90
[22] Lewis B A and Engelman D M 1983 J. Mol. Biol. 166 211–7 [59] Santra T S, Wang P-C, Chang H-Y and Tseng F-G 2013 Appl.
[23] Carlo D D, Jeong K-H and Lee L P 2003 Lab Chip 3 287–91 Phys. Lett. 103 233701
[24] Yun S-S, Yoon S Y, Song M-K, Im S-H, Kim S, Lee J-H and [60] Jokilaakso N et al 2013 Lab on a Chip 13 336–9
Yang S 2010 Lab Chip 10 1442–6 [61] Riaz K, Leung S-F, Fan Z and Lee Y-K 2017 Sens. Actuators
[25] Kim J, Hong J W, Kim D P, Shin J H and Park I 2012 Lab A: Physical 255 10–20
Chip 12 2914–21 [62] Liu C, Xie X, Zhao W, Liu N, Maraccini P A, Sassoubre L M,
[26] So H, Lee K, Seo Y H, Murthy N and Pisano A P 2014 ACS Boehm A B and Cui Y 2013 Nano Lett. 13 4288–93
Appl. Mater. Interfaces 6 6993–7 [63] Pavlin M, Leben V and Miklavčič D 2007 Biochimica et
[27] So H, Lee K, Murthy N and Pisano A P 2014 ACS Appl. Biophysica Acta (BBA)—General Subjects 1770 12–23
Mater. Interfaces 6 20693–9 [64] Pavlin M and Miklavčič D 2008 Bioelectrochemistry 74
[28] Xie X, Xu A M, Angle M R, Tayebi N, Verma P and 38–46
Melosh N A 2013 Nano Lett. 13 6002–8 [65] Boukany P E et al 2011 Nat. Nanotechnol. 6 747–54

11
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

[66] Xie X, Xu A M, Leal-Ortiz S, Cao Y, Garner C C and [95] Shah D A, Kwon S-J, Bale S S, Banerjee A, Dordick J S and
Melosh N A 2013 ACS Nano 7 4351–8 Kane R S 2011 Biomaterials 32 3210–9
[67] Caprettini V, Cerea A, Melle G, Lovato L, Capozza R, [96] Kucsko G, Maurer P C, Yao N Y, Kubo M, Noh H J, Lo P K,
Huang J-A, Tantussi F, Dipalo M and Angelis F D 2017 Sci. Park H and Lukin M D 2013 Nature 500 54–8
Rep. 7 8524 [97] Heller D A, Baik S, Eurell T E and Strano M S 2005 Adv.
[68] Chang L et al 2016 Nanoscale 8 243–52 Mater. 17 2793–9
[69] Xie C, Lin Z, Hanson L, Cui Y and Cui B 2012 Nat. [98] Fakhri N, Wessel A D, Willms C, Pasquali M,
Nanotechnol. 7 185–90 Klopfenstein D R, MacKintosh F C and Schmidt C F 2014
[70] Lin Z C, Xie C, Osakada Y, Cui Y and Cui B 2014 Nat. Science 344 1031–5
Commun. 5 3206 [99] Yum K, Na S, Xiang Y, Wang N and Yu M-F 2009 Nano
[71] Duan X, Gao R, Xie P, Cohen-Karni T, Qing Q, Choe H S, Lett. 9 2193–8
Tian B, Jiang X and Lieber C M 2012 Nat. Nanotechnol. 7 [100] Hara C, Tateyama K, Akamatsu N, Imabayashi H, Karaki K,
174–9 Nomura N, Okano H and Miyawaki A 2006 Brain Cell Bio.
[72] Ashkin A, Dziedzic J M, Bjorkholm J E and Chu S 1986 Opt. 35 229–37
Lett. 11 288–90 [101] Chiappini C, Rosa E D, Martinez J O, Liu X, Steele J,
[73] Ding X, Lin S-C S, Kiraly B, Yue H, Li S, Chiang I-K, Shi J, Stevens M M and Tasciotti E 2015 Nat. Mater. 14 532–9
Benkovic S J and Huang T J 2012 PNAS 109 11105–9 [102] Chiappini C, Martinez J O, De Rosa E, Almeida C S,
[74] Guan J, Boukany P E, Hemminger O, Chiou N-R, Zha W, Tasciotti E and Stevens M M 2015 ACS Nano 9 5500–9
Cavanaugh M and Lee L J 2010 Adv. Matter. 22 3997–4001 [103] Shalek A K et al 2010 PNAS 107 1870–5
[75] VanDersarl J J, Xu A M and Melosh N A 2012 Nano Lett. 12 [104] Chen X, Zhu G, Yang Y, Wang B, Yan L, Zhang K Y,
3881–6 Lo K K-W and Zhang W 2014 Adv. Healthcare Mater. 2
[76] Govorov A O, Zhang W, Skeini T, Richardson H, Lee J and 1103–7
Kotov N A 2006 Nanoscale Res. Lett. 1 84 [105] Park S, Choi S-O, Paik S, Choi S, Allen M and Prausnitz M
[77] Wang Y, Zaytsev M E, Lajoinie G, The H L, Eijkel J C T, 2016 Biomed. Microdevices 18 10
van den Berg A, Versluis M, Weckhuysen B M, Zhang X, [106] Matsumoto D, Yamagishi A, Saito M, Sathuluri R R,
Zandvliet H J W and Lohse D 2018 PNAS 115 7676–81 Silberberg Y R, Iwata F, Kobayashi T and Nakamura C
[78] Xiong R, Raemdonck K, Peynshaert K, Lentacker I, 2016 J. Biosci. Bioeng. 122 748–52
De Cock I, Demeester J, De Smedt S C, Skirtach A G and [107] Mann D G J, McKnight T E, McPherson J T, Hoyt P R,
Braeckmans K 2014 ACS Nano 8 6288–96 Melechko A V, Simpson M L and Sayler G S 2008 ACS
[79] Baffou G, Quidant R and Girard C 2009 Appl. Phys. Lett. 94 Nano 2 69–76
153109 [108] McKnight T E, Melechko A V, Griffin G D, Guillorn M A,
[80] Courvoisier S, Saklayen N, Huber M, Chen J, Diebold E D, Merkulov V I, Serna F, Hensley D K, Doktycz M J,
Bonacina L, Wolf J-P and Mazur E 2015 Nano Lett. 15 Lowndes D H and Simpson M L 2003 Nanotechnology
4461–6 14 551
[81] Saklayen N, Kalies S, Madrid M, Nuzzo V, Huber M, [109] Park S, Kim Y-S, Kim W B and Jon S 2009 Nano Lett. 9
Shen W, Sinanan-Singh J, Heinemann D, 1325–9
Heisterkamp A and Mazur E 2017 Biomed. Opt. Express 8 [110] Peer E, Artzy-Schnirman A, Gepstein L and Sivan U 2012
4756–71 ACS Nano 6 4940–6
[82] Wu T-H, Teslaa T, Teitell M A and Chiou P-Y 2010 Opt. [111] Kim K H, Kim J, Choi J S, Bae S, Kwon D, Park I,
Express 18 23153–60 Kim D H and Seo T S 2015 Small 11 6215–24
[83] Wu T-H, Teslaa T, Kalim S, French C T, Moghadam S, [112] Chang L et al 2015 Lab on a Chip 15 3147–53
Wall R, Miller J F, Witte O N, Teitell M A and Chiou P-Y [113] Cao Y, Chen H, Qiu R, Hanna M, Ma E, Hjort M, Zhang A,
2011 Anal. Chem. 83 1321–7 Lewis R S, Wu J C and Melosh N A 2018 Sci. Adv. 4
[84] Xu J, Teslaa T, Wu T-H, Chiou P-Y, Teitell M A and Weiss S eaat8131
2012 Nano Lett. 12 5669–72 [114] Sharei A et al 2014 Integr. Biol. 6 470–5
[85] Wu T-H et al 2016 Cell Metab. 23 921–9 [115] Felgner P L, Gadek T R, Holm M, Roman R, Chan H W,
[86] Wu Y-C, Wu T-H, Clemens D L, Lee B-Y, Wen X, Wenz M, Northrop J P, Ringold G M and Danielsen M 1987
Horwitz M A, Teitell M A and Chiou P-Y 2015 Nat. PNAS 84 7413–7
Methods 12 439–44 [116] Tian B et al 2018 Phys. Biol. 15 031002
[87] Lyu Z, Zhou F, Liu Q, Xue H, Yu Q and Chen H Adv. Funct. [117] Jiang Y et al 2018 Nat. Biomed. Eng. 2 508
Mater. 26 5787–95 [118] Robinson J T, Jorgolli M, Shalek A K, Yoon M-H,
[88] Saklayen N, Huber M, Madrid M, Nuzzo V, Vulis D I, Gertner R S and Park H 2012 Nat. Nanotechnol. 7 180–4
Shen W, Nelson J, McClelland A A, Heisterkamp A and [119] Abbott J, Ye T, Qin L, Jorgolli M, Gertner R S, Ham D and
Mazur E 2017 ACS Nano 11 3671–80 Park H 2017 Nat. Nanotechnol. 12 460–6
[89] Messina G C, Dipalo M, Rocca R L, Zilio P, Caprettini V, [120] Alivisatos A P et al 2013 ACS Nano 7 1850–66
Zaccaria R P, Toma A, Tantussi F, Berdondini L and [121] Tian B and Lieber C M 2013 Annu. Rev. Anal. Chem. 6 31–51
Angelis F D 2015 Adv. Mater. 27 7145–9 [122] Zhang A and Lieber C M 2016 Chem. Rev. 116 215–57
[90] Mullett W M, Lai E P C and Yeung J M 2000 Methods 22 [123] Parameswaran R and Tian B 2018 Acc. Chem. Res. 51
77–91 1014–22
[91] Duan X, Huang Y, Agarwal R and Lieber C M 2003 Nature [124] Abbott J, Ye T, Ham D and Park H 2018 Acc. Chem. Res. 51
421 241–5 600–8
[92] Ellis B, Mayer M A, Shambat G, Sarmiento T, Harris J, [125] Hällström W, Mårtensson T, Prinz C, Gustavsson P,
Haller E E and Vučković J 2011 Nat. Photonics 5 297–300 Montelius L, Samuelson L and Kanje M 2007 Nano Lett. 7
[93] Chen R, Tran T-T D, Ng K W, Ko W S, Chuang L C, 2960–5
Sedgwick F G and Chang-Hasnain C 2011 Nat. Photonics 5 [126] Piret G, Perez M-T and Prinz C N 2015 ACS Appl. Mater.
170–5 Interfaces 7 18944–8
[94] Doherty G J and McMahon H T 2009 Annu. Rev. Biochem. 78 [127] Oh S, Daraio C, Chen L-H, Pisanic T R, Fiñones R R and
857–902 Jin S 2006 J. Biomed. Mater. Res. Part A 78A 97–103

12
Nanotechnology 30 (2019) 264002 A K Fajrial and X Ding

[128] Oh S, Brammer K S, Li Y S J, Teng D, Engler A J, [135] Gates B D, Xu Q, Stewart M, Ryan D, Willson C G and
Chien S and Jin S 2009 PNAS 106 2130–5 Whitesides G M 2005 Chem. Rev. 105 1171–96
[129] Dalby M J, Gadegaard N and Oreffo R O C 2014 Nat. Mater. [136] Vazquez-Mena O, Gross L, Xie S, Villanueva L G and
13 558–69 Brugger J 2015 Microelectron. Eng. 132 236–54
[130] Capozza R, Caprettini V, Gonano C A, Bosca A, Moia F, [137] Chern W, Hsu K, Chun I S, de Azeredo B P, Ahmed N,
Santoro F and De Angelis F 2018 ACS Appl. Mater. Kim K-H, Zuo J, Fang N, Ferreira P and Li X 2010 Nano
Interfaces 10 29107–14 Lett. 10 1582–8
[131] Dipalo M et al 2018 Nano Lett. 18 6100–5 [138] Azeredo B P et al 2013 Nanotechnology 24 225305
[132] Ding Y, Yang Z, Bi C W C, Yang M, Xu S L, Lu X, [139] Madrid M, Saklayen N, Shen W, Huber M, Vogel N and
Huang N, Huang P and Leng Y 2014 ACS Appl. Mater. Mazur E 2018 ACS Appl. Bio Mater. 1 1793–9
Interfaces 6 12062–70 [140] Lee D W et al 2015 The Lancet 385 517–28
[133] Persson H, Li Z, Tegenfeldt J O, Oredsson S and Prinz C N [141] Tumaini B, Lee D W, Lin T, Castiello L, Stroncek D F,
2015 Sci. Rep. 5 18535 Mackall C, Wayne A and Sabatino M 2013 Cytotherapy 15
[134] Gates B D, Xu Q, Love J C, Wolfe D B and Whitesides G M 1406–15
2004 Annu. Rev. Mater. Res. 34 339–72 [142] Kay M A 2011 Nat. Rev. Genet. 12 316–28

13

You might also like