You are on page 1of 16

REVIEW

www.advmatinterfaces.de

Bioelectric Interface Technologies in Cells and Organoids


Zhaoguo Xue and Jianzhong Zhao*

nanostructured bioelectronics, owing


The past two decades have witnessed breakthroughs in cellular-scale to their versatile, miniaturized de-
bioelectronics and their widespread applications in life sciences, creating signs and capabilities of in vivo (or in
many powerful platforms for studying organisms directly and efficiently. vitro) sensing of cell activities, have
These advanced devices can integrate seamlessly and intimately onto/into emerged as a very promising direc-
tion of next generation cell interfaces.
target cells and organoids, providing unprecedented functions and To better fulfill the requirements of cell
revolutionary capabilities. Bioelectronics with nanostructured designs are electrophysiology recording, the long-
developed to allow for long-term, stable monitoring of electrophysiological term stability, high spatial and tem-
activities. This review summarizes nanostructured bioelectronics for cell poral resolution, design versatility and
electrophysiology recording, emphasizing the crucial roles of structural device miniaturization, represent four
key design considerations of nanostruc-
designs on functions and capabilities, e.g., intracellular access, high-density
tured bioelectronic devices. In these de-
multiplexed recording, multifunctional interfaces and the conformability to vices, the most important components
curvy biological shapes. Finally, the remaining challenges and opportunities in are the nanostructured electrodes[21–27]
nanostructured bioelectronics are identified, and perspectives on the future or field-effect transistors (FETs).[28–38] In
developments toward their practical applications are provided. the recording of cell membrane and
transmembrane potential, the electrodes
have advantages in high-density and
low-cost fabrication, but have difficul-
1. Introduction ties in low-amplitude cell signals and
picking up subthreshold, because of
Cells serve as the basic building blocks that make up all living the intrinsically large impedance.[21] A variety of nanostructured
organisms. Versatile forms of life activities are based on the in- architectures with strategically designed layout of integrated elec-
tracellular organization[1] and intercellular communication.[2–5] trodes or FETs have been developed to offer conformal and stable
The recordings of cell activities are therefore of pivotal impor- interfaces with diversely shaped cells/organoids, and to provide
tance for the understanding and prediction of advanced life be- long-term, continuous recording of biophysiological signals with
haviors. In recent years, there has been growing interest in de- high sensitivity and high fidelity.[39–56] For example, many differ-
velopments of electrical,[6–9] optical,[10–12] chemical,[11] and ther- ent forms of needle type architectures were developed for cell
mal interfaces technologies for sensing and monitoring of cells interfacing, ranging from nanoprobes,[57–61] nanopillars,[42,62–65]
and organoids.[13–20] Among these bioelectronic interfaces, the nanotubes,[66] nanocrowns,[24] to nanovolcanos.[67] Aside from
these 1D nanostructured configurations, 2D planar type,[68–71]
as well as diverse 3D architectures have been devised to allow
Z. Xue
National Key Laboratory of Strength and Structural Integrity
monitoring with higher spatial resolutions.[10,39–49,72] Therefore,
Institute of Solid Mechanics the cell electrophysiology activities can be displayed in real-time
School of Aeronautic Science and Engineering for biological and biomedical studies. It is noteworthy that the
Beihang University structural designs of such bioelectronics play vital roles on their
Beijing 100191, P. R. China performances and capabilities. However, reviews systematically
J. Zhao elaborating the relationship between structural designs and func-
Applied Mechanics Laboratory
Laboratory of Flexible Electronics Technology tions of representative nanostructured bioelectronics are still
Department of Engineering Mechanics insufficient.
Tsinghua University In this review, we highlight the representative nanostruc-
Beijing 100084, P. R. China tured bioelectronics (Figure 1) for cell electrophysiology record-
E-mail: jianzhongzhao@tsinghua.edu.cn
ing, with an emphasis on their structural designs, functions
The ORCID identification number(s) for the author(s) of this article and potential applications. The next section summarizes nee-
can be found under https://doi.org/10.1002/admi.202300550 dle type bioelectronics with a diversity of architectural designs.
© 2023 The Authors. Advanced Materials Interfaces published by The content then shifts to planar type for cells, and scaffold,
Wiley-VCH GmbH. This is an open access article under the terms of the conformal and implantable type bioelectronics for organoids.
Creative Commons Attribution License, which permits use, distribution The summary and outlook section outline opportunities and
and reproduction in any medium, provided the original work is properly challenges that exist for future developments of nanostructured
cited.
bioelectronics.
DOI: 10.1002/admi.202300550

Adv. Mater. Interfaces 2023, 2300550 2300550 (1 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

ize NW FET nanoprobe arrays with controllable tip geometry and


FET size, enabling the study of the impact of the nanoprobe size
and geometry on the intracellular recording.[101] It was shown
that the nanoprobes with the smallest radii of curvature and FET
channel lengths could facilitate the recording of full amplitude
intracellular action potentials and subthreshold features.[102] In
addition, arrays of these nanoprobes can be exploited to allow
multiplexed recording from single cells and cell networks.[103,104]
Based on the AFM platform, the electroporation of nanofoun-
tain probe technology can deliver molecules into cells in a man-
ner that is highly efficient and gentler to cells.[105,106] The electro-
poration of single HeLa cells was demonstrated. Furthermore, a
system for combined optical, force, and electrical measurements
Figure 1. A summary of nanostructured bioelectronics for cells and based on the AFM nanoprobe was developed,[107] which consists
organoids. of a nanovolcano probe at the tip of a suspended microcantilever.
Impedance measurements on mechanically stimulated neona-
tal rat cardiomyocytes were achieved using these nanovolcano
2. Needle Type Bioelectronics for Cells probes. The nanoprobes can also be constructed with various
As a class of most wide-used structures of nanomaterials, needle materials and nanodevices, such as carbon nanotubes (CNTs),
type devices have become a powerful tool for studying biologi- Si NWs, and gold nanopillars.[77,78,108] Owing to their high me-
cal interfaces.[9,73–85] The development of such devices could not chanical strength and high electrical conductivity, CNTs are at-
only meet the growing demand for highly sensitive and selec- tractive for cell probing, noticing that the dimensions of CNTs
tive detection of single cells, but also provide an ideal platform to would potentially allow the interrogation of submicrometresized
penetrate the cell membrane for intracellular detection, which is organelles deep within the cell, without causing evident damage.
difficult to achieve with other structural configurations.[74,86] The Singhal et al.[108] developed a multiwalled CNT-based endoscope
existing penetration methods for intracellular access can be clas- for interrogating cells, which could transport fluids and perform
sified into two main categories,[87–90] , i.e., spontaneous penetra- optical and electrochemical diagnostics without disrupting the
tion and assisted penetration. The spontaneous penetration usu- cell at the single organelle level.
ally relies on the sharp physical geometry of the nanostructure or
the gravity/adhesion forces of cells, while the aided penetration
requires additional chemical, electrical, or optical processes. 2.2. Nanopillars

Nanopillar array-based devices can penetrate the cell membrane


2.1. Nanoprobes and enter the intracellular compartment, allowing the transfer of
various biological molecules and detection of intracellular electri-
With conventional array-based recording devices, the contact- cal and biochemical signals.[77,78] To obtain a high-fidelity intra-
ing surfaces are typically confined to regions only near the bot- cellular signal, it is essential for the nanopillar structure to pene-
tom of cells. These issues could be addressed by integrating trate the cell membrane and form a tight coupling with the mem-
needle type nanoelectronics in a support of operating platform brane. This coupling is highly dependent on the shape and size
(e.g., atomic force microscope (AFM) cantilever, glass pipette and of the contact region. Generally, it has been shown that an easier
nanomanipulator).[91–94] This kind of nanoprobes could be em- intracellular access via spontaneous penetration can be achieved
ployed for probing intracellular environment with a high spa- by decreasing the tip diameter or increasing the height.[109,110]
tial resolution (single cell level). Due to the nanoscale size and As shown in Figure 2b-i, a vertical ultrasharp nanopillar (with
semi-implantable property, the nanoprobe could greatly reduce a tip diameter below 10 nm) was reported to enable long-term
the damage of cell during membrane penetration, allowing the recordings of intracellular potentials.[63] A high-performance
precise and long-term positioning into cells.[95,96] intracellular access is crucial for the high-quality and long-
Tian et al.[57] developed a novel nanoprobe based on kinked term recording, while maintaining cell viability. In addition to
NW FETs, which allowed the localization of a point-like FET de- the spontaneous poration, the electroporation (Figure 2b-ii) is
tector at the kinked tip for the realization of facile intracellular the most widely-used method to facilitate the cell membrane
recordings (Figure 2a). The kinked Si NW modified with phos- penetration.[62,111] Due to the sharp tip (≈100 nm) of the verti-
pholipid modification enable spontaneous penetration of the cell cal nanopillar tightly coupled to the cell membrane as shown in
membrane, without applying any external forces.[98] Meanwhile, Figure 2b-iii, a large electric field can be generated with only a
the high surface-to-volume ratio of NWs and their operation as small voltage (e.g., ≈1–3 V) to locally increase the permeability
FETs afford excellent sensitivity to voltage signals, which ensured of the cell membranes.[64] When a low voltage electroporation
the real-time electrical recording of the spontaneous penetration was applied on the nanopillars, nanopores were introduced in the
process.[99,100] Based on a similar principle, a scalable U-shaped cell membrane, and high-quality intracellular potentials could be
Si NW FET-based nanoprobe with the ability to record full am- recorded continuously for several minutes.[112] Furthermore, the
plitude intracellular action potentials was proposed,[58] as shown optoporation was also developed as an effective method to im-
in Figure 2a. A key advancement is an assembly method to real- prove the penetration, where focused high-power laser pulses

Adv. Mater. Interfaces 2023, 2300550 2300550 (2 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Figure 2. Needle type bioelectronics for cells. a) Nanoprobes. i) Kinked NW-based probes. Reproduced with permission.[57] Copyright 2010, American
Association for the Advancement of Science. ii) U-shaped NW FET probe. Reproduced with permission.[58] Copyright 2019, Nature Publishing Group.
b) Nanopillars. i) Ultra-sharp nanopillars. Reproduced with permission.[63] Copyright 2021, Wiley-VCH. ii) Membrane poration mechanisms. Reproduced
with permission.[62] Copyright 2019, Wiley-VCH GmbH. iii) Vertical electrode arrays. Reproduced with permission.[64] Copyright 2012, Nature Publishing
Group. iv) 3D vertical electrode arrays. Reproduced with permission.[42] Copyright 2022, Nature Publishing Group. c) Quasi-1D tubes. i) Hollow nanotube
electrode. Reproduced with permission.[66] Copyright 2019, American Chemical Society. ii) Nanocrown electrodes. Reproduced with permission.[24]
Copyright 2022, Nature Publishing Group. iii) Nanovolcano electrode. Reproduced with permission.[67] Copyright 2019, American Chemical Society.
iv) Patch clamp technique. Reproduced with permission.[97] Copyright 2019, Nature Publishing Group.

Adv. Mater. Interfaces 2023, 2300550 2300550 (3 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

were introduced at the interface to penetrate the cell membrane, the neuroscience toolbox.[97] As the gold standard device for
allowing simultaneously recording of both extracellular and in- electrophysiology,[124,125] patch clamps could provide high-quality
tracellular potentials.[113–116] When comparing to the electropora- action potential recordings by adsorbing cell membranes to form
tion, the optoporation does not interfere with spontaneous cell a coupling interface with the intracellular environment.[126]
activity, does not imply any recording blind time, and enables a Needle type bioelectronics have great advantages in penetrat-
very long-term observation. ing the cell membrane for intracellular detection (i.e., sponta-
To fulfill the requirements of high-fidelity transmembrane neous penetration and assisted penetration), which is difficult to
potential recording in single cells or multicellular networks, achieve with other configurations of bioelectronics. Three types
Gu et al. reported a scalable 3D FET device (Figure 2b-iv) of needle type bioelectronics are classified due to their geome-
transformed from a 2D precursor for intra- and inter-cellular tries, such as nanoprobes, nanopillars and quasi-1D nanotubes.
recording.[42] The 3D geometry allows the FETs to penetrate the Nanoprobe devices can detect the entire cell membrane and in-
cell membrane and record low-amplitude subthreshold signals tracellular domain with a high spatial resolution in support of
inside the cell. On the basis of the developed devices, the intracel- the operating platform, while such nanoprobe is inadequate in
lular action potentials and intercellular signal conductions were simultaneous multisite measurements. Nanopillar and quasi-1D
accurately recorded in real time. An array of the 3D FET devices tube devices are specially made for recording multicellular net-
with different structural designs was also demonstrated, which work, while the contacting surfaces are typically confined to the
could interrogate cells at three different depths in a 3D microtis- bottom of cells. Further advances will follow from the develop-
sue. ment of routes to high spatial resolution, multiplexed, intracellu-
lar and intercellular recording bioelectronics.

2.3. Quasi-1D Tubes


3. Planar Type Bioelectronics for Cells
The complex vertical quasi-1D nanotubes have been demon- With the fast developments of micro/nanofabrication and pro-
strated, thanks to the fast developments of advanced manufac- cessing technologies, 2D bioelectronics with a high-density
turing technologies at nanoscale.[24,66,67,97] Methods for intracel- integration have been devised to achieve a noninvasive,
lular delivery of molecules or nanoparticles (e.g., DNA plasmids, long-term, and multiplexed extracellular recording of cell
drug molecules, quantum dots, and gold nanoparticles) are es- electrophysiology.[127–130] The planar microelectrode array is a
sential for the development of modern biological and biomed- conventional tool for the electrophysiological investigation of
ical techniques, and have been widely used in single-cell stud- cells, with the advantages of low-cost fabrication, minimal in-
ies, drug delivery, disease diagnosis, pharmaceutical therapeu- vasiveness to cells and high-throughput recordings.[131–134] How-
tics, gene regulation, and intracellular imaging.[117,118] Different ever, due to the 2D geometry of the electrode, it is basically not
from nanopillars, the hollow nanotube structures can provide a capable of directly recording the intracellular action potential. Be-
unique nanochannel that bridges the extracellular and intracel- sides, the micron-sized electrode usually records the electrical po-
lular microenvironments, facilitating the delivery of bio-cargoes tentials of the cell population rather than a single cell, which can
into cells through the nanochannel. be well addressed by introducing nanoscale 2D structures.[135]
Hollow nanotubes have been exploited to serve as reliable
means of delivery with high cell viability.[119–122] For example,
3.1. Nanowells
an electrophoretic system based on vertical hollow nanotubes
(Figure 2c-i) was developed to enable intracellular delivery of in-
A bioelectronic system consisting of a planar microelectrode and
dividual nanoparticles and real-time monitoring of the single-
five patterned nanowells was proposed to record the intracellu-
particle delivery process.[66] The hollow tubular nanoelectrodes
lar action potential of a single cardiomyocyte,[136] as shown in
were shown to be able to delay the resealing of the cell mem-
Figure 3a. The electroporation of nanowell electrodes could gen-
branes and induce the active membrane fusion, thereby im-
erate electric fields across the cardiomyocytes, and then pene-
proving the sealing resistance and prolonging the intracellu-
trate the cell membrane to allow a good electrical coupling be-
lar recording time.[123] Jahed et al.[24] proposed semi-hollow
tween the microelectrode and cardiomyocytes for the intracel-
nanocrown electrodes with mechanical robustness, which could
lular monitoring. The micron-sized nanowell array enabled the
achieve high electroporation success rates (≈99%) with high ac-
penetration point to stay under a single cell for the recording of
curacy and signal strength for cardiomyocyte intracellular ac-
permeability and action potential. Compared to those of the ex-
cess (Figure 2c-ii). The nanocrown structures could induce the
tracellular recording by conventional planar microelectrode array,
cell membrane to wrap around the outer surface and adhere
the nanowell microelectrode array could measure the intracellu-
to the inner core, which stabilizes the membrane-electrode in-
lar potential with higher quality, reduced noise root-mean-square,
terface. Similarly, volcano-shaped nanoelectrode (nanovolcano)
and higher signal-to-noise ratio.
was also proposed[67] for long-term electrophysiological record-
ings (Figure 2c-iii). The 100-nm-thick nanovolcano wall has a
nanopillar-like shape, which could induce high-curvature re- 3.2. Planar FETs
gions in the cell membrane, thereby increasing the chance
of intracellular access. The patch-clamp technique which is The function of biological organs relies on the dynamics of con-
originally developed to record currents of ions flowing in the stituent cells and their coupling, and thus the simultaneous ob-
cell membranes (Figure 2c-iv), has become a true stalwart of servation of a large number of cells over long time is essential for

Adv. Mater. Interfaces 2023, 2300550 2300550 (4 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Figure 3. Planar type bioelectronics for cells. a) Nanowells. Nanowell-based microelectrode for penetrating the cardiomyocyte membrane by electro-
poration and recording intracellular action potentials. Reproduced with permission.[136] Copyright 2022, Nature Publishing Group. b) Planar FETs. i)
Colorized electron micrograph image of a neuron on a silicon chip with a linear array of buried-channel FETs. Reproduced with permission.[137] Copyright
2005, Wiley-VCH. ii) Optical image of a cortex neuron connected to a NW device. Reproduced with permission.[138] Copyright 2006, American Associa-
tion for the Advancement of Science. iii) Schematic illustration of graphene-based solution gated FETs for the detection of cell signals. Reproduced with
permission.[139] Copyright 2011, Wiley-VCH.

the biological research.[140] Based on the complementary metal guided neuronal growth over Si NW FETs. Then a highly local-
oxide silicon (CMOS) technology, Voelker et al.[137,141] demon- ized synapse-like junction could be formed at regions where the
strated low-noise electrolyte–oxide–silicon FETs for parallel mon- neuronal axon or dendrite crosses the NW device.
itoring of neurons at high spatial and temporal resolution. Dur- Moreover, the graphene electrolyte-gated FET array was de-
ing the transistor recording, a neuron was directly placed onto veloped for the detection of the cell electrophysiological activity
to the exposed gate oxide of a FET, as illustrated in Figure 3b- (Figure 3b-iii), using large-area graphene films grown by chem-
i. The source–drain current could be directly modulated by the ical vapor deposition on copper foil.[139] The action potentials of
change in the local extracellular potential in the electrolyte of the these cardiomyocyte-like HL-1 cells could be effectively detected
cleft between the cell and transistor. The cell–transistor junction and resolved by culturing cells on the FET array. Due to the low
could be regarded as a type of amplifier without interference of noise and the large transconductive sensitivity, these graphene
the electrolyte–metal contact or stray capacitances. transistors offer an outstanding advantage of high signal-to-noise
In addition, planar FETs based on semiconductor nanoma- ratios. Through the entire transistor array, the propagation of the
terials (such as Si NWs,[142–144] CNTs[145–147] and graphene,[148] cell signals across the layer was successfully tracked by analyzing
MoS2 [149] ) have attracted widespread attention, because of their the multiplexed data. Integration of different types of devices to
capabilities of highly-sensitive and label-free detections. The ap- measure the same cell is also possible. The simultaneous record-
plication of Si NW FETs for extracellular recording from cul- ing of electrophysiological signals from the same cardiomyocyte
tured neurons was first reported by the Lieber group in 2006. As was carried out by both graphene and Si NW FETs.[150]
shown in Figure 3b-ii, the as-fabricated Si NW FET array could The research on planar type bioelectronics remains insuffi-
be selectively passivated by patterning polylysine to promote the cient. Planar type bioelectronics are relatively low-cost because

Adv. Mater. Interfaces 2023, 2300550 2300550 (5 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

of their compatibility with state-of-art large-scale integrated pla- 4.2. Porous Foams
nar fabrication techniques, thereby being applicable to high-
throughput recordings. Similar to nanopillar and quasi-1D tube The emerging 3D cell culture systems have boosted the de-
devices, the recording area is typically confined to the bottom of velopment of many fields, such as pathophysiological study,
cells. The 2D geometry of the electrodes lead to a weak coupling tissue regeneration, and drug screening. Among the various
interface. Opportunities may exist in the multiplexed extracellu- 3D cell culture systems, scaffold-based 3D cell culture sys-
lar recording of cell electrophysiology, which are useful for un- tems have attracted extensive attention,[40,69,72,126,157–159,172] be-
derstanding intercellular communications, such as neurons. cause such porous scaffolds can simultaneously offer mechanical
support and 3D microenvironments for the cell attachment and
tissue formation in tissue engineering.
Qin et al. developed a stretchable and multifunctional scaffold
4. Scaffold Type Bioelectronics for Organoids type platform capable of 3D cell culture, mechanical loading, and
Organoid, as a miniaturized and simplified version of an electrochemical sensing.[40] The scaffold type 3D bioelectronics
organ, can self-organize in 3D culture, owing to their self- were fabricated by attaching the networks of gold nanotubes (Au
renewal and differentiation capacities.[10,13,43–45,151] 3D nanos- NTs) on porous polydimethylsiloxane (PDMS), and linking Gly-
tructured bioelectronics have advantages in native state of Arg-Gly-Asp (GRGD) with Au NTs via covalent bond (Figure 4b).
cells, as well as multisite, concurrent electrophysiological Such scaffold type 3D bioelectronics offered very good biocom-
monitoring.[10,18,23,43,44,46,48,152–156] In this section, the rep- patibility, excellent stretchability, and stable electrochemical sens-
resentative scaffold type bioelectronics for organoids are ing performance, with capabilities of mimicking the mechan-
introduced.[40,152,157–159] Owing to the geometrical features, otransduction of articular cartilage and monitoring the stretch-
such bioelectronics can provide the platforms for the struc- induced signaling molecules in real time.
tural support and growth of cells, while accommodating the
spatially distributed electrodes capable of recording the cell
activities. Hence, scaffold type bioelectronics have potentials in
integrating a higher density of electrodes. Such potentials com- 4.3. Vertical Electrode Arrays
bined with structural designs and functionalized materials can
serve as a multifunctional platform in exploring the generation The shape morphing between free-standing 3D frameworks
and conduction of electrical impulse in cells or intercellular and their 2D precursors can be achieved by introduc-
networks.[39,40,42,156,160] ing plastic strain programming. Unlike the 3D assembly
methods,[161,162,165,166,168,169] such strategy could provide free-
standing 3D geometries. By exploiting the plastic deformations,
Soscia et al. developed a 3D flexible spike-like multi-electrode ar-
4.1. Assembled Scaffolds rays (Figure 4c), where the hinges were plastically deformed by a
customized apparatus, allowing the probes to stand upright with-
The mechanically guided 3D assembly methods provide a power- out additional supports.[39] The developed 3D multi-electrode
ful route to 3D electronic devices over different length scales, with devices containing 10 probes and 80 electrodes can be used
a rich diversity of accessible 3D geometric configurations.[161–171] to non-invasively interrogate the suspended neurons, with a
The excellent compatibility of such assembly methods with well- capability of visualizing the spatial and temporal mapping of
established planar nanofabrication and processing techniques al- electrophysiological data across a 3D volume.
lows their applicability to a broad set of high-performance materi- Such vertical 3D multi-electrode arrays with precisely con-
als. Besides, the development of routes to complex 3D structures trolled configurations can easily integrates with standard com-
with feature sizes in the mesoscopic range is of increasing. The mercially available electrophysiology hardware.[39] To precisely
purpose is to establish methods for controlling the structural de- control the bending location during actuation, a hinge region was
signs for applications as metamaterials, offering engineered be- incorporated into the structural designs. The bending region fea-
haviors with optical, thermal, acoustic, mechanical that do not tures a void in the polyimide, resulting in an inherently flexible
occur in the natural world, which can be summarized folding, region compared to the rest part. Polyimide was chosen as the
rolling and mechanical assembly.[166] main structural component of the probes due to its flexibility and
Yan et. al. reported a mechanically guided assembled strat- biocompatibility. To ensure rapid and reproducible actuation of
egy for advanced 3D designs in micro/nanomanufacturing which the probes, a costumed actuation apparatus was also developed.
have potentials in many fields including biomedical engineering, Such customized platforms are important step in facilitating non-
metamaterials, electronics, electromechanical components.[156] invasive electrophysiological characterization of 3D networks of
Sophisticated cell scaffolds can leverage high-performance com- electroactive cells in vitro.
ponents to allow interaction and communication with live cells The rich diversity of accessible 3D geometric configurations
and tissues. The developed electronic scaffolds were used for en- is crucial to the culture of organoids. Three techniques are de-
gineered dorsal root ganglion neural networks. As a demonstra- tailly discussed to show their design strategies, such as 3D assem-
tion, 3D bilayer nested cages of epoxy transferred onto optical- bly, porous foam, and plastic deformation methods. Integration
quality glass to enable high-resolution, in situ imaging, serve of functional components (metal electrodes or FETs) on the 3D
as growth platforms for neural networks of dorsal root ganglion scaffold structures, enables multisite, concurrent electrophysio-
cells dissociated from explants from rats. logical monitoring within organoids. Such bioelectronics provide

Adv. Mater. Interfaces 2023, 2300550 2300550 (6 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Figure 4. Scaffold type bioelectronics for organoids. a) Assembled scaffolds. 3D mesostructures as electronic cell scaffolds. Reproduced with
permission.[156] Copyright 2017, National Academy of Sciences. b) Porous foams. Stretchable scaffold-like 3D bioelectronics. Reproduced with
permission.[40] Copyright 2021, Wiley-VCH. c) Vertical electrode arrays. 3D multi-electrode arrays transformed from a 2D precursor by plastic defor-
mations of the hinges. Reproduced with permission.[39] Copyright 2020, The Royal Society of Chemistry.

reliable, advanced platforms for studying the mechanism of self- and shape morphing capability (e.g., transformable from 2D to
renewal and differentiation capacities of organoids. 3D).[10,43–45,151,155,160,173–175] In this section, several conformal type
bioelectronics for organoids and issues are introduced. The ad-
5. Conformal Type Bioelectronics for Organoids vanced devices consist of multilayered stacks of ultrathin active
components, including sensors that have dimensions compara-
To allow conformal wrapping around 3D organoids, the bioelec- ble to those of a single cell. For example, these systems can mon-
tronics should be well designed to offer high degree of flexibility itor specific collections of neurons in the central and peripheral

Adv. Mater. Interfaces 2023, 2300550 2300550 (7 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Figure 5. Conformal 3D bioelectronics for organoids. a) Self-bending leaflets. 3D shell microelectrode arrays composed of self-bending polymer leaflets
with metal coating for electrophysiology recording of brain organoids. Reproduced with permission.[43] Copyright 2022, American Association for
the Advancement of Science. b) Assembled frameworks. 3D frameworks as compliant, multifunctional interfaces for spheroids. Reproduced with
permission.[10] Copyright 2021, American Association for the Advancement of Science. c) Self-rolling/-twining shells. i) 3D self-rolled biosensor arrays.
Reproduced with permission.[44] Copyright 2019, American Association for the Advancement of Science. ii) 3D microchannels of the cuff-type implant.
Reproduced with permission.[154] Copyright 2015, Wiley-VCH. iii) 3D twining electrode. Reproduced with permission.[176] Copyright 2019, American
Association for the Advancement of Science.

nervous systems, as means to elaborate connections to complex Inspired by the shape of electroencephalography caps, Huang
behavioral responses. et al. developed 3D shell microelectrode arrays composed of self-
bending polymer leaflets with metal coating for electrophysiology
recording of brain organoids.[43] The solvent exchange between
5.1. Self-Bending Leaflets acetone and water for the gradient cross-linked SU8 triggers the
controlled bending of leaflets, resulting in the 2D-to-3D transfor-
Conformal multielectrode arrays can provide noninvasive record- mation of shell microelectrode arrays (Figure 5a). The thickness
ing and network mapping of extracellular potential for organoids. and extent of cross-linking extent are two key design parameters

Adv. Mater. Interfaces 2023, 2300550 2300550 (8 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

for achieving different bending angles. By tuning the bending an- dius of curvature can be controlled by the geometric design and
gle, shape, and electrode pattern, a customizable conformability the residual stress level.
can be realized for organoids with different sizes. Karnaushenko et al. reported mechanically adaptive mi-
When comparing to conventional planar microelectrode ar- crochannels of the cuff-type implant with integrated high-
rays, such 3D microelectrode arrays can address the limitations performance electronics (Figure 5c-ii), including signal ampli-
that the recording contact area is restricted to the bottom of the fiers and logic circuitry based on indium gallium zinc oxide
3D organoids. For a better signal-to-noise ratio, the needle type (IGZO) transistors.[154] The device can be bent, or self-assembled
electrodes are used to provide recording access to the interior of into a swiss roll like microtube,[178–180] by adjusting the envi-
the measured cell via penetration, while such 3D microelectrode ronmental conditions, including the solution composition and
arrays have advantages in providing noninvasive and high-speed pH. The diameter of the tubular structures can be tuned over a
recording and network mapping of extracellular electric field wide range from 50 μm to 1 mm. The differentiation and guided
potential. growth of neural stem cells in the microchannels of the 3D device
were demonstrated. The integrated electronics allowed the detec-
tion of tiny amounts of ionic liquids in the microchannel, mim-
5.2. Assembled Frameworks icking the detection of polarization/depolarization processes in
neural microconduits.
Mechanically-guided 3D assembly methods typically involve the Peripheral neuromodulation has been widely used both in
2D-to-3D geometric transformation of a patterned high-modulus clinical practices and neuroscience research.[181–184] However, the
thin film on a low-modulus elastomer substrate much thicker mechanical and geometrical mismatches at electrode nerve inter-
than the film, driven by compressive buckling.[162] Park et al. faces and surgical implantation often induce irreversible neural
developed a class of compliant 3D electrical interfaces, through damage. Zhang et. al. reported climbing-inspired twining elec-
compressive buckling of patterned 2D electrode array consist- trodes for peripheral nerve stimulation and recording.[176] The
ing of polymer support and metal electrode/interconnect.[10] In reported 3D twining electrode has permanent shape reconfigura-
particular, the 2D precursor pattern can be customized to al- bility, distinct elastic modulus controllability (from ≈100 MPa to
low the resulting 3D electronics to be matched with various dif- ≈300 kPa), and shape memory recoverability at body tempera-
ferent shapes of organoids, highlighting the design versatility ture (Figure 5c-iii). Importantly, the 3D twining electrode is also
(Figure 5b). Two examples of 3D electrical interfaces are provided compatible with traditional 2D planar processing. Similar to the
to study the spreading of coordinated bursting events of an iso- climbing process of twining plants, the 2D stiff twining elec-
lated cortical spheroid and of a pair of such spheroids. The multi- trode can naturally self-climb onto nerves driven by 37 °C normal
functional interface is capable of simultaneous electrophysiolog- saline.
ical monitoring of 16 spheroids. Conformal type bioelectronics are designed to offer high de-
With the tailored geometric features and low bending stiff- gree of flexibility and shape morphing capability. The electrodes
nesses, the assembled 3D framework can gently envelop and hold or FETs are distributed on the surface of organoids, capable of
an individual neural spheroid, with multifunctional devices on monitoring the multisite potentials. Importantly, the 3D shape
the individual wings, such as electrical, optical, chemical, and of such bioelectronics can be tailored by changing the geome-
thermal interfaces. These design features, together with soft, tries and bending stiffnesses, thereby showing potential in on-
proximity contacts to the surface of the spheroid, yields high per- demand design for versatile organoids. The challenges and op-
formance in recording of neural activity.[10] Based on finite ele- portunities remain in morphing capability and miniaturization
ment analysis (FEA), the optimized selection of design parame- of such bioelectronics.
ters to ensure contact with extremely low forces at the soft tissue
interfaces can be easily realized. Besides, the elastomeric sub-
strate can be reversibly compressed or stretched to alter the 3D 6. Implantable Type Bioelectronics for Organoids
geometry to accommodate dynamic, natural changes in the size By matching the subcellular feature sizes and mechanical prop-
of the spheroid as it evolves and grows. erties of cells, the 3D implantable bioelectronics could be seam-
lessly integrated with organoids and tissues to achieve recording
of neural activities in brain,[7,25,48,185–191] as well as mapping of the
5.3. Self-Rolling/-Twining Shells action potential propagation in cardiac tissues.[49,192–195]

3D rolled-up nanomembrane bioelectronics can be robustly at-


tached to a tissue to enable the enclosure of neuronal cells or 6.1. Porous 3D Mesh
nervous fibers. The rolled-up nanomembrane structures can be
actuated from planar configurations mainly by light, pH, tem- To address the dimensional and mechanical mismatch of probes
perature, electrical or magnetic stimuli.[154,177,178] Kalmykov et al. with the brain tissue, Xie et al. developed a 3D ultra-flexible bio-
reported 3D self-rolled biosensor arrays through use of residual electronic network consisting of NW FET sensors to serve as
stresses in the metal/polymer support bilayer structure.[44] The minimally invasive brain probes.[48] The high flexibility of such
3D FETs and microelectrodes were implemented to interface hu- probes precludes direct insertion into tissue, hence, rapid freez-
man cardiac spheroids for electrophysiological monitoring and ing provides sufficient rigidity to allow controlled insertion into
investigation of the complex signal transduction in 3D cell assem- neural tissue. Due to the high flexibility of the network configu-
blies toward an organ-on-an-electronic-chip (Figure 5c-i). The ra- ration, the porous network probes (Figure 6a-i) can be integrated

Adv. Mater. Interfaces 2023, 2300550 2300550 (9 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Figure 6. Implantable type bioelectronics for organoids. a) Porous 3D mesh. 3D nanoelectronic networks serve as minimally invasive brain probes.
Reproduced with permission.[48] Copyright 2015, Nature Publishing Group. b) Nanofilm. Self-assembled 3D nanofilm electrode arrays as long-term
neural interfaces. Reproduced with permission.[47] Copyright 2021, Wiley-VCH. c) Neuron-like probes. Bioinspired neuron-like electronics as 3D neural
probes for stable, long-term recording. Reproduced with permission.[46] Copyright 2019, Nature Publishing Group.

with the brain tissue without inducing large mechanical con- be naturally deformed to conform to the entire surface of brain
straints. The biocompatible porous probes were implanted into organoids for long-term, multisites, continuous recording.[45]
rodent brains with minimal surgical tissue damage, with capabil- The stretchable mesh nanoelectronics can well match the me-
ities of long-term stable recording. chanical properties of brain organoids and be folded by the
Floch et. al. developed 3D mesh nanoelectronics consisting organogenetic process of progenitor or stem cells. The inte-
of highly stretchable serpentine wires(Figure 6a-ii), which can grated stretchable electrode arrays show no interruption to brain

Adv. Mater. Interfaces 2023, 2300550 2300550 (10 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

organoids, adapt to the volume and morphological changes. The 7. Summary and Outlook
seamless and noninvasive coupling of electrodes to neurons en-
ables long-term stable recording. This review summarizes the developments of three general
classes of nanostructured bioelectronics for cells (Table 1), high-
lighting their structural designs, device functions and potential
applications. First, the representative architectures (needle type)
6.2. Nanofilm of 1D nanostructured bioelectronics are demonstrated for highly
sensitive and selective intracellular detection of single cells by
Gao et al. presented high-density, free-standing gold nanofilm penetrating the cell membrane. While these 1D nanostructured
electrode arrays as a stable electrode-tissue interface.[47] The 2D bioelectronic devices offer tight coupling interfaces with cells, the
nanofilm electrode arrays (10 nm in thickness) were encapsu- invasiveness could affect the long-term recording of cell activities.
lated by biodissolvable polymer carriers, enabling their reliable Based on the well-established planar nanofabrication and pro-
implantation into deep brain tissues. After implantation and dis- cessing techniques, large-area, 2D nanostructured bioelectronics
solution of polymer carriers, the ultrathin electrode array was re- with high-density integration of electrodes/FETs have been de-
leased and assembled into 3D configurations to interface with veloped for multisite recording of cells, which are useful for un-
neural tissues (Figure 6b). Chronically implanted nanofilm elec- derstanding of intercellular communications. In addition, three
trode arrays can form intimate and innervated interfaces with types of representative architectures of 3D nanostructured bio-
neural tissue, enabling stable recordings across multiple brain electronics are discussed, including scaffold, conformal, and im-
regions over several months. plantable types. The scaffold type bioelectronics can be effectively
Neural electrodes based on free-standing nanoscale materi- used for studying the intercellular communication, whereas the
als offer unique opportunities to interrogate neural systems at reported devices all had a relative low electrode/FET density as
unprecedented spatiotemporal scales, such as nanowires, nan- compared to 2D nanostructured bioelectronics due to the compli-
otubes, and nanofilms. However, such ultra-flexible materials cated fabrication. The architecture of conformal type bioelectron-
have precluded their direct implantation into the brain tissues. ics can be exploited to non-invasively wrap around the organoids
The methods of dissolution of polymer carriers can be extended with diverse shapes. However, it remains challenging to scale
to assemble a variety of free-standing nanoscale materials into down such devices for single cell monitoring, based on the exist-
implantable, high-density neural probes, which offer important ing fabrication/assembly methods. The implantable nanostruc-
opportunities in basic neuroscience and biomedical applications. tured bioelectronics consisting of highly flexible network archi-
tecture allow for multisite long-term monitoring of tissue cul-
ture with negligible mechanical constraints. But it is difficult to
6.3. Neuron-Like Probes achieve a precise positioning of recording sites in the implanted
condition.
To study the brain, bioinspired and biomimetic strategies have Bioelectric interface technologies aspire to explore the under-
begun to be applied to the development of neural probes. In- lying mechanism of life activities in cells and organoids by bring-
spired by the structural features and mechanical properties of ing together the most important advances in the discipline, en-
neurons, Yang et al. developed neuron-like electronics (NeuE) as hancing the comprehensive performance excellence that cov-
neural probes with negligible immune responses, which exhibit ers various metrics/aspects. The needle type bioelectronics have
seamless interpenetrating interfaces with the brain.[46] NeuE great potentials in the fields of intracellular monitoring, such
probes demonstrated a stable single-unit recording of individ- as the silicon nanoprobes can be exploited to record intracel-
ual cells in the nearly native physiological context without loss in lular electrical signals of a single living cell. In contrast, the
recording quality (Figure 6c). NeuE are capable of mimicking the patch-clamp technique could provide high-quality action poten-
structural features (e.g., shape and size) and mechanical prop- tial recordings at the cell membranes. The 2D and 3D bioelec-
erties (e.g., flexibility) of neurons, representing a key advance tronics have more advantages in studying intercellular commu-
of this work. In this biomimetic design, the sizes of the metal nications within organoids. Such platforms with unprecedented
recording electrode and interconnect match those of the soma functions and revolutionary capabilities enable in vivo (or in
and neurite of the typical pyramidal neuron. The interconnect vitro) sensing of cell activities, showing potential in revealing the
and axon have similar and the thin polymer insulation is analo- fundamental mechanisms.
gous to the myelin sheath. Time-dependent histology and electro- Looking to the future, many challenges and opportunities re-
physiology studies also reveal a functionally stable interface with main in developing and improving nanostructured bioelectron-
the neuronal and glial networks, thus opening opportunities for ics for cells. For examples, the development of novel, efficient
next-generation brain–machine interfaces. and low-cost methods for nanofabrication and/or assembly is
Implantable type bioelectronics capable of multisite long-term the key to promote nanostructured bioelectronics toward prac-
monitoring of tissue culture with negligible mechanical con- tical applications. In view of the current research status of bio-
straints. Such flexible bioelectronics are compatible with in vivo logical nanodevices, the design optimization of device architec-
tissues and organoids with minimal surgical tissue damage, with ture with enhanced cell-device interface coupling and fabrica-
capabilities of long-term stable recording. By customizing the tion feasibility still requires further efforts. Opportunities exist in
structural features and mechanical properties, such implantable the incorporation of artificial intelligence (AI) techniques to as-
bioelectronics could serve as biomimetic cells, capable of electro- sist the design and fabrication of nanostructured bioelectronics
physiological monitoring inside in vivo tissues and organoids. devices.

Adv. Mater. Interfaces 2023, 2300550 2300550 (11 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

Table 1. Summary of nanostructured bioelectronics.

Dimensions Architectures Advantages Disadvantages

1D Needle • Intracellular recording • Invasiveness


• High sensitivity and selectivity • Restricted contact region
• Tight coupling interface • Single site monitoring
2D Planar • Low-cost fabrication with large-area and high-density integration • Extracellular detection
• Non-invasiveness • Weak coupling interface
• Intercellular communication • Low sensitivity
3D Scaffold • Spatial integration • Complicated fabrication
• Multisite recording • Low spatial resolution of multisite monitoring
• Long-term stability
Conformal • Programmable 3D shapes • Extracellular detection
• Multisite recording • Low sensitivity
• Long-term stability
Implantable • Spatial integration • Difficult to achieve precise positioning of
• Multisite in vivo recording recording sites

• Negligible mechanical constraints


• Long-term stability

A comprehensive performance excellence that covers vari- Keywords


ous metrics/aspects (e.g., high selectivity, high sensitivity, high
signal-to-noise ratio, long-term stability, high density, and etc.) bioelectronics, electrophysiology recording, nanoprobes, nanostructures,
structural designs
remains a long-term goal pursued in the development of bio-
electronics. Additionally, the ability to simultaneously monitor a
Received: June 26, 2023
large number of cells over long time is essential for the biological Revised: July 21, 2023
and pathophysiological studies, but quite a few CMOS-based de- Published online:
vices could achieve simultaneous, high-fidelity, high-throughput
and large-scale intracellular recordings. Opportunities exist in
the combination of cell nanoelectronics with CMOS integrated
circuits, which cannot only improve the signal quality, but also
provide a feasible strategy for building a large-scale, parallel mon- [1] M. P. Viana, J. Chen, T. A. Knijnenburg, R. Vasan, C. Yan, J. E. Arakaki,
itoring platform. M. Bailey, B. Berry, A. Borensztejn, E. M. Brown, S. Carlson, J.
To explore the diversity and complexity of life activities from A. Cass, B. Chaudhuri, K. R. Cordes Metzler, M. E. Coston, Z. J.
cells and tissues to organisms, 3D bioelectronic devices are pow- Crabtree, S. Davidson, C. M. DeLizo, S. Dhaka, S. Q. Dinh, T. P. Do,
erful tools for multiplexed signal collection and multi-modality J. Domingus, R. M. Donovan-Maiye, A. J. Ferrante, T. J. Foster, C.
studies. Multifunctional nanostructured bioelectronics that can L. Frick, G. Fujioka, M. A. Fuqua, J. L. Gehring, K. A. Gerbin, et al.,
achieve electrical/optical/chemical monitoring/stimulation as Nature 2023, 613, 345.
well as cell screening and intracellular delivery/extraction also [2] K. J. Yu, D. Kuzum, S.-W. Hwang, B. H. Kim, H. Juul, N. H. Kim,
represent a promising area for further explorations. The devel- S. M. Won, K. Chiang, M. Trumpis, A. G. Richardson, H. Cheng, H.
Fang, M. Thompson, H. Bink, D. Talos, K. J. Seo, H. N. Lee, S.-K.
opment of bioelectric interface technologies could promote the
Kang, J.-H. Kim, J. Y. Lee, Y. Huang, F. E. Jensen, M. A. Dichter, T. H.
emerging of advanced nanofabrication technologies and power- Lucas, J. Viventi, B. Litt, J. A. Rogers, Nat. Mater. 2016, 15, 782.
ful studying platforms, which are crucial to potential biomedi- [3] E. Song, C.-H. Chiang, R. Li, X. Jin, J. Zhao, M. Hill, Y. Xia, L. Li, Y.
cal applications. The bioelectronics could also provide support of Huang, S. M. Won, K. J. Yu, X. Sheng, H. Fang, M. A. Alam, Y. Huang,
fundamental breakthroughs in life science. J. Viventi, J.-K. Chang, J. A. Rogers, Proc. Natl. Acad. Sci. USA 2019,
116, 15398.
Acknowledgements [4] D. Zhao, R. Huang, J.-M. Gan, Q.-D. Shen, ACS Nano 2022, 16,
19892.
Z.X. acknowledges support from the National Natural Science Foundation [5] H. Fang, K. J. Yu, C. Gloschat, Z. Yang, E. Song, C.-H. Chiang, J. Zhao,
of China (grant No. 12272023) and the Fundamental Research Funds for S. M. Won, S. Xu, M. Trumpis, Y. Zhong, S. W. Han, Y. Xue, D. Xu, S.
the Central Universities. J.Z. acknowledges support from the National Nat- W. Choi, G. Cauwenberghs, M. Kay, Y. Huang, J. Viventi, I. R. Efimov,
ural Science Foundation of China (grant No. 12202233) and China Post- J. A. Rogers, Nat. Biomed. Eng. 2017, 1, 0038.
doctoral Science Foundation (grant No. 2022M711832). [6] E. Song, J. Li, S. M. Won, W. Bai, J. A. Rogers, Nat. Mater. 2020, 19,
590.
Conflict of Interest [7] R. Chen, A. Canales, P. Anikeeva, Nat. Rev. Mater. 2017, 2, 16093.
[8] Y. Jiang, B. Tian, Nat. Rev. Mater. 2018, 3, 473.
The authors declare no conflict of interest. [9] A. Zhang, C. M. Lieber, Chem. Rev. 2016, 116, 215.

Adv. Mater. Interfaces 2023, 2300550 2300550 (12 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

[10] Y. Park, C. K. Franz, H. Ryu, H. Luan, K. Y. Cotton, J. U. Kim, T. S. [37] P. Fromherz, A. Offenhäusser, T. Vetter, J. Weis, Science 1991, 252,
Chung, S. Zhao, A. Vazquez-Guardado, D. S. Yang, K. Li, R. Avila, J. 1290.
K. Phillips, M. J. Quezada, H. Jang, S. S. Kwak, S. M. Won, K. Kwon, [38] H. Gao, F. Yang, K. Sattari, X. Du, T. Fu, S. Fu, X. Liu, J. Lin, Y. Sun,
H. Jeong, A. J. Bandodkar, M. Han, H. Zhao, G. R. Osher, H. Wang, J. Yao, Sci. Adv. 2022, 8, eabn2485.
K. Lee, Y. Zhang, Y. Huang, J. D. Finan, J. A. Rogers, Sci. Adv. 2021, [39] D. A. Soscia, D. Lam, A. C. Tooker, H. A. Enright, M. Triplett, P.
7, eabf9153. Karande, S. K. G. Peters, A. P. Sales, E. K. Wheeler, N. O. Fischer,
[11] Y. Park, T. S. Chung, G. Lee, J. A. Rogers, Chem. Rev. 2022, 122, 5277. Lab Chip 2020, 20, 901.
[12] E. Menard, M. A. Meitl, Y. Sun, J.-U. Park, D. J.-L. Shir, Y.-S. Nam, S. [40] Y. Qin, X.-B. Hu, W.-T. Fan, J. Yan, S.-B. Cheng, Y.-L. Liu, W.-H. Huang,
Jeon, J. A. Rogers, Chem. Rev. 2007, 107, 1117. Adv. Sci. 2021, 8, 2003738.
[13] L. Wang, P. Cai, J. Luo, F. Zhang, J. Liu, Y. Chen, Z. Zhu, Y. Song, B. [41] F. A. Pennacchio, L. D. Garma, L. Matino, F. Santoro, J. Mater. Chem.
Yang, X. Liu, X. Chen, S. Wang, Nano Res. 2018, 11, 5704. B 2018, 6, 7096.
[14] B. Hu, W. R. Leow, P. Cai, Y.-Q. Li, Y.-L. Wu, X. Chen, ACS Nano 2017, [42] Y. Gu, C. Wang, N. Kim, J. Zhang, T. M. Wang, J. Stowe, R. Nasiri,
11, 12302. J. Li, D. Zhang, A. Yang, L. H.-H. Hsu, X. Dai, J. Mu, Z. Liu, M. Lin,
[15] C. Y. Tay, P. Cai, M. I. Setyawati, W. Fang, L. P. Tan, C. H. L. Hong, X. W. Li, C. Wang, H. Gong, Y. Chen, Y. Lei, H. Hu, Y. Li, L. Zhang, Z.
Chen, D. T. Leong, Nano Lett. 2014, 14, 83. Huang, X. Zhang, S. Ahadian, P. Banik, L. Zhang, X. Jiang, P. J. Burke,
[16] K.-I. Jang, H. N. Jung, J. W. Lee, S. Xu, Y. H. Liu, Y. Ma, J.-W. Jeong, Y. et al., Nat. Nanotechnol. 2022, 17, 292.
M. Song, J. Kim, B. H. Kim, A. Banks, J. W. Kwak, Y. Yang, D. Shi, Z. [43] Q. Huang, B. Tang, J. C. Romero, Y. Yang, S. K. Elsayed, G. Pahapale,
Wei, X. Feng, U. Paik, Y. Huang, R. Ghaffari, J. A. Rogers, Adv. Funct. T.-J. Lee, I. E. M. Pantoja, F. Han, C. Berlinicke, T. Xiang, M. Solazzo,
Mater. 2016, 26, 7281. T. Hartung, Z. Qin, B. S. Caffo, L. Smirnova, D. H. Gracias, Sci. Adv.
[17] J.-W. Jeong, W.-H. Yeo, A. Akhtar, J. J. S. Norton, Y.-J. Kwack, S. Li, 2022, 8, eabq5031.
S.-Y. Jung, Y. Su, W. Lee, J. Xia, H. Cheng, Y. Huang, W.-S. Choi, T. [44] A. Kalmykov, C. Huang, J. Bliley, D. Shiwarski, J. Tashman, A.
Bretl, J. A. Rogers, Adv. Mater. 2013, 25, 6839. Abdullah, S. K. Rastogi, S. Shukla, E. Mataev, A. W. Feinberg, K. J.
[18] S. M. Won, E. Song, J. Zhao, J. Li, J. Rivnay, J. A. Rogers, Adv. Mater. Hsia, T. Cohen-Karni, Sci. Adv. 2019, 5, eaax0729.
2018, 30, 1800534. [45] P. L. Floch, Q. Li, Z. Lin, S. Zhao, R. Liu, K. Tasnim, H. Jiang, J. Liu,
[19] A. Mariano, C. Lubrano, U. Bruno, C. Ausilio, N. B. Dinger, F. Adv. Mater. 2022, 34, 2106829.
Santoro, Chem. Rev. 2022, 122, 4552. [46] X. Yang, T. Zhou, T. J. Zwang, G. Hong, Y. Zhao, R. D. Viveros, T.-M.
[20] X. Gao, Y. Jiang, Y. Lin, K.-H. Kim, Y. Fang, J. Yi, L. Meng, H.-C. Lee, Fu, T. Gao, C. M. Lieber, Nat. Mater. 2019, 18, 510.
Z. Lu, O. Leddy, R. Zhang, Q. Tu, W. Feng, V. Nair, P. J. Griffin, F. Shi, [47] L. Gao, J. Wang, Y. Zhao, H. Li, M. Liu, J. Ding, H. Tian, S. Guan, Y.
G. S. Shekhawat, A. R. Dinner, H.-G. Park, B. Tian, Sci. Adv. 2020, 6, Fang, Adv. Mater. 2022, 34, 2107343.
eaay2760. [48] C. Xie, J. Liu, T.-M. Fu, X. Dai, W. Zhou, C. M. Lieber, Nat. Mater.
[21] G. Hong, C. M. Lieber, Nat. Rev. Neurosci. 2019, 20, 330. 2015, 14, 1286.
[22] Y. Zhu, Z. Sun, Z. Yan, Z. Jin, J. M. Tour, ACS Nano 2011, 5, [49] X. Dai, W. Zhou, T. Gao, J. Liu, C. M. Lieber, Nat. Nanotechnol. 2016,
6472. 11, 776.
[23] Y. H. Cho, Y.-G. Park, S. Kim, J.-U. Park, Adv. Mater. 2021, 33, [50] G. Zheng, X. Chen, C. A. Mirkin, Small 2009, 5, 2537.
2005805. [51] S. Yin, Y.-L. Wu, B. Hu, Y. Wang, P. Cai, C. K. Tan, D. Qi, L. Zheng, W.
[24] Z. Jahed, Y. Yang, C.-T. Tsai, E. P. Foster, A. F. McGuire, H. Yang, A. R. Leow, N. S. Tan, S. Wang, X. Chen, Adv. Mater. Interfaces 2014, 1,
Liu, C. Forro, Z. Yan, X. Jiang, M.-T. Zhao, W. Zhang, X. Li, T. Li, A. 1300043.
Pawlosky, J. C. Wu, B. Cui, Nat. Commun. 2022, 13, 2253. [52] Y.-S. Guan, F. Ershad, Z. Rao, Z. Ke, E. C. da Costa, Q. Xiang, Y. Lu,
[25] J. M. Lee, D. Lin, G. Hong, K.-H. Kim, H.-G. Park, C. M. Lieber, Nano X. Wang, J. Mei, P. Vanderslice, C. Hochman-Mendez, C. Yu, Nat.
Lett. 2022, 22, 4552. Electron. 2022, 5, 881.
[26] J. Fang, S. Huang, F. Liu, G. He, X. Li, X. Huang, H.-j. Chen, X. Xie, [53] S.-W. Hwang, C. H. Lee, H. Cheng, J.-W. Jeong, S.-K. Kang, J.-H. Kim,
Nano-Micro Lett. 2022, 14, 125. J. Shin, J. Yang, Z. Liu, G. A. Ameer, Y. Huang, J. A. Rogers, Nano Lett.
[27] D. Xu, J. Mo, X. Xie, N. Hu, Nano-Micro Lett. 2021, 13, 127. 2015, 15, 2801.
[28] H. Shim, F. Ershad, S. Patel, Y. Zhang, B. Wang, Z. Chen, T. J. Marks, [54] Y. Huang, H. Li, T. Hu, J. Li, C. K. Yiu, J. Zhou, J. Li, X. Huang, K. Yao,
A. Facchetti, C. Yu, Nat. Electron. 2022, 5, 660. X. Qiu, Y. Zhou, D. Li, B. Zhang, R. Shi, Y. Liu, T. H. Wong, M. Wu,
[29] H. Shim, S. Jang, A. Thukral, S. Jeong, H. Jo, B. Kan, S. Patel, G. H. Jia, Z. Gao, Z. Zhang, J. He, M. Zheng, E. Song, L. Wang, C. Xu,
Wei, W. Lan, H.-J. Kim, C. Yu, Proc. Natl. Acad. Sci. USA 2022, 119, X. Yu, Nano Lett. 2022, 22, 5944.
2204852119. [55] D.-H. Kim, J.-H. Ahn, W. M. Choi, H.-S. Kim, T.-H. Kim, J. Song, Y. Y.
[30] H. Fang, S. Chuang, T. C. Chang, K. Takei, T. Takahashi, A. Javey, Huang, Z. Liu, C. Lu, J. A. Rogers, Science 2008, 320, 507.
Nano Lett. 2012, 12, 3788. [56] J. Zhao, W. Li, X. Guo, H. Wang, J. A. Rogers, Y. Huang, MRS Bull.
[31] H. Ko, K. Takei, R. Kapadia, S. Chuang, H. Fang, P. W. Leu, K. 2020, 46, 107.
Ganapathi, E. Plis, H. S. Kim, S.-Y. Chen, M. Madsen, A. C. Ford, [57] B. Tian, T. Cohen-Karni, Q. Qing, X. Duan, P. Xie, C. M. Lieber, Sci-
Y.-L. Chueh, S. Krishna, S. Salahuddin, A. Javey, Nature 2010, 468, ence 2010, 329, 830.
286. [58] Y. Zhao, S. S. You, A. Zhang, J.-H. Lee, J. Huang, C. M. Lieber, Nat.
[32] M. Tosun, S. Chuang, H. Fang, A. B. Sachid, M. Hettick, Y. Lin, Y. Nanotechnol. 2019, 14, 783.
Zeng, A. Javey, ACS Nano 2014, 8, 4948. [59] Z. Liu, J. Yan, H. Ma, T. Hu, J. Wang, Y. Shi, J. Xu, K. Chen, L. Yu,
[33] L. Li, L. Shao, X. Liu, A. Gao, H. Wang, B. Zheng, G. Hou, K. Shehzad, Nano Lett. 2021, 21, 2773.
L. Yu, F. Miao, Y. Shi, Y. Xu, X. Wang, Nat. Nanotechnol. 2020, 15, [60] Z. Xue, M. Sun, T. Dong, Z. Tang, Y. Zhao, J. Wang, X. Wei, L. Yu, Q.
743. Chen, J. Xu, Y. Shi, K. Chen, P. Roca i Cabarrocas, Nano Lett. 2017,
[34] X. Song, T. Zhang, L. Wu, R. Hu, W. Qian, Z. Liu, J. Wang, Y. Shi, J. 17, 7638.
Xu, K. Chen, L. Yu, Adv. Sci. 2022, 9, 2105623. [61] Z. Xue, W. Chen, X. Meng, J. Xu, Y. Shi, K. Chen, L. Yu, P. Roca i
[35] S.-K. Lee, H. Y. Jang, S. Jang, E. Choi, B. H. Hong, J. Lee, S. Park, Cabarrocas, Appl. Surf. Sci. 2022, 593, 153435.
J.-H. Ahn, Nano Lett. 2012, 12, 3472. [62] M. Dipalo, V. Caprettini, G. Bruno, F. Caliendo, L. D. Garma, G.
[36] M. Xu, J. Wang, Z. Xue, J. Wang, P. Feng, L. Yu, J. Xu, Y. Shi, K. Chen, Melle, M. Dukhinova, V. Siciliano, F. Santoro, F. De Angelis, Adv.
P. Roca i Cabarrocas, Nanoscale 2017, 9, 10350. Biosyst. 2019, 3, 1900148.

Adv. Mater. Interfaces 2023, 2300550 2300550 (13 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

[63] R. Liu, J. Lee, Y. Tchoe, D. Pre, A. M. Bourhis, A. D’Antonio- [92] K. Kampasi, D. F. English, J. Seymour, E. Stark, S. McKenzie, M.
Chronowska, G. Robin, S. H. Lee, Y. G. Ro, R. Vatsyayan, K. J. Vöröslakos, G. Buzsáki, K. D. Wise, E. Yoon, Microsyst. Nanoeng.
Tonsfeldt, L. A. Hossain, M. L. Phipps, J. Yoo, J. Nogan, J. S. 2018, 4, 10.
Martinez, K. A. Frazer, A. G. Bang, S. A. Dayeh, Adv. Funct. Mater. [93] K. Kim, M. Vöröslakos, J. P. Seymour, K. D. Wise, G. Buzsáki, E.
2022, 32, 2108378. Yoon, Nat. Commun. 2020, 11, 2063.
[64] J. T. Robinson, M. Jorgolli, A. K. Shalek, M.-H. Yoon, R. S. Gertner, [94] X. Jiao, Y. Wang, Q. Qing, Nano Lett. 2017, 17, 7315.
H. Park, Nat. Nanotechnol. 2012, 7, 180. [95] T.-M. Fu, X. Duan, Z. Jiang, X. Dai, P. Xie, Z. Cheng, C. M. Lieber,
[65] C. Chiappini, ACS Sens. 2017, 2, 1086. Proc. Natl. Acad. Sci. USA 2014, 111, 1259.
[66] J.-A. Huang, V. Caprettini, Y. Zhao, G. Melle, N. Maccaferri, L. [96] R. Gao, S. Strehle, B. Tian, T. Cohen-Karni, P. Xie, X. Duan, Q. Qing,
Deleye, X. Zambrana-Puyalto, M. Ardini, F. Tantussi, M. Dipalo, F. C. M. Lieber, Nano Lett. 2012, 12, 3329.
De Angelis, Nano Lett. 2019, 19, 722. [97] A. D. Reyes, Nature 2019, 575, 38.
[67] B. X. E. Desbiolles, E. de Coulon, A. Bertsch, S. Rohr, P. Renaud, [98] B. Tian, P. Xie, T. J. Kempa, D. C. Bell, C. M. Lieber, Nat. Nanotechnol.
Nano Lett. 2019, 19, 6173. 2009, 4, 824.
[68] S. J. Kim, K. W. Cho, H. R. Cho, L. Wang, S. Y. Park, S. E. Lee, T. [99] Q. Qing, Z. Jiang, L. Xu, R. Gao, L. Mai, C. M. Lieber, Nat. Nanotech-
Hyeon, N. Lu, S. H. Choi, D.-H. Kim, Adv. Funct. Mater. 2016, 26, nol. 2014, 9, 142.
3207. [100] L. Xu, Z. Jiang, Q. Qing, L. Mai, Q. Zhang, C. M. Lieber, Nano Lett.
[69] S. J. Kim, H. R. Cho, K. W. Cho, S. Qiao, J. S. Rhim, M. Soh, T. Kim, 2013, 13, 746.
M. K. Choi, C. Choi, I. Park, N. S. Hwang, T. Hyeon, S. H. Choi, N. [101] Y. Zhao, J. Yao, L. Xu, M. N. Mankin, Y. Zhu, H. Wu, L. Mai, Q. Zhang,
Lu, D.-H. Kim, ACS Nano 2015, 9, 2677. C. M. Lieber, Nano Lett. 2016, 16, 2644.
[70] J.-H. Ahn, H.-S. Kim, K. J. Lee, S. Jeon, S. J. Kang, Y. Sun, R. G. Nuzzo, [102] Y. Fang, B. Tian, Nat. Nanotechnol. 2019, 14, 733.
J. A. Rogers, Science 2006, 314, 1754. [103] X. Zhang, Matter 2019, 1, 560.
[71] K. S. Kim, Y. Zhao, H. Jang, S. Y. Lee, J. M. Kim, K. S. Kim, J.-H. Ahn, [104] A. Zhang, Y. Zhao, S. S. You, C. M. Lieber, Nano Today 2020, 31,
P. Kim, J.-Y. Choi, B. H. Hong, Nature 2009, 457, 706. 100821.
[72] K. W. Cho, S. J. Kim, J. Kim, S. Y. Song, W. H. Lee, L. Wang, M. [105] W. Kang, F. Yavari, M. Minary-Jolandan, J. P. Giraldo-Vela, A. Safi,
Soh, N. Lu, T. Hyeon, B.-S. Kim, D.-H. Kim, Nat. Commun. 2019, 10, R. L. McNaughton, V. Parpoil, H. D. Espinosa, Nano Lett. 2013, 13,
4824. 2448.
[73] H. Amin, M. Dipalo, F. De Angelis, L. Berdondini, ACS Appl. Mater. [106] O. Loh, R. Lam, M. Chen, N. Moldovan, H. Huang, D. Ho, H. D.
Interfaces 2018, 10, 15207. Espinosa, Small 2009, 5, 1667.
[74] X. Duan, T. M. Fu, J. Liu, C. M. Lieber, Nano Today 2013, 8, 351. [107] B. X. E. Desbiolles, M. T. M. Hannebelle, E. de Coulon, A. Bertsch,
[75] A. Noy, Adv. Mater. 2011, 23, 807. S. Rohr, G. E. Fantner, P. Renaud, Nano Lett. 2020, 20, 4520.
[76] M. Kwak, L. Han, J. J. Chen, R. Fan, Small 2015, 11, 5600. [108] R. Singhal, Z. Orynbayeva, R. V. K. Sundaram, J. J. Niu, S.
[77] C. Jia, Z. Lin, Y. Huang, X. Duan, Chem. Rev. 2019, 119, 9074. Bhattacharyya, E. A. Vitol, M. G. Schrlau, E. S. Papazoglou, G.
[78] B. Tian, C. M. Lieber, Chem. Rev. 2019, 119, 9136. Friedman, Y. Gogotsi, Nat. Nanotechnol. 2011, 6, 57.
[79] Z. Xue, M. Xu, X. Li, J. Wang, X. Jiang, X. Wei, L. Yu, Q. Chen, J. Wang, [109] J. Abbott, T. Ye, D. Ham, H. Park, Acc. Chem. Res. 2018, 51, 600.
J. Xu, Y. Shi, K. Chen, P. Roca i Cabarrocas, Adv. Funct. Mater. 2016, [110] M. Dipalo, A. F. McGuire, H.-Y. Lou, V. Caprettini, G. Melle, G.
26, 5352. Bruno, C. Lubrano, L. Matino, X. Li, F. De Angelis, B. Cui, F. Santoro,
[80] Z. Xue, M. Xu, Y. Zhao, J. Wang, X. Jiang, L. Yu, J. Wang, J. Xu, Y. Shi, Nano Lett. 2018, 18, 6100.
K. Chen, P. Roca i Cabarrocas, Nat. Commun. 2016, 7, 12836. [111] J. Abbott, T. Ye, K. Krenek, R. S. Gertner, S. Ban, Y. Kim, L. Qin, W.
[81] H. Seong, S. G. Higgins, J. Penders, J. P. K. Armstrong, S. W. Wu, H. Park, D. Ham, Nat. Biomed. Eng. 2019, 4, 232.
Crowder, A. C. Moore, J. E. Sero, M. Becce, M. M. Stevens, ACS [112] C. Xie, Z. Lin, L. Hanson, Y. Cui, B. Cui, Nat. Nanotechnol. 2012, 7,
Nano 2020, 14, 5371. 185.
[82] T. Yasui, T. Yanagida, T. Shimada, K. Otsuka, M. Takeuchi, K. [113] Y. Tchoe, J. Lee, R. Liu, A. M. Bourhis, R. Vatsyayan, K. J. Tonsfeldt,
Nagashima, S. Rahong, T. Naito, D. Takeshita, A. Yonese, R. S. A. Dayeh, Appl. Phys. Rev. 2021, 8, 041317.
Magofuku, Z. Zhu, N. Kaji, M. Kanai, T. Kawai, Y. Baba, ACS Nano [114] J. Yoo, H. Kwak, J. Kwon, G. E. Ha, E. H. Lee, S. Song, J. Na, H.-J.
2019, 13, 2262. Lee, J. Lee, A. Hwangbo, E. Cha, Y. Chae, E. Cheong, H.-J. Choi, Sci.
[83] G. L. Saux, N. Bar-Hanin, A. Edri, U. Hadad, A. Porgador, M. Rep. 2020, 10, 4279.
Schvartzman, Adv. Mater. 2019, 31, 1805954. [115] R. Parameswaran, J. L. Carvalho-de-Souza, Y. Jiang, M. J. Burke, J. F.
[84] W. Kim, J. K. Ng, M. E. Kunitake, B. R. Conklin, P. Yang, J. Am. Chem. Zimmerman, K. Koehler, A. W. Phillips, J. Yi, E. J. Adams, F. Bezanilla,
Soc. 2007, 129, 7228. B. Tian, Nat. Nanotechnol. 2018, 13, 260.
[85] Y. Kubota, H. Oi, H. Sawahata, A. Goryu, Y. Ando, R. Numano, M. [116] M. Dipalo, H. Amin, L. Lovato, F. Moia, V. Caprettini, G. C. Messina,
Ishida, T. Kawano, Small 2016, 12, 2846. F. Tantussi, L. Berdondini, F. De Angelis, Nano Lett. 2017, 17,
[86] X. Li, J. Mo, J. Fang, D. Xu, C. Yang, M. Zhang, H. Li, X. Xie, N. Hu, 3932.
F. Liu, J. Mater. Chem. B 2020, 8, 7609. [117] Y. Liu, J. Wang, X. Huang, Y. Zhao, Y. Sun, S. Zhang, H. Wang, L.
[87] X. Xie, A. M. Xu, M. R. Angle, N. Tayebi, P. Verma, N. A. Melosh, Yuan, H. Chen, ACS Appl. Mater. Interfaces 2019, 11, 46515.
Nano Lett. 2013, 13, 6002. [118] Z. Liu, J. Nie, B. Miao, J. Li, Y. Cui, S. Wang, X. Zhang, G. Zhao, Y.
[88] X. Zhu, M. F. Yuen, L. Yan, Z. Zhang, F. Ai, Y. Yang, P. K. N. Yu, G. Deng, Y. Wu, Z. Li, L. Li, Z. L. Wang, Adv. Mater. 2019, 31, 1807795.
Zhu, W. Zhang, X. Chen, Adv. Healthcare Mater. 2016, 5, 1157. [119] G. C. Messina, M. Dipalo, R. La Rocca, P. Zilio, V. Caprettini, R.
[89] A. Zhang, J. Fang, X. Li, J. Wang, M. Chen, H.-j. Chen, G. He, X. Xie, Proietti Zaccaria, A. Toma, F. Tantussi, L. Berdondini, F. De Angelis,
Nanoscale Adv. 2022, 4, 1844. Adv. Mater. 2015, 27, 7145.
[90] C. Chiappini, Y. Chen, S. Aslanoglou, A. Mariano, V. Mollo, H. Mu, [120] X. Xie, A. M. Xu, S. Leal-Ortiz, Y. Cao, C. C. Garner, N. A. Melosh,
E. De Rosa, G. He, E. Tasciotti, X. Xie, F. Santoro, W. Zhao, N. H. ACS Nano 2013, 7, 4351.
Voelcker, R. Elnathan, Nat. Protoc. 2021, 16, 4539. [121] E. Peer, A. Artzy-Schnirman, L. Gepstein, U. Sivan, ACS Nano 2012,
[91] D. Li, N. Wang, T. Zhang, G. Wu, Y. Xiong, Q. Du, Y. Tian, W. 6, 4940.
Zhao, J. Ye, S. Gu, Y. Lu, D. Jiang, F. Xu, Adv. Photonics 2022, 4, [122] J. J. VanDersarl, A. M. Xu, N. A. Melosh, Nano Lett. 2012, 12,
016001. 3881.

Adv. Mater. Interfaces 2023, 2300550 2300550 (14 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

[123] Z. C. Lin, C. Xie, Y. Osakada, Y. Cui, B. Cui, Nat. Commun. 2014, 5, [158] S. Inal, A. Hama, M. Ferro, C. Pitsalidis, J. Oziat, D. Iandolo, A.-M.
3206. Pappa, M. Hadida, M. Huerta, D. Marchat, P. Mailley, R. M. Owens,
[124] J. Gao, C. Liao, S. Liu, T. Xia, G. Jiang, J. Nanobiotechnol. 2021, 19, Adv. Biosyst. 2017, 1, 1700052.
97. [159] C. Angulo-Pineda, K. Srirussamee, P. Palma, V. M. Fuenzalida, S. H.
[125] X. Wang, M. Li, Assay Drug Dev. Technol. 2003, 1, 695. Cartmell, H. Palza, Nanomaterials 2020, 10, 428.
[126] V. I. Chubinskiy-Nadezhdin, A. V. Sudarikova, M. A. Shorokhova, V. [160] X. Li, M. Mao, K. Han, C. Yao, B. Gu, J. He, D. Li, Prog. Mater Sci.
Y. Vasileva, Z. M. Khairullina, Y. A. Negulyaev, Biochem. Biophys. Res. 2023, 133, 101041.
Commun. 2022, 619, 22. [161] Z. Fan, K.-C. Hwang, J. A. Rogers, Y. Huang, Y. Zhang, J. Mech. Phys.
[127] A. Spanu, L. Martines, A. Bonfiglio, Lab Chip 2021, 21, 795. Solids 2018, 111, 215.
[128] S. Kim, B. Keisham, V. Berry, Mater. Today Nano 2020, 9, 100063. [162] Y. Shuai, J. Zhao, R. Bo, Y. Lan, Z. Lv, Y. Zhang, J. Mech. Phys. Solids
[129] J. Yoon, J. Lim, M. Shin, T. Lee, J.-W. Choi, Appl. Phys. Rev. 2023, 10, 2023, 173, 105203.
011302. [163] J. Zhao, F. Zhang, X. Guo, Y. Huang, Y. Zhang, H. Wang, J. Mech.
[130] N. Schaefer, R. Garcia-Cortadella, J. Martínez-Aguilar, G. Schwesig, Phys. Solids 2021, 155, 104568.
X. Illa, A. Moya Lara, S. Santiago, C. Hébert, G. Guirado, R. Villa, A. [164] H. Fu, K. Nan, W. Bai, W. Huang, K. Bai, L. Lu, C. Zhou, Y. Liu, F.
Sirota, A. Guimerà-Brunet, J. A. Garrido, 2D Mater. 2020, 7, 025046. Liu, J. Wang, M. Han, Z. Yan, H. Luan, Y. Zhang, Y. Zhang, J. Zhao,
[131] H.-G. Jahnke, A. Mewes, F. D. Zitzmann, S. Schmidt, R. Azendorf, X. Cheng, M. Li, J. W. Lee, Y. Liu, D. Fang, X. Li, Y. Huang, Y. Zhang,
A. A. Robitzki, Sci. Rep. 2019, 9, 13875. J. A. Rogers, Nat. Mater. 2018, 17, 268.
[132] J. Yao, X. A. Liu, K. D. Gillis, Anal. Methods 2015, 7, 5760. [165] W. Pang, X. Cheng, H. Zhao, X. Guo, Z. Ji, G. Li, Y. Liang, Z. Xue, H.
[133] Z. Zhang, T. Zheng, R. Zhu, Microsyst. Nanoeng. 2020, 6, 81. Song, F. Zhang, Z. Xu, L. Sang, W. Huang, T. Li, Y. Zhang, Natl. Sci.
[134] S. Xie, Y. Lu, S. Zhang, L. Wang, X. Zhang, Small 2010, 6, 1897. Rev. 2020, 7, 342.
[135] J. J. VanDersarl, P. Renaud, Sci. Rep. 2016, 6, 32485. [166] Y. Zhang, F. Zhang, Z. Yan, Q. Ma, X. Li, Y. Huang, J. A. Rogers, Nat.
[136] Y. Xiang, H. Liu, W. Yang, Z. Xu, Y. Wu, Z. Tang, Z. Zhu, Z. Zeng, D. Rev. Mater. 2017, 2, 17029.
Wang, T. Wang, N. Hu, D. Zhang, Microsyst. Nanoeng. 2022, 8, 70. [167] Y. Zhang, Z. Yan, K. Nan, D. Xiao, Y. Liu, H. Luan, H. Fu, X. Wang, Q.
[137] M. Voelker, P. Fromherz, Small 2005, 1, 206. Yang, J. Wang, W. Ren, H. Si, F. Liu, L. Yang, H. Li, J. Wang, X. Guo,
[138] F. Patolsky, B. P. Timko, G. Yu, Y. Fang, A. B. Greytak, G. Zheng, C. H. Luo, L. Wang, Y. Huang, J. A. Rogers, Proc. Natl. Acad. Sci. USA
M. Lieber, Science 2006, 313, 1100. 2015, 112, 11757.
[139] L. H. Hess, M. Jansen, V. Maybeck, M. V. Hauf, M. Seifert, M. [168] S. Xu, Z. Yan, K.-I. Jang, W. Huang, H. Fu, J. Kim, Z. Wei, M. Flavin,
Stutzmann, I. D. Sharp, A. Offenhäusser, J. A. Garrido, Adv. Mater. J. McCracken, R. Wang, A. Badea, Y. Liu, D. Xiao, G. Zhou, J. Lee, H.
2011, 23, 5045. U. Chung, H. Cheng, W. Ren, A. Banks, X. Li, U. Paik, R. G. Nuzzo,
[140] J. Müller, M. Ballini, P. Livi, Y. Chen, M. Radivojevic, A. Shadmani, V. Y. Huang, Y. Zhang, J. A. Rogers, Science 2015, 347, 154.
Viswam, I. L. Jones, M. Fiscella, R. Diggelmann, A. Stettler, U. Frey, [169] Z. Xue, T. Jin, S. Xu, K. Bai, Q. He, F. Zhang, X. Cheng, Z. Ji, W. Pang,
D. J. Bakkum, A. Hierlemann, Lab Chip 2015, 15, 2767. Z. Shen, H. Song, Y. Shuai, Y. Zhang, Sci. Adv. 2022, 8, eabm6922.
[141] M. Voelker, P. Fromherz, Phys. Rev. Lett. 2006, 96, 228102. [170] X. Cheng, Z. Fan, S. Yao, T. Jin, Z. Lv, Y. Lan, R. Bo, Y. Chen, F.
[142] K.-I. Chen, B.-R. Li, Y.-T. Chen, Nano Today 2011, 6, 131. Zhang, Z. Shen, H. Wan, Y. Huang, Y. Zhang, Science 2023, 379,
[143] Q. Qing, S. K. Pal, B. Tian, X. Duan, B. P. Timko, T. Cohen-Karni, V. 1225.
N. Murthy, C. M. Lieber, Proc. Natl. Acad. Sci. USA 2010, 107, 1882. [171] J. Zhao, J. Appl. Mech. 2022, 89, 041008.
[144] Y. Cui, Q. Wei, H. Park, C. M. Lieber, Science 2001, 293, 1289. [172] R. Feiner, L. Engel, S. Fleischer, M. Malki, I. Gal, A. Shapira, Y.
[145] Y. Huang, H. G. Sudibya, D. Fu, R. Xue, X. Dong, L.-J. Li, P. Chen, Shacham-Diamand, T. Dvir, Nat. Mater. 2016, 15, 679.
Biosens. Bioelectron. 2009, 24, 2716. [173] H. Li, H. Liu, M. Sun, Y. Huang, L. Xu, Adv. Mater. 2021, 33,
[146] J. Kim, G. Park, S. Lee, S.-W. Hwang, N. Min, K.-M. Lee, Biosens. 2004425.
Bioelectron. 2017, 90, 238. [174] J. Cools, Q. Jin, E. Yoon, D. Alba Burbano, Z. Luo, D. Cuypers,
[147] S. Liu, X. Guo, NPG Asia Mater 2012, 4, 23. G. Callewaert, D. Braeken, D. H. Gracias, Adv. Sci. 2018, 5,
[148] D. S. Roman, R. Garg, T. Cohen-Karni, APL Mater. 2020, 8, 100906. 1700731.
[149] D. Sarkar, W. Liu, X. Xie, A. C. Anselmo, S. Mitragotri, K. Banerjee, [175] S. Kim, Y. S. Oh, K. Lee, S. Kim, W.-Y. Maeng, K. S. Kim, G.-B. Kim,
ACS Nano 2014, 8, 3992. S. Cho, H. Han, H. Park, M. Wang, R. Avila, Z. Xie, K. Ko, J. Choi, M.
[150] T. Cohen-Karni, Q. Qing, Q. Li, Y. Fang, C. M. Lieber, Nano Lett. 2010, Je, H. Lee, S. Lee, J. Koo, I. Park, Small 2023, 19, 2206839.
10, 1098. [176] Y. Zhang, N. Zheng, Y. Cao, F. Wang, P. Wang, Y. Ma, B. Lu, G. Hou,
[151] Q. Li, K. Nan, P. Le Floch, Z. Lin, H. Sheng, T. S. Blum, J. Liu, Nano Z. Fang, Z. Liang, M. Yue, Y. Li, Y. Chen, J. Fu, J. Wu, T. Xie, X. Feng,
Lett. 2019, 19, 5781. Sci. Adv. 2019, 5, eaaw1066.
[152] B. Tian, J. Liu, T. Dvir, L. Jin, J. H. Tsui, Q. Qing, Z. Suo, R. Langer, [177] W. Xi, C. K. Schmidt, S. Sanchez, D. H. Gracias, R. E. Carazo-Salas,
D. S. Kohane, C. M. Lieber, Nat. Mater. 2012, 11, 986. S. P. Jackson, O. G. Schmidt, Nano Lett. 2014, 14, 4197.
[153] J. Wang, H. Wang, X. Mo, H. Wang, Adv. Mater. 2020, 32, 2004555. [178] Y. Mei, A. A. Solovev, S. Sanchez, O. G. Schmidt, Chem. Soc. Rev.
[154] D. Karnaushenko, N. Münzenrieder, D. D. Karnaushenko, B. Koch, 2011, 40, 2109.
A. K. Meyer, S. Baunack, L. Petti, G. Tröster, D. Makarov, O. G. [179] Y. Mei, G. Huang, A. A. Solovev, E. B. Ureña, I. Mönch, F. Ding, T.
Schmidt, Adv. Mater. 2015, 27, 6797. Reindl, R. K. Y. Fu, P. K. Chu, O. G. Schmidt, Adv. Mater. 2008, 20,
[155] Y. Park, T. S. Chung, J. A. Rogers, Curr. Opin. Biotechnol. 2021, 72, 1. 4085.
[156] Z. Yan, M. Han, Y. Shi, A. Badea, Y. Yang, A. Kulkarni, E. Hanson, [180] E. J. Smith, S. Schulze, S. Kiravittaya, Y. Mei, S. Sanchez, O. G.
M. E. Kandel, X. Wen, F. Zhang, Y. Luo, Q. Lin, H. Zhang, X. Guo, Y. Schmidt, Nano Lett. 2011, 11, 4037.
Huang, K. Nan, S. Jia, A. W. Oraham, M. B. Mevis, J. Lim, X. Guo, [181] P. Fattahi, G. Yang, G. Kim, M. R. Abidian, Adv. Mater. 2014, 26, 1846.
M. Gao, W. Ryu, K. J. Yu, B. G. Nicolau, A. Petronico, S. S. Rubakhin, [182] W. M. Grill, S. E. Norman, R. V. Bellamkonda, Annu. Rev. Biomed.
J. Lou, P. M. Ajayan, K. Thornton, et al., Proc. Natl. Acad. Sci. USA Eng. 2009, 11, 1.
2017, 114, E9455. [183] G. G. Naples, J. T. Mortimer, A. Scheiner, J. D. Sweeney, IEEE Trans.
[157] X. Liu, M. N. George, S. Park, A. L. Miller Ii, B. Gaihre, L. Li, B. E. Biomed. Eng. 1988, 35, 905.
Waletzki, A. Terzic, M. J. Yaszemski, L. Lu, Acta Biomater. 2020, 111, [184] D. J. Tyler, D. M. Durand, IEEE Trans. Neural Syst. Rehabil. Eng. 2002,
129. 10, 294.

Adv. Mater. Interfaces 2023, 2300550 2300550 (15 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH
21967350, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/admi.202300550 by CochraneItalia, Wiley Online Library on [21/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advmatinterfaces.de

[185] M. W. Sang, E. Song, J. T. Reeder, J. A. Rogers, Cell 2020, 181, 115. [191] R. Dong, L. Wang, C. Hang, Z. Chen, X. Liu, L. Zhong, J. Qi, Y. Huang,
[186] J. Li, E. Song, C.-H. Chiang, K. J. Yu, J. Koo, H. Du, Y. Zhong, S. Liu, L. Wang, Y. Lu, X. Jiang, Small 2021, 17, 2006612.
M. Hill, C. Wang, J. Zhang, Y. Chen, L. Tian, Y. Zhong, G. [192] H.-P. Phan, Y. Zhong, T.-K. Nguyen, Y. Park, T. Dinh, E. Song, R. K.
Fang, J. Viventi, J. A. Rogers, Proc. Natl. Acad. Sci. USA 2018, Vadivelu, M. K. Masud, J. Li, M. J. A. Shiddiky, D. Dao, Y. Yamauchi,
115, E9542. J. A. Rogers, N.-T. Nguyen, ACS Nano 2019, 13, 11572.
[187] Z. Zhao, H. Zhu, X. Li, L. Sun, F. He, J. E. Chung, D. F. Liu, L. Frank, [193] D. M. Cox-Pridmore, F. A. Castro, S. R. P. Silva, P. Camelliti, Y. Zhao,
L. Luan, C. Xie, Nat. Biomed. Eng. 2022, 7, 520. Small 2022, 18, 2105281.
[188] A. Natalia, L. Zhang, N. R. Sundah, Y. Zhang, H. Shao, Nat. Rev. [194] C. Y. Tay, S. A. Irvine, F. Y. C. Boey, L. P. Tan, S. Venkatraman, Small
Bioeng. 2023, 1, 481. 2011, 7, 1361.
[189] H. A. Ledesma, X. Li, J. L. Carvalho-de-Souza, W. Wei, F. Bezanilla, [195] A. Ashok, T.-K. Nguyen, M. Barton, M. Leitch, M. K. Masud, H. Park,
B. Tian, Nat. Nanotechnol. 2019, 14, 645. T.-A. Truong, Y. V. Kaneti, H. T. Ta, X. Li, K. Liang, T. N. Do, C.-
[190] J. A. Frank, M.-J. Antonini, P. Anikeeva, Nat. Biotechnol. 2019, 37, H. Wang, N.-T. Nguyen, Y. Yamauchi, H.-P. Phan, Small 2023, 19,
1013. 2204946.

Zhaoguo Xue is an associate professor at Beihang University. He received his Ph.D. degree from Nan-
jing University in 2018, and worked as a postdoctoral fellow at Tsinghua University from 2018 to 2020.
His research interests include mechanically-guided 3D assembly, stretchable and flexible nanoelec-
tronics, and semiconductor nanomaterials.

Jianzhong Zhao received his Ph.D. degree from Shanghai University in 2021. He is now a postdoctoral
fellow at Tsinghua University. His research interests focus on mechanically-guided 3D assembly and
mechanics of flexible electronics.

Adv. Mater. Interfaces 2023, 2300550 2300550 (16 of 16) © 2023 The Authors. Advanced Materials Interfaces published by Wiley-VCH GmbH

You might also like