ArticlePDF AvailableLiterature Review

Biomimetic cell culture for cell adhesive propagation for tissue engineering strategies

Authors:

Abstract

Biomimetic cell culture, which involves creating a biomimetic microenvironment for cells in vitro by engineering approaches, has aroused increasing interest given that it maintains the normal cellular phenotype, genotype and functions displayed in vivo. Therefore, it can provide a more precise platform for disease modelling, drug development and regenerative medicine than the conventional plate cell culture. In this review, initially, we discuss the principle of biomimetic cell culture in terms of the spatial microenvironment, chemical microenvironment, and physical microenvironment. Then, the main strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a biomimetic microenvironment for cells, a variety of strategies has been developed, ranging from conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to emerging scaffold-free strategies, such as spheroids, organoids, and assembloids, to simulate the native cellular microenvironment. Recently, 3D bioprinting and microfluidic chip technology have been applied as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this area are discussed and future directions are discussed to shed some light on the community.
Materials
Horizons
rsc.li/materials-horizons
REVIEW ARTICLE
Zhaoying Wu, Lin Xiao et al .
Biomimetic cell culture for cell adhesive propagation for
tissue engineering strategies
ISSN 2051-6347
Volume 10
Number 11
November 2023
Pages 4637–5316
4662 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
Cite this: Mater. Horiz., 2023,
10, 4662
Biomimetic cell culture for cell adhesive
propagation for tissue engineering strategies
Qiuchen Luo,
a
Keyuan Shang,
a
Jing Zhu,
a
Zhaoying Wu, *
a
Tiefeng Cao,
b
Abeer Ahmed Qaed Ahmed,
c
Chixiang Huang
a
and Lin Xiao *
a
Biomimetic cell culture, which involves creating a biomimetic microenvironment for cells in vitro by
engineering approaches, has aroused increasing interest given that it maintains the normal cellular
phenotype, genotype and functions displayed in vivo. Therefore, it can provide a more precise platform
for disease modelling, drug development and regenerative medicine than the conventional plate cell
culture. In this review, initially, we discuss the principle of biomimetic cell culture in terms of the spatial
microenvironment, chemical microenvironment, and physical microenvironment. Then, the main
strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a
biomimetic microenvironment for cells, a variety of strategies has been developed, ranging from
conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to
emerging scaffold-free strategies, such as spheroids, organoids, and assembloids, to simulate the native
cellular microenvironment. Recently, 3D bioprinting and microfluidic chip technology have been applied
as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this
area are discussed and future directions are discussed to shed some light on the community.
Wider impact
Biomimetic cell culture, which is committed to creating a biomimetic microenvironment for cells in vitro by engineering approaches, has aroused increasing
interest. In this review, initially we discuss the principle of biomimetic cell culture in terms of the spatial, chemical, and physical microenvironments. Then, the
main strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a biomimetic microenvironment for cells, a variety of
strategies has been developed ranging from conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to emerging scaffold-
free strategies, such as spheroids, organoids, and assembloids, to simulate the native cellular microenvironment. Recently, 3D bioprinting and microfluidic
chip technology have been applied as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this area are discussed,
and future directions outlooked. To the best of our knowledge, this is the first review discussing material design and manufacture from the perspective of
biomimetic cell culture, which is a new concept or new way of thinking. It will shed some light for the materials science community, particularly concerning
biological, therapeutic and tissue engineering materials.
1. Introduction
Biomimetic cell culture involves creating a biomimetic micro-
environment for cells by engineering approaches in vitro, where
cells can grow, proliferate, differentiate, communicate, and
maintain their metabolic activities and functions as in vivo.
Thus, biomimetic culture of cells has great potential in
applications such as disease modelling, drug development
and screening, and stem cell research.
1
Initially, cell cultures
are performed on two-dimensional (2D) planes,
2
such as Petri
dishes and porous plates, on which cells are grown against
the wall. However, traditional cell 2D culture methods cannot
simulate the in vivo growth environment of cells, leading to a
distorted cellular morphology and function and limitations in
the formation of tissue-specific structures.
1,3
In addition, live animal experiments are often required after
in vitro studies with 2D cultured cells. Living animal models
can provide in vivo conditions, which are deemed more reliable
than 2D cell culture systems. However, animal experiments are
also limited due to intrinsic species differences, ethical issues,
and high costs. Recently, it was reported that animal tests are
a
School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University,
Shenzhen 518107, China. E-mail: xiaolin23@mail.sysu.edu.cn,
wuzhy37@mail.sysu.edu.cn
b
Department of Gynaecology, First Affiliated Hospital of Sun Yat-Sen University,
Guangzhou 510070, China
c
Department of Molecular Medicine, Biochemistry Unit, University of Pavia,
27100 Pavia, Italy
Received 3rd June 2023,
Accepted 29th August 2023
DOI: 10.1039/d3mh00849e
rsc.li/materials-horizons
Materials
Horizons
REVIEW
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
View Journal
| View Issue
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4663
no longer required before human drug trials by the FDA.
4
This
calls for more advanced and precise in vitro models, in which
achieving biomimetic cell culture is a fundamental issue.
Accordingly, tissue and organ engineering based on biomi-
metic cell culture may provide reliable in vitro models.
2. The principle of biomimetic cell
culture
The extracellular matrix (ECM) is a non-cellular three-
dimensional (3D) macromolecular network outside of cells.
It provides structural support, regulates and facilitates cell–cell
and cell–tissue signaling and controls the morphology, structure
and function of cells.
5
The three main components of the ECM
are fibrin, glycoproteins and proteoglycans, which regulate the
biochemical and mechanical properties, provide sufficient cell–
ECM interactions and mediate cell–cell interactions.
6,7
Therefore,
the key to achieving biomimetic cell culture is to recapitulate the
ECM in vitro to provide a simulated growth environment for cells
as in vivo.
8
In general, the cellular microenvironment in vivo can be
approximately divided into three aspects, i.e.,thespatialmicro-
environment, chemical microenvironment, and physical microen-
vironment (Fig. 1). The spatial microenvironment refers to the 3D
spatial niches where cells live in terms of the spatial geometry,
composition and spatial distribution of cells and ECM. In addi-
tion, cells in vivo grow in a 3D space with multiple chemical
signals, including growth factors, nutritional factors and signaling
factors, and various physical signals, such as mechanical cues of
the ECM.
9
Therefore, the basic principle of biomimetic cell culture
lies in the simulation of the spatial, chemical, and physical
microenvironments in vitro for cells.
2.1 Spatial microenvironment simulation
The 3D spatial microenvironment is crucial for cells to maintain
their normal morphology, metabolism, function, and communi-
cation.
10
In terms of spatial geometry, the construction of porous
scaffolds is a regular approach to simulate the spatial charac-
teristics of cells for biomimetic culture. The 3D spatial structures
of porous scaffolds, including porosity, pore size and orientation,
have a remarkable impact on the fate of cells. Porous materials
with a high specific surface area will increase the adsorption of
proteins, thus promoting cell adhesion.
11
In addition, a porous
surface improves the mechanical interlock between the implant
biomaterials and surrounding tissues, such as natural bone,
providing greater mechanical stability at this critical interface.
Studies have shown that a porosity of more than 50% and an
interconnected pore size of more than 100 mm are the minimum
requirements for cell growth and migration.
12
He et al. prepared
a porous scaffold with a pore size of ca. 167 mm, which was
suitable for osteoblasts to infiltrate the scaffold and facilitate the
transport of nutrients and metabolic waste.
13
In bone tissue
engineering, biomaterials with smaller pore sizes are beneficial
for the formation of anoxic conditions and induce osteochon-
drogenesis before osteogenesis, while biomaterials with larger
pore sizes lead to vascularization and direct osteogenesis with-
out forming previous cartilage.
14
In addition, the orientation
of pores is closely related to the direction of cell migration. For
example, Jiang et al. prepared a radial porous nanocomposite
chitosan (CS)/HA scaffold using HA nanoparticles from porcine
cortical bone and CS by freeze-casting technology (Fig. 2a).
15
The
scaffold exhibited excellent biological functions and enabled
cells to attach and migrate along the pore channels in vitro.In
addition, Zhang et al. reported that oriented pores are more
conducive to cell ingrowth and interaction and more efficient
nutrient transport and ECM deposition than scaffolds with a
random orientation.
16
The spatial microenvironment also involves the interaction
between cells and the ECM. The components and microstruc-
ture of the ECM provide an elaborate 3D niche for cells to carry
out physiological activities and largely affect cell behaviors.
17
For example, collagen and its derivatives are widely applied as
biomimetic ECM in cartilage tissue engineering.
18
In addition,
the distribution of cells also features the spatial microenviron-
ment. This may only be a single layer of cells with cell–cell
interactions in the horizontal direction in conventional 2D cell
culture, while the distribution of cells is multilayered with
all-directional cell–cell interactions in biomimetic cell culture.
For example, in cellular spheroids, the distribution of cells is
compact with all-directional cell–cell interactions, which is
conducive to maintaining cell activity and function.
19
In addition, the spatial microenvironment in vivo is dynamic
in response to various chemical and physical cues. To achieve a
dynamic spatial microenvironment, intelligent constructs
whose properties change over time based on 3D cell culture
systems were developed, also known as four-dimensional (4D)
cell culture.
20
They respond to different stimuli, including
electric field, magnetic field, temperature, humidity, pH and
light.
21,22
Miao et al. prepared an intelligent 4D scaffold with
shape-memory characteristics (Fig. 2b).
23
Its topography can be
changed dynamically from microwell arrays to aligned patterns
over time by changing the environmental temperature. The
results showed that the microwell arrays promoted the for-
mation of neural stem cell (NSC) aggregates and early differ-
entiation, while the aligned patterns were conducive to neurite
extension and elongation.
23
Some researchers also designed
Fig. 1 Typical schematic of the cellular microenvironment.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4664 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
chemically modular microgels for logical degradation to achieve
customizable void ratios in 3D-printed scaffolds at specific
times.
24
2.2 Chemical microenvironment simulation
Cells are in a microenvironment with a large number of
biochemical factors. Due to the presence of these factors, cells
can normally grow, proliferate and perform a series of physio-
logical functions. For example, mesenchymal stem cells (MSCs)
require a series of growth factors, including transforming
growth factor-b(TGF-b), hepatocyte growth factor (HGF),
interleukin-1 (IL-1) and interleukin-6 (IL-6),
25
to achieve func-
tions such as fixation, migration, proliferation, and differentia-
tion. Additionally, the communication between cells includes
bidirectional transmission of various biochemical factors. Also,
the chemical signals in the intracellular environment play a
certain role in regulating the growth and differentiation of cells.
Therefore, including these chemical signals is an important
aspect of biomimetic cell culture. For example, Li et al.
constructed scaffolds that can release TGF-b3 in an orderly
manner, which is conducive to the migration, proliferation,
and fibrocartilage differentiation of cells in vitro in the culture
system, and also remodelling the fibrous matrix into a fibrous
cartilage matrix.
26
In another study, Lee et al. developed a dual
cryogel system consisting of a gelatin/CS cryogel surrounded by
a gelatin/heparin cryogel for the dual delivery of vascular
endothelial growth factor (VEGF) and bone morphogenetic
protein-4 (BMP-4) (Fig. 2c).
27
The release of these two factors
enhances the osteogenic differentiation of adipose-derived
stem cells (ADSCs).
The chemical microenvironment of cells in vivo also includes
the biochemical properties of ECM components, which have
remarkable impacts on cell adhesion, growth, proliferation, and
migration. Based on this, the biochemical properties of bio-
materials in contact with cells are also closely related to the
growth of cells in vitro. For instance, dopamine (DA) was
partially covalently grafted onto a hyaluronic acid–chondroitin
sulfate composite gel to enhance the matrix stability in the cell
culture medium. DA, as a neurotransmitter, plays a critical role
in axonal outgrowth and stimulates the remodelling properties
of gels by capturing cell-secreted laminin and binding brain-
derived neurotrophic factor.
29
This study implemented the
interaction between cells and biological scaffold materials,
and ultimately successfully simulated the formation of neuro-
nal networks. Additionally, RGD peptides are often used to
modify biomaterials to increase their cell adhesion ability.
30
Fig. 2 (a) Schematic diagram of the structure and application of nanocomposite CS/HA scaffolds. Radial porous scaffolds can cause cells to go deep
into the scaffolds along the pore channel (curved arrows), while preventing unrelated cells from invading the scaffold. Axial porous scaffolds allow cells to
migrate from the bottom pore channel (curved arrows) and allow fibroblasts to invade the scaffolds. Reproduced with permission.
15
Copyright 2022,
Wiley-VCH. (b) Schematic diagram of the preparation of new intelligent 4D scaffolds. (I) Fused deposition modelling (FDM) printing molds. (II) Coating
molds with polydimethylsiloxane (PDMS) to solidify to obtain PDMS microgrooves. (III) Extruding 4D printed materials into PDMS microgrooves for
solidification. (IV) Imprinted polymethyl methacrylate microwell arrays by stereolithography (STL). (V) Imprinted 4D scaffolds from microwell molds at
60 1C for 10 min. (VI) Microwell array fabrication. (VII) Cell seeding scaffolds. (VIII) Time-dependent scaffold materials. Reproduced with permission.
23
Copyright 2020, Wiley-VCH. (c) Research diagram of the double cryogel system. Reproduced with permission.
27
Copyright 2020, Elsevier B.V.
(d) Photosensitive hypertensive metamaterial scaffolds for dynamic cell culture systems. (i) Dynamic culture system consists of an elastic styrene
ethylene–butylene–styrene framework and photoactive surface, allowing the response to light and mechanical stimulation. (ii) Dynamic regulationof
cells by dynamic culture systems under different external stimulations. Reproduced with permission.
28
Copyright 2022, the American Chemical Society.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4665
In addition, to achieve a biomimetic culture of cells in vitro,
it is also necessary to have an adequate nutrient supply
and meet the growth conditions of cells, such as an appro-
priate oxygen concentration and soluble nutrient molecule
concentration.
2.3 Physical microenvironment simulation
The cellular microenvironment involves various physical signals
mediated by the ECM and interstitial fluid, which in turn affect
the fate of cells. These physical signals include not only mechan-
ical effects produced by cell–cell interactions but also mechanical
cues produced by physical factors such as the morphological
structure of materials, mechanical properties of the ECM, internal
stress, and external forces in the cellular microenvironment.
Essential physical signals, such as the appropriate mechan-
ical stimulation and mechanical properties of materials, are
crucial in the biomimetic culture of cells.
31
Zhang et al. applied
cyclic compression to MSCs, inducing them to produce func-
tional osteocyte bone organoids.
32
It was found that cyclic
pressure enhanced the differentiation of osteoblasts and orga-
noid mineral density and stiffness. Nguyen et al. used aldehyde
hyaluronic acid to oxidize gelatin microcarriers to produce a
smoother morphology and stronger stiffness.
33
The resultant
microcarriers had a higher affinity for corneal stromal cells and
were conducive to their attachment and growth.
Mechanical signals from internal stress and external stimu-
lation are essential for many cells and tissues in vivo, such as
chondrocytes and articular cartilage. Physical signals generated
by proper mechanical loading (such as exercise) can induce
homeostatic regulation in vivo to achieve reorganization and
maintenance of the cartilage ECM.
34
This inspired researchers
to optimize the in vitro culture of chondrocytes (or precursor
cells) and cartilage tissue engineering by introducing appro-
priate mechanical stimulation. It has been verified that
mechanical stimulation can trigger several responses in vitro,
such as increased secretion of osteopontin and other ECM
proteins.
35
For example, Paggi et al. applied multidirectional
compression to hydrogels containing chondrocytes by a
cartilage-on-chip device.
34
It was found that the glycosamino-
glycans secreted by the cells significantly increased after this
mechanical stimulation. Additionally, the expression of specific
cartilage ECM markers, such as aggrecan, collagen II, and
collagen VI, increased. Table 1 presents a summary of the
typical mechanical stimulation compatible with different cells.
In addition to mechanical signals, other physical signals,
such as light and electrical stimulation, have also been applied
to cell culture in vitro. Chen et al. constructed a dynamic cell
culture system capable of responding to mechanical and light
stimulation (Fig. 2d).
28
The flexibility and tensile elasticity of
the dynamic culture system enabled it to serve as a dynamic
matrix for activating cell activity. Light stimulation was applied
to produce mild heat stress, which together with mechanical
stimulation could effectively improve the activity of stem cells
and their ability to differentiate into cardiomyocytes. In
another study, appropriate electrical stimulation was used to
control the cell morphology and phenotype in biomimetic
cultures of human-induced stem cell-derived cardiomyocytes
(hPSC-CMs).
43
Electrical stimulation was also employed in the
biomimetic culture of chondrocytes, which has broad poten-
tial for exploring mechanotransduction in cartilage tissues.
44
Recently, researchers developed electroactive/conducting
hydrogels and scaffolds.
45
Electroactive materials can not only
promote cell activity but also simulate the electrophysiological
microenvironment of natural electrically excitable cells through
their own electroactive properties and transmit biochemical
signals.
46,47
For example, Li et al. prepared electroactive fibrous
scaffolds by incorporating barium titanate nanoparticles. The
dielectric permittivity of the scaffolds is comparable to that of
natural bone, which enhanced the osteogenic differentiation of
MSCs.
48
Malki et al. reported the preparation of an electroactive
Table 1 Application of mechanical stimulation in biomimetic cell culture
Cell type Mechanical stimulation Effectiveness Application Ref.
Preadipocyte 930 kPa culture substrate rigidity; 12% static
stretch
Higher cell metabolism and mitochondrial
activity
Acceleration of adipocyte
differentiation
36
Osteocyte-like
mouse cell
Unidirectional shear stress of 0.03 Pa Enhancement of cancer cell invasion with
mechanical stimulation
Construction of in vitro
prediction model of bone
metastasis
37
MSC Cyclic hydrostatic pressure with 10–300 kPa
pressure and 2 Hz frequency
Promotion of collagen synthesis and
mineral deposition
Promotion of osteogenic
differentiation
38
0.07 N preload, 1% strain, 5 Hz, 5 min per
loading time, 5 times per week
Enhancement of organoid mineral density,
stiffness, and osteoblast differentiation
Construction of functional
osteocyte bone organoids
32
Nucleus pulpo-
sus derived
stem cell
Hypoxic environment; chamber pressure
1.0 MPa
Exhibition of stronger resistance to
overloading
Treatment of disc degen-
eration and low back pain
39
Chondrocyte Cyclic tensile strain at 0.33 Hz, 0–10% strain Inhibition of inflammatory response Research of signal pathway 40
compression and shear strain (300 mbar
positive pressure, 350 mbar negative
pressure, 0.33 Hz)
Promotion of the expression of glycosami-
noglycans and specific cartilage ECM
markers
Cartilage tissues basic
research and drug testing
34
Mammary
tumor cell
1 N force at 5 Hz for 3 min Inhibition of osteoclastogenesis and the
levels of tumorigenic genes; promotion of
osteogenesis
Reduction of breast cancer-
associated bone metastasis
41
Heart-derived
ventricular cell
2% elongation (strain) at 0.25 Hz Reduction of apoptosis and inflammatory
factors
A new target for the
treatment of heart disease
42
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4666 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
hydrogel of chondroitin sulphate containing stromal cell-
derived factor 1 (SDF-1). The release of SDF-1 could be con-
trolled by externally applied electrical stimulation to promote
cell migration and infiltration.
49
3. The main strategies of biomimetic
cell culture
Many strategies have been developed to create a biomimetic
microenvironment for cells in vitro. Conventional scaffolds can
provide support and create a spatial microenvironment,
chemical microenvironment, and physical microenvironment
for cells to some extent.
50
However, the cellular microenviron-
ment constructed by this method is relatively simple, with
limited capacities for cell arrangement and nutrient
supplementation.
51
Scaffold-free strategies, such as spheroids,
organoids, and assembloids, have enriched the cell types and
arrangements and enabled the formation of complex cellular
aggregate structures through the self-assembly of cells, which
mimic the process of natural tissue and organ formation.
Recently, fast-growing 3D bioprinting and microfluidic chip
technology provided potent platforms for integrating emerging
biomimetic culture strategies, creating more complicated and
precise biomimetic constructs.
52
Beyond the regular use of
individual cells, cellular aggregates such as spheroids, orga-
noids and assembloids can be applied in 3D bioprinting and
microfluidic chips.
3.1 Conventional scaffold strategies
The simplest way to realize biomimetic culture in vitro is to seed
cells on scaffolds. In this case, a variety of materials (natural or
synthetic) in different states (gel or solid) can be used as
scaffolds. Porous scaffolds with appropriate porosities and pore
sizes can provide an ideal 3D space for cell attachment
and migration. With specific physical and chemical signals by
surface modification, mechanical adjustment or chemical/
biological functionalization of the scaffolds, the cells in the
scaffolds can grow, proliferate, and differentiate to form tissue-
like structures.
53
3.1.1 Solid scaffolds. Metals, ceramics, polymers and other
materials have been used to fabricate solid scaffolds for bio-
mimetic cell culture.
54
Metals such as titanium (Ti)
55,56
and
magnesium (Mg)
57
and ceramics such as hydroxyapatite (HA),
bioactive glass and b-tricalcium phosphate (b-TCP) are com-
monly used for porous scaffolds. As an example, Zhang et al.
developed a biodegradable, bioactive, and elastic calcium
phosphide (CaP) ceramic sponge with high porosity (approxi-
mately 99%).
58
It was self-assembled by HA nanowires and
b-TCP nanofibers into a seamless interwoven network, which
was conducive for cell attachment, proliferation, and osteogenic
differentiation. Recently, many techniques, such as non-solvent-
induced phase separation, thermally induced phase separation,
foaming, electrospinning, magnetic levitation, self-assembly,
extrusion-based cell entrapment and bioprinting, have been
developed for the fabrication of scaffolds.
59
The topological
structure (porosity and pore size), chemical (biocompatibility
and degradability) and physical (mechanical and electroactive
properties) characteristics of the materials are essential for the
construction of the spatial, chemical and physical microenviron-
ment. The porous structure and mechanical properties of scaf-
folds can affect the morphology, adhesion, growth, maturation,
and metabolism of cells.
60,61
However, there are some limitations
in the solid scaffold approach, especially when using large scaf-
folds. For example, cells cannot migrate into the inner pores of
scaffolds and only adhere to the shallow surface. Even if there are
cells in the inner pores, their activity will be reduced due
to insufficient nutrient supplementation and signal exchange.
Moreover, a suitable degradation rate of scaffold materials that
matches the cell recovery rate is required for most applications of
scaffold materials. It is expected that with the degradation of
scaffold materials, cells undergo proliferation and differentiation
and form tissue structures with biological functions. However,
this is currently a major challenge in the development of scaffold
materials. This has caused numerous failures in scaffold material
applications due to the unmatchable material degradation and
cell recovery. The rapid degradation of scaffold materials may lead
to inadequate cell development and poor treatment efficacy, while
delayed material degradation may cause inflammation.
62
Addi-
tionally, the degradation products of the scaffold materials should
be non-cytotoxic and cause minimal fibrosis and foreign body
reactions, which further increase the difficulties in material
development.
63
Compared with macroscopic solid scaffolds, microcarriers
based on porous microspheres have significant advantages in
cell loading capacity and nutrient supplementation because of
their microscopic size and high surface area to mass ratio.
64–66
Microcarriers are micron-level porous systems, with a particle
size in the range of 100–300 mm.
67
Microcarriers with appro-
priate porous structures allow cells to attach, grow, and migrate
within the internal areas and facilitate the diffusion of nutri-
ents, oxygen and metabolites. For example, Huang et al. pre-
pared porous chitosan microcarriers with interconnected pores
of a suitable size using a method of emulsion-based thermally
induced phase separation. By using these microcarriers, 3D
culture of L-02 hepatocytes was achieved, where cells grew and
migrated in the internal pores and formed multidirectional
cell–cell interactions.
68
In addition, microcarriers can protect
cells from shear damage when incubated in an agitating
suspension in bioreactors.
69
3.1.2 Hydrogels. Hydrogels are a type of 3D network
formed by embedding hydrophilic polymer chains in a water-
rich environment.
70
Hydrogels are promising materials for
biomimetic cell culture because of their porous network struc-
ture, high water content (470% water) and good biocompat-
ibility, which are similar to the natural ECM.
6
For example, a
curdlan/polyvinyl alcohol composite hydrogel had an appropri-
ate pore size and porosity to simulate the spatial microenviron-
ment and could maintain cell activity when mouse fibroblasts
were cultured inside.
71
Hydrogels have tailorable viscoelastic
and stiffness properties depending on their composition
and crosslinking mechanisms, including physical crosslinking
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4667
(ionic, H-bonding, or hydrophobic) and chemical crosslinking
(covalent bonds between macromolecular chains).
72–74
The
viscoelastic nature of hydrogels endow them with the ability
to resist mechanical deformation. In particular, dynamic cross-
linked hydrogels will relax and deform under stress to promote
cell diffusion, proliferation and differentiation.
75,76
In addition,
the structure of hydrogels can include a variety of biochemical
signal molecules and allow their diffusion and release in a
preset manner, which simulates the biochemical features of
the cellular microenvironment. Hydrogels can also deliver
external physical stimuli to cells to further shape the cellular
microenvironment.
However, similar to solid scaffolds, large hydrogels also have
limitations in cell arrangement and nutrient supplementation.
Alternatively, microgels have attracted increasing attention
for biomimetic cell culture.
77
The surface wettability, hydro-
phobicity, charge, and chemical properties of microgels
can affect cell attachment, proliferation, migration, and self-
organization.
78,79
Microgels are also used to fabricate macro-
porous hydrogels by assembly. Rommel et al. reported a 3D
soft macroporous hydrogel scaffold by the assembly of two-
component rod-like microgels with a defined size, stiffness,
and reactivity.
80
In this system, rod-like microgels act as
artificial ‘‘blood vessels’’ to deliver oxygen and nutrients, and
thus cells in both the internal and external areas of the
hydrogels can obtain sufficient oxygen and nutrients.
3.2 Scaffold-free strategies
Cellular aggregates such as spheroids, organoids and assem-
bloids have been developed as next-generation strategies of
biomimetic cell culture, in which cells are self-organized into
aggregates without a scaffold support. A variety of methods can
be used to promote the formation of cellular aggregates, such
as using hanging drop microplates and low adhesion plates
with ultralow attachment coatings.
81
Because of the native-like
microtissue structures with abundant cell–cell and cell–ECM
interactions, the cells in these aggregates have a microenviron-
ment closer to that of cells in vivo than cells cultured in
scaffolds.
19
3.2.1 Spheroids. Spheroids are simple clusters of multiple
cell types, such as tumor spheroids,
82
neurospheroids,
83
hepa-
tocyte spheroids
84
and stem cell spheroids.
85
Compared with
2D cell lines, cells in spheroids are much closer to cells in vivo
in terms of cellular morphology, metabolic activity, and
function.
86
In addition to their 3D spatial structure, spheroids
can form a biomimetic chemical microenvironment with
gradients of nutrients, gases, growth factors and signaling
factors.
87
Moreover, mechanical stimulation, such as compres-
sion force, shear force, hydrostatic pressure and osmotic pres-
sure, can be applied to spheroids by adjusting the culture
environment.
88
Spheroids are formed because adherent cells tend to aggre-
gate under low attachment conditions. The main mechanism of
spheroid formation is that cells self-organize to establish cell–
cell contacts in response to external physical stimulation, such
as gravity, magnetic force, and sound waves. For example,
Byun et al. used magnetic force to self-assemble cells to form
spheroids of ADSCs (Fig. 3a).
89
They prepared a magnetized
artificial ECM containing magnetic nanoparticles (MNPs). The
artificial ECM had a stable spatial structure, and ADSCs were
assembled into composite magnetized spheroids, which were
used as building blocks for further 3D tissue engineering. The
ADSC spheroids showed a crucial influence on cell proliferation
and differentiation, and they successfully induced chondrocyte
differentiation. Rasouli and Tabrizian reported a fast spheroid
formation technique based on boundary-driven acoustic waves
to produce dense cellular aggregates (Fig. 3b).
90
In this study,
when solutions were introduced into microfluidic channels and
reached the acoustic region, cells were condensed and com-
pacted into spheroid clusters within seconds by acoustically
induced hydrodynamics. Using this method, biomimetic cocul-
ture spheroids could be obtained from malignant breast tumor
cells (MDA-MB-231) and human breast adenocarcinoma cells
(MCF-7).
Moreover, the effects of chemical signals can also cause cells
to form spheroids. Because the hydrated surface of hydrogels
can inhibit the adsorption of proteins and mediate the inter-
action force between cells, cells will not adhere to the surface of
Fig. 3 (a) Schematic of the construction of composite stem cell spher-
oids. They are constructed from synthetic fibres (MSFs) and complex tissue
structures (rings and laminates) by magnetic assembly. Because MSFs
hinder cell contraction, cell spheroids still exhibit structural stability over
7 days of culture. Reproduced with permission.
89
Copyright 2021, IOP
Publishing. (b) Formation mechanism of acoustic spheroids assembled by
collagen. Cells surrounded by collagen fibres are injected into microfluidic
channels. Driven by sound waves, cells are captured and remodelled into
spheroids. Reproduced with permission.
90
Copyright 2021, Wiley-VCH.
(c) Schematic of spheroid formation and extraction. Reproduced with
permission.
91
Copyright 2022, Science China Press.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4668 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
the hydrogel material, which results in the formation of spher-
oids. Ai et al. developed a supramolecular polypeptide hydrogel
via the self-assembly of biotinylated peptides for biomimetic
cell culture (Fig. 3c).
91
The introduction of a D-amino acid into
the polypeptide may reduce the cell–ECM interaction, and thus
promote the spontaneous formation of spheroids. In another
study, thermo-responsive hydrogels with surface micropore
patterns were developed to culture ADSC spheroids and deliver
them into Matrigel to study their migration and fusion
behaviors.
92
In addition, Im et al. investigated the effect of
seeding density on the formation of spheroids.
93
Studies have
found that the expression levels of angiogenic paracrine factors
and the secretion of cell adhesion molecules in spheroids are
different with various seeding densities of human ADSCs.
Under appropriate seeding density conditions, the spheroids
showed significantly enhanced therapeutic angiogenic capa-
city. Through the above-mentioned physical and chemical
methods, it is possible to control the number of cells and the
size of the spheroids, to produce high-density cell clusters and
increase the ratio of cells to fluid.
3.2.2 Organoids. More than 60 experts have accurately
defined organoids as three-dimensional structures derived
from (pluripotent) stem cells, progenitors, and/or differentiated
cells that self-organize through cell–cell and cell–matrix
interactions to recapitulate aspects of the native tissue archi-
tecture and function in vitro.
97
Organoid technology is one of
the most potent approaches for biomimetic cell culture. Orga-
noids have organ specificity in many aspects, such as cell type,
function, and spatial structure.
98
The cell source, culture
method and ECM material are three decisive factors in the
culture of organoids. Organoid formation requires cells to be
embedded in an appropriate ECM and supplemented with an
appropriate organic medium and a range of growth factors to
promote the self-organization and growth of the cells
(Fig. 4a).
94
With specific growth factors, stem cells proliferate,
differentiate and self-organize into organ-like structures, which
can be passaged and cultured and exhibit a high degree of
genetic stability.
99
A variety of stem cells, including embryonic
stem cells (ESCs), adult stem cells (ASCs) and induced plur-
ipotent stem cells (iPSCs), has been used for the formation of
organoids. To induce the specific development of organoids,
the culture medium is usually supplemented with several
ligands or compounds, such as retinoic acid, BMP, and epider-
mal growth factor (EGF), which can activate pivotal modal
signaling pathways.
95
The physical conditions of the ECM
environment significantly influence the morphology of the
self-organized organoids (Fig. 4b). During the initial stages of
organoid development, the suspension culture conditions
Fig. 4 (a) Schematic of organoid formation and self-organization: diverse types of cells (different colors) sort themselves to form the embryo body over
spatial constraints, eventually generating more differentiated cells and developing organoids. Reproduced with permission.
94
Copyright 2021, Wiley-
VCH. (b) Methods to induce organoid formation: suspension culture, bioreactor and ECM. Reproduced with permission.
95
Copyright 2021, Wiley-VCH.
(c) Methods for the preparation of the 3D structure of human islet organoids. (i) Methods for the construction of the islet organoid 3D structure by cell
self-aggregation. (ii) Mixed hydrogels with human islet lineage cells to generate 3D structures vi a 3D bioprinting or biomaterial embedding. (iii) Methods
to maintain islet organoids, including implantation in 3D printing scaffolds, decellularized pancreatic scaffolds and organs-on-a-chip. Reproduced with
permission.
96
Copyright 2022, Ivyspring International.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4669
facilitate the reaggregation and self-sorting of the cells. After
the cells aggregate into a 3D structure, the use of bioreactors
can improve the perfusion levels of nutrient molecules and
oxygen, prolong the organoid culture time and increase the size
of organoids. In addition, the use of naturally derived ECM
facilitates the real simulation of the cellular environment and
produces organoids closer to the actual situation.
100
The methods used for the construction of human organoids
include traditional self-aggregation (such as the use of ultralow
attachment plates, spinner flasks, and biomaterial-coated
plates), modified self-aggregation (such as suspended droplets
and microporous platforms), and hydrogel embedding,
96
as well
as emerging methods such as 3D bioprinting, decellularized
organ scaffolds, and organ-on-a-chip (Fig. 4c).
96
Using these
methods, many types of organoids have been developed and
applied in human healthcare areas. Table 2 presents a sum-
mary of the representative organoids reported in the last few
years. For instance, Yoshihara et al. used Matrigel to culture islet
organoids in vitro and maintain their physiological functions,
which can be used to explore insulin-dependent diabetes
therapies.
101
In the generation of liver organoids, the selection
of source cells, the type and concentration of growth factors, and
thegrowthprocessoforganoidshavebeenextensivelyexplored
(Fig. 5a). During the construction of human liver organoids, the
activities of cyclic adenosine monophosphate (cAMP) and TGF-b
need to be strictly regulated to successfully expand the cells.
102
Table 2 Representative organoids developed in recent years
Type Source Culture method Results Ref.
Endoderm-
derived
organoid
Liver
organoid
Human fetal liver pro-
genitor cells
Culture in scaffolds made from
liver ECM
Formation of hepatocytes and bile duct
structures at the same time
106
Pluripotent stem cells
(PSCs)
Coculture in plate Long-term expansion with competent liver
functionality
107
Hepatobiliary
tubular
organoid
Hepatocytes,
cholangiocytes
Coculture in plate Reproduction of hepatocyte metabolite
transport in liver tissue
108
Hepatobiliary
organoid
iPSCs, ESCs Coculture on Matrigel-coated
plates
Demonstration of hepatic and biliary
functional attributes
109
Lung
organoid
ESCs Culture on poly (lactide-co-
glycolide) or polycaprolactone
(PCL) scaffolds
Formation of tubular structures similar to
adult airways
110
Bronchial epithelial cells,
lung fibroblasts, smooth
muscle cells
Triple culture in stiffer matrix Formation of airway simulated tubular
morphology
111
Intestinal
organoid
Small intestinal mucosa
cells
Coculture in plate Organoids containing enterocytes, goblet cells,
Paneth cells and entero-endocrine cells
112
Cells from human ileum
and duodenum
Coculture in plate, CRISPR
engineering
Demonstration of widespread germination
and nourish all sorts of cells
113
Mesoderm-
derived
organoid
Kidney
organoid
PSCs Coculture in plate and modula-
tion of Wnt
Vascularized and segmentally patterned
organoids
114
PSCs Extrusion-based cellular
bioprinting
Increasement of throughput, reduction of
organoid size and demonstration of improved
maturation and nephron number
115
PSCs Culture in kidney decellularized
ECM hydrogel
Demonstration of extensive vascular network
and endothelial cells
116
Breast
organoid
Breast epithelial cells
and breast cancer cells
Culture in 3D bioprinting ECM
hydrogel
Production of organoids/tumoroids in a
tissue-specific matrix
117
Trabecular
bone
organoid
Bone marrow mono-
nuclear cells and primary
murine osteoblasts
Coculture on thin slices of
demineralized cortical bone
Reproduction of the bone remodelling cycle 118
Cartilage
organoid
PSCs Suspension culture Successfully bridge of critical size long bone
defects
119
Bone callus
organoid
MSCs Digital light processing (DLP)
printing of cell-loaded hydrogel
microspheres
Maturation and formation of bone-like tissue
in vivo
120
Woven bone
organoid
MSCs 3D self-organizing coculture of
osteoblasts and osteocytes
Formation of network and communicate by
expression of sclerostin
121
Bone marrow
organoid
MSCs Coculture in transglutaminase-
polyethylene glycol/hyaluronic
acid hybrid hydrogels
Functional bone marrow organoids with a
higher level of complexity
122
Ectoderm-
derived
organoid
Sweat gland
organoid
Reprogram human epi-
dermal keratinocytes
Coculture in plate Self-organization of into monoclonal spheres
and organoids
123
Skin organoid iPSCs Coculture in plate Production of hair follicles and nervous system 124
and
125
Cerebral
organoid
iPSCs Coculture in plate Research of human neuronal cells migration 126
iPSCs Culture in spinning bioreactor Similar transcriptome and response to
inflammatory stimulation
127
iPSCs Culture in spinning bioreactor Repeatable realization of cell diversity 128
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4670 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
Bone organoids are 3D self-organizing bone microtissues that can
reflect the complex bone physiological microenvironment and
structure as ideal models in vitro.
103
Bone microenvironment
recapitulation is the theoretical basis of bone organoid formation.
Functionally, the bone microenvironment mainly consists of three
units, i.e., bone formation, bone resorption and hematopoiesis.
There are several pivotal steps in establishing bone organoids
(Fig. 5b). Firstly, stem cells are isolated for differentiation. Then,
for the growth and differentiation of cells, biomaterials are
introduced as a matrix. Finally, some construction methods are
used to construct various organoid models, including woven bone
organoids, bone marrow organoids, cartilage organoids and
trabecular bone organoids.
104
Because of their high biomimetic efficacy from the cellular
level to organ level, organoids have shown great potential in
human disease modelling. Organoids cultured from patient-
derived cells can achieve personalized treatment and perform
drug development, evaluation and screening.
105
Moreover,
organoids with complex 3D structures and specific functions
can be used in tissue engineering and regenerative medicine.
3.2.3 Assembloids. Assembloids were defined by Pas-ca
et al. as self-organizing cellular systems resulting from the
combination of a type of organoid with another type of orga-
noid or with different specialized cell types, resulting in their
integration.
129
However, some researchers may define assem-
bloids as some type of organoid. As an example, Vogt claimed
that assembloids are organoids generated by spatially organiz-
ing multiple cell types.
130
It should be noted that we adopt the
definition of assembloids as a combination of different types of
organoids in this work. Assembloids are more advanced
organoid-based cellular aggregates produced by a variety of cell
types with a spatial organization structure, which can integrate
different types of organoids.
131
Compared with organoids,
assembloids can better achieve the complexity of the micro-
environment in vitro and the specificity of tissue structure,
as well as maintain the stability of cell genetic information.
Fig. 5 (a) Method for the construction of liver organoids and tumors from healthy livers. Isolated cells or tissues are seeded in suitable matrix scaffolds.
After 1–2 weeks of amplification, liver organoids are formed, and liver tumors are formed after 3–4 weeks. Reproduced with permission.
102
Copyright
2021, Elsevier B.V. (b) Schematic diagram of the process for the biofabrication of bone organoids.
104
Copyright 2022, Elsevier B.V.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4671
They provide a new solution to overcome the shortcomings of
organoids, such as their single-cell type, lack of vascular system
and complex interstitial structure.
The successful construction of assembloidsismainlyachieved
by suspension culture, matrix culture, 3D bioprinting or organ-on-
a-chip technology, which are similar to the methods of organoid
construction. Suspension culture is the earliest method, which is
simple and time-consuming. It cannot provide matrix compo-
nents and a vascular system for cells, leading to an unstable
assembly structure and poor permeability in the growth process.
Alternatively, the matrix culture method can provide a matrix that
is rich in growth factors and provides mechanical support and
necessary substances for growth for the self-organization process
of cells. In addition, it can make the assembloid structure more
complete and plays a role in prolonging the cell culture cycle. 3D
bioprinting technology and organ-on-a-chip technology have
shown great potential in a wide area of biofabrication, including
the construction of assembloids, in which chemical and physical
signals can be applied and controlled more accurately. These
emerging technologies enable assembloids to achieve more
complex, complete, and delicate physiological structures,
132
better
compatibility
133
and higher throughput in vitro biomimetic cul-
ture. Further discussion is presented in Section 3.3.
In addition to the necessary physical and chemical stimula-
tions, the methods for the assembly of cells are also crucial for
the formation of assembloids. Different protocells are used in
different assembly methods. The common assembly methods
include spontaneous assembly and directed assembly. Sponta-
neous assembly is a process that mainly involves the sponta-
neous growth of stem cells to achieve assembly, while directed
assembly involves coculturing a variety of highly differentiated
cells in proportion to achieve assembly.
134
At present, different assembloids have been constructed,
such as brain,
135
heart,
136
islet,
137,138
and tumor
139
assem-
bloids. For example, iPSCs were encapsulated in microcapsules
by microfluidic electrospray technology to effectively form
brain region-specific organoids, which were then fused and
assembled into brain assembloids.
140
Brain assembloids can
grow and function well and are composed of cortical, hippo-
campal and thalamic organoids, indicating that they have
significant potential for a wide range of applications in the
neurological and biomedical fields. In addition, iPSCs are used
to generate 3D cortical and striatal organoids, which can be
assembled to form the corticostriatal circuitry of the forebrain
(Fig. 6a).
135
The neural circuit formed by the complete assembly
of the cortex, spinal cord and skeletal muscle can also be
cultured in vitro for up to 10 weeks, and control of muscle
contraction by the cortex can be simulated. The construction of
these models is of great significance for the study of neurode-
velopment and mental illness.
141
For the construction of heart
assembloids, PSCs are seeded on ultralow attachment plates
and cultured under chemical stimulation. Cardiac differentia-
tion and spontaneous assembly were induced to form heart
assembloids (Fig. 6b).
136
3.3 Integration platforms
Existing strategies for biomimetic cell culture have made great
progress in the simulation of spatial, chemical, and physical
microenvironments. However, the structural complexity, func-
tional diversity, and complicated communications at different
levels (cell, tissue, and organ) are still insufficient compared
with that in vivo. With the rapid development of 3D bioprinting
technology and microfluidic chip technology, they may serve as
platforms to improve existing biomimetic cell culture strategies
Fig. 6 (a) Schematic diagram of region-specific neural culture and assembly simulating human brain development in vitro. Reproduced with
permission.
135
Copyright 2023, Springer Nature. (b) Diagram showing the construction of self-organizing heart assembloids. Reproduced with
permission.
136
Copyright 2021, Elsevier B.V.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4672 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
such as microcarriers, spheroids and organoids.
142
3D bioprint-
ing is manufacturing technology for the precise allocation of
biomaterials containing cells to construct complex living tis-
sues or artificial organs with specific functions.
143
It can be
used to print larger-sized complex constructs based on cellular
aggregates. Microfluidic chip systems are potent for culturing
living cells in micron-level chambers under continuous flow
conditions.
144
They can also be used to enable multiple biomi-
metic structures, such as organoids, to communicate with each
other to achieve more complicated biomimetic structures.
These integrative platforms can control the assembly of cellular
aggregates in a larger spatial range and improve the physiolo-
gical relevance of the models.
3.3.1 3D bioprinting technology. As additive manufactur-
ing technology, 3D bioprinting has great application prospects
in the construction of 3D spatial niches for cells in terms of
spatial geometry, compositions and spatial distribution of cells
and ECM. 3D bioprinting can give rise to biomimetic 3D
scaffolds with a uniform distribution of cells or cellular aggre-
gates.
145
There are different bioprinting technologies based on
the basic working principle of manufacturing functional tissue
structures, including inkjet-based bioprinting,
146–148
laser-
assisted bioprinting,
149,150
extrusion-based bioprinting,
151,152
acoustic bioprinting,
153–155
photocuring-based bioprinting,
156–159
and magnetic bioprinting.
160–162
Table 3 presents a summary of
the main applications of these bioprinting technologies in
biomimetic cell culture with a brief description of each techno-
logy. These bioprinting strategies can achieve the requested
additive manufacturing goals and tissue manufacturing via
single or multiple uses, as shown in Fig. 7. As an example,
magnetic 3D bioprinting is used to form cellular aggregates in a
noncontact manner, where the cell medium is exposed to an
external magnetic field after mixing with a paramagnetic
buffer.
160
Nanoparticles can also be used to magnetize cells,
and thus they can be manipulated to suspend from the surface
into the culture medium to form cellular aggregates.
161
Magnetic
3D bioprinting has become an exocrine gland culture platform
with non-heterogeneity, high expansibility and reproduci-
bility.
162
In addition to bioprinting methods, bioink is another
essential part of the bioprinting strategy. Bioinks are blends
of cells and materials used for printing in 3D bioprinting
technology.
163,164
3D bioprinting has been extensively employed
to fabricate various porous scaffolds using bioinks from natural or
synthetic biological materials toward biomimetic cell culture and
tissue engineering.
As discussed above, microtissues, including cell-containing
microspheres and cellular aggregates such as spheroids, orga-
noids and assembloids, are superior to individual cells in terms
of biomimetic culture. Therefore, 3D bioprinting using bioinks
containing microtissues will have significant advantages over
regular 3D bioprinting using bioinks containing individual
cells. In the construction of the spatial microenvironment, it
not only realizes the construction of spatial geometry but also
precisely controls the compositions and spatial distribution of
cells and the matrix. For example, DLP bioprinting technology
was used to print microtissues from chondrocytes and cartilage
acellular matrix to form an auricular shape (Fig. 8a). 3D
bioprinting on a needle array, known as Kenzan bioprinting,
was used to fix spheroids on an array of microneedles and
manufacture tabular and tubular tissue constructs, as shown in
Fig. 8b.
198,199
Heo et al. printed spheroids formed by cocultured
MSCs and HUVECs by aspiration-assisted bioprinting
technology.
196
The printed tissue constructs have high spatial
structure stability and cell activity. Organoids or assembloids
have also been applied in 3D bioprinting (Fig. 8c) to control the
spatial position of cellular aggregates on a larger scale and
construct more complicated tissue structures. This process is
called bioprinting-assisted tissue emergence (BATE).
132
In the
BATE process, organoids or individual cells can be used as 3D
biological printing units, which can be forced to fuse and
recombine according to the preset shapes and sizes by 3D
printing. By utilizing this universal strategy, large-scale
complex cell constructs (such as the gut, Fig. 8d) can be printed
with particular cell types to generate tissue–tissue interactions
by simulating the process of native organ development or
homeostasis.
Additionally, the combination of spheroids and 3D bioprint-
ing technology improves the accuracy of cell regulation, which
simulates the cellular microenvironment more precisely, espe-
cially the chemical microenvironment.
200
To construct human
hypertrophic scars in vitro, Bin et al. developed a new bioink
containing cellular aggregates of fibroblasts, and then prepared
a scar tissue model with a precise arrangement and multiple
functions using bioprinting technology.
201
This model can not
only be used to study the key mechanisms of hypertrophic
scarring but also to rapidly search for drug targets and study
new optimal treatments. In addition, using 3D bioprinting
technology, liver organoids undergo more advanced assembly
and fusion, hepatocyte differentiation, albumin synthesis, liver-
specific enzyme activity, and a highly biomimetic polarization
phenotype.
188
These results prove that the bioprinting of 3D
cellular aggregates can achieve more complex regulation of
biochemical factors than the bioprinting of cells.
In terms of the physical microenvironment, studies have
shown that bioinks containing compacted cellular aggregates
exhibit higher yield stress than slurries of individual cells,
enabling the printing of more mechanically stable
structures.
202
Daly et al. developed a bioprinting method in
which spheroids could move within a shear-thinning hydrogel,
which could self-heal to localize the spheroids.
203
High cell-
density microtissues with specific shapes could be formed
by directed fusion between spheroids within the hydrogel.
By bioprinting this hydrogel containing spheroids composed
of iPSC-derived cardiomyocytes and cardiac fibroblasts, it was
found that the culture system supported cell differentiation,
proliferation, and migration, allowing the growth of multi-
cellular structures in prescribed 3D patterns, successfully
mimicking the global reduction in contractility that occurs
after myocardial infarction.
3.3.2 Microfluidic chip technology. Microfluidics is a tech-
nique for precisely handling small amounts of fluid in the form
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4673
Table 3 Application of 3D bioprinting in biomimetic cell culture
Method Principle Cell type Material Printing structure Results Ref.
Inkjet-based
bioprinting
Division of bioink into
droplets and print layer by
layer
Fibroblasts Alginate, silk fibroin Tubular constructs Long-term cell culture in vitro 166
Fibroblasts Alginate, surfactants Hollow cell-containing
channels
Long-term cultures of larger samples 167
Fibroblasts Sodium alginate Cell-laden microsphere Construction of circulatory system and
reduction of cell sedimentation and
aggregation
168
Corneal epithelial/endothelial
cells
Poly-e-lysine, gellan gum Porous bitmaps Favor of the growth of monolayer cells 169
Type I and II alveolar cells, lung
fibroblasts, lung endothelial cells
Collagen Progressive deposition
of each layer of alveolar
tissue
Construction of the alveolar barrier
models
170
Laser-
assisted
bioprinting
Vaporization of the metal
film depositing on bioink
by laser; ejection of bioink
droplets
Stromal cells from apical papilla/
HUVECs
Collagen I, tricalcium silicate Disc Osteogenic differentiation and
angiogenesis
171
HUVECs N.A. Deposition on
biopaper
Production of vascularized bone
structure
172
MSCs Rat collagen I, nano HA Disc, ring Bone repair 173
ESCs, ADSCs Human laminin, collagen I Layer-by-layer printed
stacking structures
Simulation of corneal tissue structures 174
Extrusion-
based
bioprinting
Extrusion of bioink in the
syringe and cross-link
Fibroblasts GelMA, silicate nanoparticles Hollow cylinders Maximization of printability and
shape fidelity
175
HUVECs/breast cancer cells/
fibroblasts
GelMA, alginate core, sheath
microfibers
Hollow constructs with
curved and straight
channels
Favor of cellular proliferation and
spread
176
Muscle progenitor cells Acellular gelatin hydrogel, PCL Multilayer structure
containing
microchannels
Excellent cell proliferation and
differentiation
177
Dermal papilla cells/
keratinocytes
GelMA/gelatin Filament segments
containing hollow
channels
Construction of the intestinal villi/
human hair follicles structure
178
Small intestinal organoids Collagen I Tube constructs Control of tissue self-organization on
a larger scale
132
Acoustic
bioprinting
Movement of cells in dif-
ferent directions by sound
waves
Fibroblasts GelMA Pyramid structure Construction of tumor microtissue
invasion model
179
MSCs Pluronic F-127, Matrigel Cylinders consisting of
96 droplets per layer
Control of droplet size by changing
acoustic frequency
180
Colorectal organoids GelMA Single, line, cross, and
triangle patterns
Reestablishment of tumor structures
and simulation of tumor invasion
181
Photo-curing
based
bioprinting
Photocuring of bioinks by
light projection
Fibroblasts GelMA Lattice structures Production of steak-type cultured
meat
182
HUVECs CS, acrylamide Nose, ear, lattice
structures
Provision of bioactive sites for cell
growth
183
HUVECs GelMA Lattice structures Excellent biocompatibility for cultur-
ing cells in vitro
184
MSCs GelMA Cylinders Cartilage regeneration 185
MSCs GelMA, LAP, New Coccine Grid structures Excellent shape fidelity, efficient and
low-cost cell culture strategy
186
Microtissue of chondrocytes on
acellular matrix microparticles
GelMA 3D auricular model Accurate assembly of organ building
blocks and increase in cell viability
187
Liver organoids GelMA Cylindrical organoid-
laden constructs
Reproduction of liver-specific ammo-
nia detoxification function
188
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4674 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
of discrete droplets.
205–208
It can meet conditions such as a high
surface volume ratio and shear stress, and also realize the
control of physical or chemical conditions for the growth
microenvironment.
The cellular microenvironment, including cytokines, ECM,
physical cues and cocultured cell types, can be integrated in the
microfluidic platform (Fig. 9a), which supports cell growth,
metabolism, proliferation, differentiation, apoptosis and other
physiological activities. Recently, microfluidic technology has
made great progress in the construction of a biomimetic tumor
microenvironment (TME).
209
Chung et al. constructed a bio-
mimetic TME using a microfluidic platform to achieve the
coculture of tumor cell lines with primary fibroblasts (Fig. 9b).
210
The biomimetic culture system could be used to directly observe
angiogenesis induced by tumor–matrix interactions and to
reconstruct simultaneous angiogenesis and lymphangiogenesis
induced by tumor–matrix interactions and TME-simulating
extrinsic factors. The microenvironment of steatohepatitis
could also be constructed on the microfluidic chip (Fig. 9c). It
realized the steatohepatitis-related interactions of multiple
cells, multiple biochemical factors and matrices.
211
To precisely
control the oxygen tension in the cellular microenvironment,
Gao et al. fabricated a microfluidic device using oxygen-
permeable and water-impermeable materials.
212
By incorporat-
ing a gas-permeable zone in the top of the cell culture chamber
and equipping it with a tank of chemical caramel alcohol for
oxygen consumption, local control of the oxygen tension in the
cell culture was achieved. Integrated microfluidic devices could
be used to produce alginate microgels that encapsulate
cells and to gently separate the microgels from immiscible oil
solutions, resulting in a significant increase in cell survival.
213
Given the importance of chemical signals to cells, it is essential
to efficiently deliver related signal molecules such as biological
signal peptides in the microfluidic platform. Zhao et al. con-
structed GelMA microspheres integrated with biological signal
peptides in a microfluidic system, realizing the effective deliv-
ery of integrated biological signal peptides.
214
The system
constructed an accurate chemical microenvironment, which
substantially promoted the osteogenic differentiation of MSCs
and the vascularization of HUVECs in vitro.
Microfluidics have been used to regulate the biomimetic cell
culture environment in real time, such as controlling the
delivery of oxygen and nutrients, increasing the concentration
of metabolites and secretory factors, and regulating the
mechanical stimulation applied to cells.
216
For example, phy-
sical cues have been applied to finely tune cell culture channels
to reconstruct the spatial and physical microenvironment.
217
Microfluidic chip technology has also provided a potent plat-
form to study the communications between different types of
cells by restricting the cells to different chambers, between
which the exchange of signal molecules is allowed.
215
Microfluidic chip technology has been employed to estab-
lish in vitro microscopic physiological models, which are
known as organs-on-a-chip. Various in vitro physiological
models in microfluidic platforms have been developed to study
pathogenesis and pharmacology, aiming at overcoming severe
Table 3 (continued )
Method Principle Cell type Material Printing structure Results Ref.
Magnetic
bioprinting
Guidance of the magne-
tized cells into 3D patterns
by mild magnetic
Fibroblasts Alginate, methylcellulose, polyacrylic
acid-stabilized magnetite nano-
particles of Fe
3
O
4
Cubes Remote control and navigate hydrogel
actuators
189
Dental pulp stem cells Gold and iron oxide nanoparticles
crosslinked by poly-L-lysine
3D spheroids Formation of biofunctional salivary
gland organoids
190
Kenzan
bioprinting
Using the microneedles as
temporary support to pro-
vide spatial organization
for the spheroids to merge
Spheroid composed of MSCs N.A. Tubular constructs Dispensation of spheroids according
to the size and shape of defect
191
Spheroid composed of
hepatocytes
N.A. Spheres Construction of a novel liver tissue-
like model
192
Spheroid composed of dermal
fibroblasts
N.A. Ring-like tissues Reestablishment of tendon-like struc-
tures under tensile culture
193
Spheroid composed of chon-
drocytes, endothelial cells and
MSCs
N.A. Tubular constructs Generation of tubular artificial
tracheas
194
Spheroid composed of fibro-
blasts, HUVECs, MSCs and
smooth muscle cells
N.A. Tubular structures Generation of artificial esophagus 195
Aspiration-
assisted
Bioprinting
Pick of cells to the speci-
fied position by aspiration
forces
Spheroid composed of MSCs,
HUVECs
N.A. Geometrical shapes in
a single layer
Osteogenic spheroids as building
blocks
196
Spheroid composed of ADSCs Sacrificial alginate hydrogel Single layer structure
of different component
Simulation of the bone and cartilage
structures in natural tissues
197
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4675
health risks such as cancer and COVID-19. For example, Liu
et al. proposed a microfluidic method for the large-scale
production of multi-size 3D tumors from human hepatocarci-
noma HepG2 cells and human glioma U251 cells with a
biomimetic TME, which were used to monitor the tumor
response in different fluid microenvironments and test multi-
ple anticancer drugs.
218
It provides an efficient and low-cost
method for the development of anticancer drugs. To investigate
the underlying pathogenesis of COVID-19, Zhang et al. cultured
human alveolar epithelial cells, pulmonary microvascular
endothelial cells, and immune cells in a microfluidic chip to
mimic alveolar capillary barrier damage and the pulmonary
inflammatory response after SARS-CoV-2 infection.
219
The
microfluidic chip device consisted of an upper alveolar epithelial
channel and a lower pulmonary microvascular endothelial chan-
nel, and the cells were cocultured under perfused media flow,
thereby simulating the spatial structure of the cellular microenvir-
onment and the chemical microenvironment by cytokine secretion
(Fig. 10a). Guo et al. also studied the intestinal response to viral
infection in a microfluidic chip.
220
The upper channel cocultured
intestinal epithelial Caco-2 cells and HT-29 cells to establish the
intestinal barrier. The lower channel was cocultured with HUVECs
and immune cells to simulate the microvascular endothelium
(Fig. 10b). These organs-on-chips provided potent tools for testing
drugs and exploring disease mechanisms.
In addition to individual cells, microfluidic chip technology
can also be used to integrate cellular aggregates, especially
organoids,
221
to greatly improve the complexity and accuracy of
organ models in vitro. For example, Liu et al. constructed a
multiorgan-on-a-chip system consisting of upstream ‘‘lung
organs’’ and downstream ‘‘brain organs’’ (Fig. 10c).
222
The
multiorgan microfluidic chip was fabricated using two PDMS
layers and a microporous membrane. The upstream ‘‘lung
organs’’ are organoids cocultured with a sandwich structure,
including tumor cells, pulmonary vascular endothelial cells,
immune cells, fibroblasts and bronchial epithelial cells. The
two sides of the vacuum channel simulated the respiratory
rhythm to reconstruct the occurrence of lung cancer. Alterna-
tively, the downstream ‘‘brain organs’’ are organoids composed
of two cerebral parenchymal chambers surrounded by vascular
channels with a bionic blood–brain barrier structure. Vascular
channels and parenchymal chambers communicate through
microporous membranes to achieve intercellular interactions
and allow tumor cells to penetrate the blood–brain barrier.
Using this multiorgan-on-a-chip system, the pathological pro-
cess of primary lung cancer to brain metastasis can be simu-
lated, which provides a new idea for the development of specific
drugs and conducive to more accurate and effective treatment.
In another example, liver, heart, and lung organoids were
combined in a modular manner and perfused with culture
Fig. 7 Schematic of bioprinting technology. (a) Inkjet bioprinting system based on thermal or voltage. (b) Laser-assisted bioprinting system.
(c) Extrusion-based bioprinting system, including pneumatic pressure, piston and screw auxiliary structure. (d) Acoustic bioprinting system.
(e) Photocuring-based bioprinting system, including stereolithography appearance (SLA) and DLP. (f) Magnetic bioprinting system. Reproduced with
permission.
165
Copyright 2020, Elsevier B.V.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4676 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
medium in a microfluidic chip system (Fig. 10d).
223
This model
revealed the cardiotoxicity of bleomycin, which was previously
believed to be non-cardiotoxic by simple models such as heart
cells and organoids. The cardiotoxicity of bleomycin is gener-
ated as a result of cytokine-mediated interactions between the
lung and heart tissues. This is a typical example that indicates
the great significance of microfluidic chip technology as an
integrated platform that is superior to individual organoids.
By combining organoids with microfluidic technology, a more
complex and accurate cellular microenvironment can be
created, and more precise in vitro physiological models can
be established. In addition to the liver–heart–lung organoids-
on-a-chip described above, several other multiorgan-on-a-
chip systems have also been developed, such as liver–heart
organoids-on-a-chip
224
and gastrointestinal-liver organoids-on-
a-chip.
225
The liver-heart organoid on-a-chip system can be
used to evaluate the safety of antidepressants to heart tissue
after liver metabolism, which provides a new possibility for
predicting the side effects of antidepressant drugs.
224
Gastro-
intestinal-liver organoids-on-a-chip can be used to predict
how the two organs jointly respond to drugs.
225
These
multiorgan-on-a-chip systems include complex interactions
between different organs, providing a more realistic micro-
environment for cells than a single organ model. More infor-
mation about multiorgan-on-a-chip is available in a review by
Picollet-D’hahan et al.
226
Theoretically, various cellular aggregates, such as spheroids,
organoids and assembloids, can be incorporated into micro-
fluidic chip systems. The reasonable selection of cellular aggre-
gates and the design of microfluidic chips can provide a potent
strategy for biomimetic cell culture, based on which in vitro
tissue and organ models with biomimetic structures and func-
tions can be developed.
4. Challenges and future directions
With the growing understanding of the cellular microenviron-
ment and the emergence of new technologies, biomimetic cell
culture has reached an unprecedented height. Due to their
simple method and low requirements, the success rate of
conventional scaffold strategies is the highest among the
Fig. 8 (a) Schematic of DLP bioprinting of the microtissue bioink. The GelMA-microtissue bioink was printed in an auricular shape. Reproduced with
permission.
187
Copyright 2022, John Wiley & Sons, Inc. (b) Construction of an artificial esophagus with multicellular spheroids. Reproduced with
permission.
195
Copyright 2019, Public Library of Science. (c) Features of bioprinters and bioinks for organoid printing. Reproduced with permission.
204
Copyright 2021, Springer Nature Switzerland AG. (d) Application schematic of BATE. It can achieve precise control of cell density and tissue structure in an
environment that allows multicell self-organization. Reproduced with permission.
132
Copyright 2021, Springer Nature.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4677
strategies. In contrast, the scaffold-free strategies of cellular
aggregates (spheroids, organoids and assembloids) are much
more complex and have higher requirements. In these strategies,
especially organoids and assembloids, the self-assembly of cells
involves complex cell communications and regulations, resulting
in lower success rates than that of conventional scaffold strategies.
Fig. 9 Construction of the cell microenvironment in a microfluidic chip. (a) Organ-on-a-chip relies on cells, biomaterials and culture systems to
reconstruct the microenvironment to replicate the key functional properties of tissues or organs. Reproduced with permission.
215
Copyright 2021,
Elsevier B.V. (b) Establishment of the TME in vitro biomimetic model in a microfluidic platform. Reproduced with permission.
210
Copyright 2017, Wiley-
VCH. (c) Construction of the steatohepatitis microenvironment in a microfluidic chip. Abbreviations: Ch., channel. Reproduced with permission.
211
Copyright 2021, Wiley Periodicals LLC.
Fig. 10 (a) Construction of a SARS-CoV-2 infection model in vitro. Reproduced with permission.
219
Copyright 2020, Wiley-VCH. (b) Multilayer intestinal
configuration of SARS-CoV-2 infection on a chip device. Reproduced with permission.
220
Copyright 2020, Elsevier B.V. (c) Schematic diagram of the
pathological process of brain metastasis from lung cancer simulated by a multiorgan microfluidic chip. Reproduced with permission.
222
Copyright 2019,
Elsevier B.V. (d) (i) Construction of an organ-on-a-chip system containing three types of organoids. (ii) Cell activity of cardiac organoids cocultured with
two other types of organoids on day 9. (iii) Cardiac organoid beating plots on day 9. Reproduced with permission.
223
Copyright 2017, Springer Nature
Limited.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4678 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
For instance, Sachs et al. constructed breast cancer organoids with
asuccessrateofca. 80%.
227
Yan et al. constructed tissue organoids
from gastric cancer patients. The success rate of non-tumor tissue
organoids was ca. 90%, while that of tumor organoids was only
ca. 50%.
228
Tsao et al. constructed a non-small cell lung cancer
organoid with a short-term (o3months)successrateof74%.
229
3D bioprinting and microfluidic chip technologies as integration
platforms for biomimetic cell culture are still being developed. 3D
bioprinting using bioinks containing individual cells has been
widely applied in tissue engineering and possesses a high success
rate, while 3D bioprinting using bioinks containing cellular aggre-
gates (spheroids, organoids and assembloids) is more challenging
and has a lower success rate, as well as higher cost in terms of
time, money and labor.
230
Although individual cells and cellular
aggregates can be integrated into microfluidic chips to achieve
more advanced biomimetic cell cultures, this is still not sufficient
for engineering the meticulous and cumbersome conditions in
microfluidic chips. Moreover, due to the lack of uniform interna-
tional standards, there may be variations between individuals,
making their success rate lower. Their success rate is also limited
by the understanding of cell signaling pathways, the sensitivity of
sensing, analysis platforms, etc.
Conventional scaffold-based strategies, including solid scaf-
folds and hydrogels, can provide a 3D environment for cell
attachment and growth. Simple chemical and physical signals
can also be included in the structures. However, it is difficult to
achieve a high cell density and precisely control the cell
distribution. It is also challenging to introduce gradients of
biochemical factors and complicated physical cues. In addi-
tion, nutrient supplementation is difficult, especially for inner
cells when the constructs are large. New attempts and progress
in this field are being made to resolve these problems. Holland
et al. used a rotational internal flow layer engineering techni-
que to achieve a high cell density (10
8
cells mL
1
) and control
the distribution of cells in different gel layers.
231
Wei et al.
constructed a bilayer hydrogel, which can achieve a biomimetic
gradient of composition, structural pore size, and biochemical
cues (kartogenin).
232
A smaller pore size (150–200 mm) and
higher concentration of kartogenin can induce the formation of
more chondrocytes, while a larger pore size (200–300 mm),
lower concentration of kartogenin and the presence of b-TCP
can induce more osteogenic differentiation.
Scaffold-free strategies, including spheroids, organoids and
assembloids, represent a significant step forward in the frontier
of biomimetic cell culture. They can achieve a biomimetic
spatial environment for cells with a high cell density and
sufficient cell–cell interactions. Moreover, gradients of bio-
chemical factors, oxygen and nutrients can also be formed in
cellular aggregates. Hubert et al. found that tumor organoids
generated nonlinear gradients of stem cell density and
hypoxia.
233
However, it is difficult to accurately control the
surrounding microenvironment of cellular aggregates, and it
cannot well reproduce the complex and dynamic microenviron-
ment in the process of organ or tissue development. Mean-
while, mature vascularization has not been achieved with these
cellular aggregates.
234
Thus, nutrient supplementation is still a
problem with an increase in the size of the aggregates. It was
found that 4 mm brain organoids began to undergo apoptosis
after approximately 100 days.
235
In addition, cellular aggregates
have variability and heterogeneity in terms of morphology and
function for different culture batches.
236
Although assembloids
increase the complexity and diversity in cell–cell and cell–
matrix interactions compared with individual organoids, infor-
mation exchange and functionality remain inadequate.
141
Recently, the converging strategies of cellular aggregates and
3D bioprinting or microfluidic chip technologies have shown
great potential in this field.
237,238
For example, Gong et al.
developed a controllable perfusion microfluidic chip to pre-
cisely control the fluidic shear stress and oxygen concentration
distribution in the process of culturing retinal organoids.
237
3D bioprinting and microfluidic chip technologies can serve
as integration platforms for different forms of biomimetic cell
culture, especially cellular aggregates, which allows the con-
struction of complex heterogeneous multi-tissue/organ struc-
tures. Also, interactions among multiple tissues and organs can
be established. Moreover, using these integration platforms,
high-throughput preparation and measurement are possible
through precise regulation of chemical and physical signals.
239
In general, multiorgan-based bioprinting and microfluidic
chips are currently the most promising systems of biomimetic
cell culture. Microfluidic chips have significant advantages
in controlling specific organizational structures and chemical
gradients and can achieve a high-level biomimetic micro-
environment.
240
However, integration platforms are also limi-
ted by some of the challenges faced by cellular aggregates, such
as nutrient supplementation and reproductivity. Recently, an
oxygen detection system that can detect the oxygen pressure in
real time was reported.
241
It could deliver oxygen when the
oxygen concentration decreased, which may facilitate oxygen
supply and diffusion in cellular aggregate culture. Nevertheless,
the resolution of nutrient supplementation ultimately relies on
the introduction of functional blood vessels.
242
In addition, to
overcome the issue of heterogeneity and variability, it is essential
to standardize the production of cellular aggregates and introduce
precise biosensor elements and automation systems.
243,244
Although biomimetic cell culture has made great progress,
there is still a long way to go to precisely engineer the micro-
environment of cells. In the future, the emergence and integra-
tion of new technologies may boost the pace of biomimetic cell
culture. For example, the application of gene editing technology
in cellular aggregates provides great potential for the develop-
ment of research, disease modelling and personalized medicine.
DuringtheCOVID-19pandemic,therapiduseofintegration
platforms to explore mechanisms and treatments has demon-
strated the advantages of using CRISPR-Cas9 technology
245
for
cellular aggregates. This technology has completely changed the
modelling of human organs and tumors, thereby deepening our
understanding of embryogenesis, organogenesis and tumori-
genesis.
246
Moreover, nanoscale biomaterials, such as nano-
particles, nanocapsules and nanofibers, are expected to be used
as functional additives to regulate physiological and metabolic
activities in biomimetic cell culture. Functional nanomaterials also
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4679
help to improve the cell density of stem cell spheroids, as well as
the activity, renewal ability, proliferation ability, differentiation
ability and paracrine effect of stem cells. The control of the spatial,
chemical and physical microenvironment in an in vitro model is
the key to realizing biomimetic cell culture. Therefore, exploring
the effects of spatial structure, chemical signals and physical cues
on cells is a prerequisite for achieving controllability. The combi-
nation of biomimetic cell culture and automation technology is an
interdisciplinary task and may be a good choice to develop
reproducible in vitro models in the future.
50
5. Conclusion
How to maintain or recapitulate the native key features of cells,
such as morphology, behavior, metabolism, and function,
in vitro is crucial for a wide range of fundamental biomedical
research and applications, especially in disease modelling and
pathological studies, drug development and evaluation, tissue
engineering and regenerative medicine. The basic principle of
biomimetic cell culture is to rebuild a microenvironment for
cells in vitro comparable to their native microenvironment
in vivo. The main task lies in the reoccurrence of the compli-
cated microenvironment in vitro by using appropriate materials,
signals and manufacturing technologies. The complicated micro-
environment of cells can be roughly simplified into three aspects,
i.e.,spatial,chemicalandphysicalfeatures.Spatialstructuressuch
as dimension and pore size can affect cell adhesion, migration,
and proliferation. The pore size in the spatial microenvironment
can affect the direction of cell differentiation, and the pore
orientation may directly affect the direction of cell migration.
The composition and spatial distribution of cells and the ECM
are also significant in the spatial microenvironment. Various
biochemical factors, such as interleukin, TGF-b,HGF,BMP-4
and O
2
, play a major role in the chemical microenvironment. They
control the growth and metabolism of cells and have profound
effects on the fate of cells by regulating gene transcription and
protein synthesis and secretion. Physical cues in the microenvir-
onment are also crucial factors that influence cell fate. Internal
stress, such as fluid shear stress, hydrostatic pressure and oxygen
partial pressure, and external stimuli may affect cell behaviors by
activating specific signaling pathways. The spatial, chemical, and
physical signals in the cellular microenvironments play a synergis-
tic role in biomimetic cell culture in vitro.
This has inspired a variety of strategies for biomimetic cell
culture involving a 3D spatial environment, specific biochem-
ical signal molecules, and appropriate physical cues. Efforts
have been made to achieve biomimetic cell culture in recent
decades with fast-growing development from conventional
scaffold-based methods to self-organized multicellular aggre-
gates such as spheroids, organoids and assembloids. Recently,
the development of 3D bioprinting and microfluidic chip
technologies has provided potent platforms to achieve higher-
level biomimetic cell culture. Conventional scaffold-based
strategies are easy to conduct, but they only involve simple
physical or chemical cues and limited cell density and
distribution. In comparison, cellular aggregates have a higher
cell density and higher-level biomimetic microenvironment,
although they suffer from issues of nutrient supplementation
and reproductivity. 3D bioprinting and microfluidic chip tech-
nologies integrated with cellular aggregates have reached a new
height in biomimetic cell culture. Despite the current issues of
variability and heterogeneity of cellular aggregates, they are
promising strategies for biomimetic cell culture by establishing
standards and introducing intelligent and automated systems.
In the era when animal experiments are no longer the standard,
biomimetic cell culture in the future will play a great role in
disease modelling, drug development, tissue engineering and
regenerative medicine with the advantages of high throughput,
low cost, high precision and low ethical risk.
Author contributions
Qiuchen Luo, Zhaoying Wu: data curation, visualization, meth-
odology, writing original draft, writing review & editing.
Keyuan Shang, Jing Zhu, Abeer Ahmed Qaed Ahmed, Chixiang
Huang: data curation, visualization, writing review & editing.
Tiefeng Cao: resources, writing review & editing, funding
acquisition. Lin Xiao: conceptualization, methodology, writing
review & editing, supervision, project administration, funding
acquisition.
Conflicts of interest
There are no conflicts of interest to declare.
Acknowledgements
The authors acknowledge the National Natural Science Founda-
tion of China (52173151, 82002734, 51803067), the Natural
Science Foundation of Guangdong Province of China (2021A15-
15011084, 2019A1515110312), the Fundamental Research
Funds for the Central Universities (22qntd1302) and Shenzhen
Outbound Postdoctoral Scientific Research Funding (SZBH202108)
for their financial support. The authors thank Prof. Antonella
Forlino from the Department of Molecular Medicine, Biochemistry
Unit, University of Pavia, 27100 Pavia, Italy, for her valuable
contribution in editing the current review.
References
1 C. Jensen and Y. Teng, Front. Mol. Biosci., 2020, 7, 33.
2 L. P. Ferreira, V. M. Gaspar and J. F. Mano, Acta Biomater.,
2018, 75, 11.
3 X. Hu, Z. Xia and K. Cai, J. Mater. Chem. B, 2022, 10, 1486.
4 M. Wadman, Science, 2023, 379, 127.
5 A. D. Theocharis, S. S. Skandalis, C. Gialeli and
N. K. Karamanos, Adv. Drug Delivery Rev., 2016, 97,4.
6 J. Lou and D. J. Mooney, Nat. Rev. Chem., 2022, 6, 726.
7 R. Li, Z. Wang, J. Huang, S. He, Y. Peng, Y. Wan and Q. Ma,
Front. Immunol., 2022, 13, 816037.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4680 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
8 O. R. Tonti, H. Larson, S. N. Lipp, C. M. Luetkemeyer,
M. Makam, D. Vargas, S. M. Wilcox and S. Calve, Acta
Biomater., 2021, 132, 83.
9 D. K. Mills, Y. Luo, A. Elumalai, S. Esteve, S. Karnik and
S. Yao, Gels, 2020, 6, 47.
10 J. H. Ylostalo, Cells, 2020, 9.
11 X. Liu and P. X. Ma, Biomaterials, 2009, 30, 4094.
12 X. Liu, M. N. Rahaman and Q. Fu, Acta Biomater., 2011,
7, 406.
13 F. L. He, D. W. Li, J. He, Y. Y. Liu, F. Ahmad, Y. L. Liu,
X. Deng, Y. J. Ye and D. C. Yin, Mater. Sci. Eng., C, 2018,
86, 18.
14 V. Karageorgiou and D. Kaplan, Biomaterials, 2005, 26,
5474.
15 S. J. Jiang, M. H. Wang, Z. Y. Wang, H. L. Gao, S. M. Chen,
Y. H. Cong, L. Yang, S. M. Wen, D. D. Cheng, J. C. He and
S. H. Yu, Adv. Funct. Mater., 2022, 32, 2110931.
16 Y. Zhang, F. Yang, K. Liu, H. Shen, Y. Zhu, W. Zhang,
W. Liu, S. Wang, Y. Cao and G. Zhou, Biomaterials, 2012,
33, 2926.
17 K. M. Yamada, A. D. Doyle and J. Lu, Trends Cell Biol., 2022,
32, 883.
18 W. Xu, J. Zhu, T. Cao, G. Yang, A. A. Q. Ahmed and L. Xiao,
Biomater. Adv., 2023, 153, 213567.
19 N. E. Ryu, S. H. Lee and H. Park, Cells, 2019, 8, 1620.
20 A. M. Hilderbrand, E. M. Ovadia, M. S. Rehmann,
P. M. Kharkar, C. Guo and A. M. Kloxin, Curr. Opin. Solid
State Mater. Sci., 2016, 20, 212.
21 B. S. Gomes, B. Simo
˜es and P. M. Mendes, Nat. Rev. Chem.,
2018, 2, 0120.
22 H. Wei, J. Cui, K. Lin, J. Xie and X. Wang, Bone Res., 2022,
10, 17.
23 S. Miao, H. Cui, T. Esworthy, B. Mahadik, S. J. Lee, X. Zhou,
S. Y. Hann, J. P. Fisher and L. G. Zhang, Adv. Sci., 2020,
7, 1902403.
24 C. E. Miksch, N. P. Skillin, B. E. Kirkpatrick, G. K. Hach,
V. V. Rao, T. J. White and K. S. Anseth, Small, 2022,
18, 2200951.
25 M. B. Al-Shaibani, A. Dickinson, X. Nong-Wang, A. S. Tulah
and P. E. Lovat, Cytotherapy, 2017, 19, e23.
26 H. Li, T. Zhao, F. Cao, H. Deng, S. He, J. Li, S. Liu, Z. Yang,
Z. Yuan and Q. Guo, Bioeng. Transl. Med., 2022, 7, e10302.
27 S. S. Lee, J. H. Kim, J. Jeong, S. H. L. Kim, R. H. Koh, I. Kim,
S. Bae, H. Lee and N. S. Hwang, Biomaterials, 2020,
257, 120223.
28 J. Chen, A. Zhou, Y. Nie, K. Chen, Y. Zhang, Y. Xu, D. Kong,
K. Shao and X. Ning, Nano Lett., 2022, 22, 135.
29 S. Samanta, L. Yla
¨-Outinen, V. K. Rangasami, S. Narkilahti
and O. P. Oommen, Acta Biomater., 2022, 140, 314.
30 L. B. Gallagher, E. B. Dolan, J. O’Sullivan, R. Levey, B. L.
Cavanagh, L. Kovarova, M. Pravda, V. Velebny, T. Farrell,
F. J. O’Brien and G. P. Duffy, Acta Biomater., 2020,
107, 78.
31 H. Li, P. Li, Z. Yang, C. Gao, L. Fu, Z. Liao, T. Zhao, F. Cao,
W. Chen, Y. Peng, Z. Yuan, X. Sui, S. Liu and Q. Guo, Front.
Cell Dev. Biol., 2021, 9, 661802.
32 J. Zhang, J. Griesbach, M. Ganeyev, A.-K. Zehnder, P. Zeng,
G. N. Scha
¨dli, A. D. Leeuw, Y. Lai, M. Rubert and R. Mu
¨ller,
Biofabrication, 2022, 14, 035018.
33 D. D. Nguyen, C. H. Yao, L. J. Luo, H. C. Chen, Y. J. Hsueh,
D. H. Ma and J. Y. Lai, Carbohydr. Polym., 2022, 292,
119668.
34 C. A. Paggi, J. Hendriks, M. Karperien and S. Le Gac, Lab
Chip, 2022, 22, 1815.
35 S. Vermeulen, Z. Tahmasebi Birgani and P. Habibovic,
Biomaterials, 2022, 283, 121431.
36 L. Mor-Yossef Moldovan, N. Kislev, M. Lustig, L. Pomeraniec
and D. Benayahu, J. Cell. Physiol., 2020, 235, 8702.
37 S. W. Verbruggen, C. L. Thompson, M. P. Duffy, S. Lunetto,
J. Nolan, O. M. T. Pearce, C. R. Jacobs and M. M. Knight,
Cancers, 2021, 13, 2906.
38 E. Stavenschi, M. A. Corrigan, G. P. Johnson, M. Riffault
and D. A. Hoey, Stem Cell Res. Ther., 2018, 9, 276.
39 R. He, Z. Wang, M. Cui, S. Liu, W. Wu, M. Chen, Y. Wu,
Y. Qu, H. Lin, S. Chen, B. Wang and Z. Shao, Autophagy,
2021, 17, 3338.
40 S. Fu, C. L. Thompson, A. Ali, W. Wang, J. P. Chapple,
H. M. Mitchison, P. L. Beales, A. K. T. Wann and
M. M. Knight, Osteoarthr. Cartil., 2019, 27, 1064.
41 M. Huang, H. Liu, L. Zhu, X. Li, J. Li, S. Yang, D. Liu,
X. Song, H. Yokota and P. Zhang, J. Cell. Physiol.,2021,
236, 6391.
42 E. Zevolis, A. Philippou, A. Moustogiannis, A. Chatzigeorgiou
and M. Koutsilieris, Cells, 2022, 11,473.
43 J. Vaithilingam, P. Sanjuan-Alberte, S. Campora, G. A.
Rance, L. Jiang, J. Thorpe, L. Burroughs, C. J. Tuck, C.
Denning, R. D. Wildman, R. J. M. Hague, M. R. Alexander
and F. J. Rawson, Adv. Funct. Mater., 2019, 29, 1902016.
44 Y. Qin, X. B. Hu, W. T. Fan, J. Yan, S. B. Cheng, Y. L. Liu
and W. H. Huang, Adv. Sci., 2021, 8, 2003738.
45 C. Ning, Z. Zhou, G. Tan, Y. Zhu and C. Mao, Prog. Polym.
Sci., 2018, 81, 144.
46 X. Zhao, H. Wu, B. Guo, R. Dong, Y. Qiu and P. X. Ma,
Biomaterials, 2017, 122, 34.
47 Q. Wang, H. Wang, Y. Ma, X. Cao and H. Gao, Biomater.
Sci., 2022, 10, 6061.
48 Y. Li, X. Dai, Y. Bai, Y. Liu, Y. Wang, O. Liu, F. Yan, Z. Tang,
X. Zhang and X. Deng, Int. J. Nanomed., 2017, 12, 4007.
49 M. Malki, A. Shapira and T. Dvir, J. Nanobiotechnol., 2022, 20, 59.
50 J. L. Gerardo-Nava, J. Jansen, D. Gu
¨nther, L. Klasen,
A. L. Thiebes, B. Niessing, C. Bergerbit, A. A. Meyer,
J. Linkhorst, M. Barth, P. Akhyari, J. Stingl, S. Nagel,
T. Stiehl, A. Lampert, R. Leube, M. Wessling, F. Santoro,
S. Ingebrandt, S. Jockenhoevel, A. Herrmann, H. Fischer,
W. Wagner, R. H. Schmitt, F. Kiessling, R. Kramann and
L. De Laporte, Adv. Healthcare Mater., 2023, 2301030.
51 B. Dhandayuthapani, Y. Yoshida, T. Maekawa and
D. S. Kumar, Int. J. Polym. Sci., 2011, 2011, 290602.
52 Y. S. Zhang and A. Khademhosseini, Bio-Des. Manuf., 2020,
3, 155.
53 D. Lantigua, Y. N. Kelly, B. Unal and G. Camci-Unal, Adv.
Healthcare Mater., 2017, 6, 1700619.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4681
54 M. P. Nikolova and M. S. Chavali, Bioact. Mater., 2019,
4, 271.
55 H. Yao, X. Zou, S. Zheng, Y. Hu, S. Zhang, C. Liang,
H. Zhou, D. Wang, H. Wang, L. Yang and Q. Li, Mater.
Sci. Eng., C, 2021, 127, 112247.
56 J. C. M. Souza, M. B. Sordi, M. Kanazawa, S. Ravindran,
B. Henriques, F. S. Silva, C. Aparicio and L. F. Cooper, Acta
Biomater., 2019, 94, 112.
57 M. Q. Cheng, T. Wahafu, G. F. Jiang, W. Liu, Y. Q. Qiao,
X. C. Peng, T. Cheng, X. L. Zhang, G. He and X. Y. Liu, Sci.
Rep., 2016, 6, 24134.
58 Y. G. Zhang, J. P. Li, M. Soleimani, F. Giacomini,
H. Friedrich, R. Truckenmuller and P. Habibovic, Adv.
Funct. Mater., 2021, 31, 2102911.
59 A. K. Badekila, S. Kini and A. K. Jaiswal, J. Cell. Physiol.,
2021, 236, 741.
60 R. Riesco, L. Boyer, S. Blosse, P. M. Lefebvre, P. Assemat,
T. Leichle, A. Accardo and L. Malaquin, ACS Appl. Mater.
Interfaces, 2019, 11, 28631.
61 S. Jiang, C. Lyu, P. Zhao, W. Li, W. Kong, C. Huang,
G. M. Genin and Y. Du, Nat. Commun., 2019, 10, 3491.
62 D. Yang, J. Xiao, B. Wang, L. Li, X. Kong and J. Liao, Mater.
Sci. Eng., C, 2019, 104, 109927.
63 H. Luo, R. Cha, J. Li, W. Hao, Y. Zhang and F. Zhou,
Carbohydr. Polym., 2019, 224, 115144.
64 S. Derakhti, S. H. Safiabadi-Tali, G. Amoabediny and
M. Sheikhpour, Mater. Sci. Eng., C, 2019, 103, 109782.
65 Y. Liu, R. Wang, S. Ding, L. Deng, Y. Zhang, J. Li, Z. Shi,
Z. Wu, K. Liang, X. Yan, W. Liu and Y. Du, Biomaterials,
2022, 287, 121615.
66 M. Kuterbekov, P. Machillot, P. Lhuissier, C. Picart,
A. M. Jonas and K. Glinel, Acta Biomater., 2018, 75, 300.
67 B. Koh, N. Sulaiman, M. B. Fauzi, J. X. Law, M. H. Ng,
R. B. H. Idrus and M. D. Yazid, Cell Biosci., 2020, 10, 75.
68 L. Huang, L. Xiao, A. Jung Poudel, J. Li, P. Zhou,
M. Gauthier, H. Liu, Z. Wu and G. Yang, Carbohydr. Polym.,
2018, 202, 611.
69 A. Ornelas-Gonza
´lez, M. Gonza
´lez-Gonza
´lez and M. Rito-
Palomares, Crit. Rev. Biotechnol., 2021, 41, 1081.
70 Y. S. Zhang and A. Khademhosseini, Science, 2017,
356, eaaf3627.
71 L. Ding, S. Song, L. Chen, J. Shi, B. Zhao, G. Teng and
J. Zhang, Int. J. Biol. Macromol., 2021, 174, 101.
72 X. Q. Dou and C. L. Feng, Adv. Mater., 2017, 29, 1604062.
73 M. D. Konieczynska and M. W. Grinstaff, Acc. Chem. Res.,
2017, 50, 151.
74 X. Xu, V. V. Jerca and R. Hoogenboom, Mater. Horiz., 2021,
8, 1173.
75 H. J. Lee, Y. Seo, H. S. Kim, J. W. Lee and K. Y. Lee, ACS
Omega, 2020, 5, 15567.
76 S. Nam, R. Stowers, J. Lou, Y. Xia and O. Chaudhuri,
Biomaterials, 2019, 200, 15.
77 Y. Kittel, A. J. C. Kuehne and L. De Laporte, Adv. Healthcare
Mater., 2022, 11, 2101989.
78 L. Huang, A. M. E. Abdalla, L. Xiao and G. Yang, Int. J. Mol.
Sci., 2020, 21, 1895.
79 X. Cui, Y. Hartanto, C. Wu, J. Bi, S. Dai and H. Zhang,
J. Biomed. Mater. Res., Part A, 2018, 106, 2899.
80 D. Rommel, M. Mork, S. Vedaraman, C. Bastard, L. P. B.
Guerzoni, Y. Kittel, R. Vinokur, N. Born, T. Haraszti and
L. De Laporte, Adv. Sci., 2022, 9, 2103554.
81 S. A. Langhans, Front. Pharmacol., 2018, 9,6.
82 S. Li, Z. Zhang and L. Han, Trends Cancer, 2020, 6, 622.
83 S. Li, H. Zhao, X. Han, B. Ni, L. He, O. Mukama, J. de Dieu
Habimana, Z. Lin, R. Huang, H. Huang, C. Tian, F. Tang
and Z. Li, Stem Cell Res. Ther., 2021, 12, 188.
84 D. Y. Ko, M. Patel, H. J. Lee and B. Jeong, Adv. Funct. Mater.,
2018, 28, 1706286.
85 W. Kim, Y. Gwon, S. Park, H. Kim and J. Kim, Bioact.
Mater., 2023, 19, 50.
86 S. Nath and G. R. Devi, Pharmacol. Ther., 2016, 163, 94.
87 S. L. Zheng and K. M. Loh, Curr. Opin. Biotechnol, 2022,
78, 102810.
88 S. M. Kang, D. Kim, J. H. Lee, S. Takayama and J. Y. Park,
Adv. Healthcare Mater., 2021, 10, e2001284.
89 H. Byun, S. Lee, G. N. Jang, H. Lee, S. Park and H. Shin,
Biofabrication, 2021, 14, 015007.
90 R. Rasouli and M. Tabrizian, Small, 2021, 17, e2101931.
91 S. Ai, H. Li, H. Zheng, J. Liu, J. Gao, J. Liu, Q. Chen and
Z. Yang, Sci. China Mater., 1655, 2022, 65.
92 S.-j Kim, H. Byun, S. Lee, E. Kim, G. M. Lee, S. J. Huh, J. Joo
and H. Shin, Acta Biomater., 2022, 142, 60.
93 G.-B. Im, S.-W. Kim and S. H. Bhang, J. Ind. Eng. Chem.,
2021, 93, 228.
94 L. Shariati, Y. Esmaeili, S. Haghjooy Javanmard, E. Bidram
and A. Amini, Stem Cells, 2021, 39, 1625.
95 S. A. Yi, Y. Zhang, C. Rathnam, T. Pongkulapa and
K. B. Lee, Adv. Mater., 2021, 33, e2007949.
96 L. Jiang, Y. Shen, Y. Liu, L. Zhang and W. Jiang, Theranos-
tics, 2022, 12, 1537.
97 A. Marsee, F. J. M. Roos, M. M. A. Verstegen, A. Marsee,
F. Roos, M. Verstegen, H. Clevers, L. Vallier, T. Takebe,
M. Huch, W. C. Peng, S. Forbes, F. Lemaigre, E. de Koning,
H. Gehart, L. van der Laan, B. Spee, S. Boj, P. Baptista,
K. Schneeberger, C. Soroka, M. Heim, S. Nuciforo, K. Zaret,
Y. Saito, M. Lutolf, V. Cardinale, B. Simons, S. van Ijzendoorn,
A. Kamiya, H. Chikada, S. Wang, S. J. Mun, M. J. Son,
T. T. Onder, J. Boyer, T. Sato, N. Georgakopoulos,
A.Meneses,L.Broutier,L.Boulter,D.Gru
¨n, J. Ijzermans,
B. Artegiani, R. van Boxtel, E. Kuijk, G. Carpino, G. Peltz,
J. Banales, N. Man, L. Aloia, N. LaRusso, G. George,
C. Rimland, G. Yeoh, A. Grappin-Botton, D. Stange, N. Prior,
J. E. E. Tirnitz-Parker, E. Andersson, C. Braconi, N. Hannan,
W.-Y. Lu, S. Strom, P. Sancho-Bru, S. Ogawa, V. Corbo,
M. Lancaster, H. Hu, S. Fuchs, D. Hendriks, H. Gehart, E. de
Koning, F. Lemaigre, S. J. Forbes, W. C. Peng, M. Huch,
T.Takebe,L.Vallier,H.Clevers,L.J.W.vanderLaanand
B. Spee, Cell Stem Cell, 2021, 28,816.
98 G. Rossi, A. Manfrin and M. P. Lutolf, Nat. Rev. Genet.,
2018, 19, 671.
99 Z. Zhao, X. Chen, A. M. Dowbaj, A. Sljukic, K. Bratlie,
L. Lin, E. L. S. Fong, G. M. Balachander, Z. Chen,
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4682 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
A. Soragni, M. Huch, Y. A. Zeng, Q. Wang and H. Yu, Nat.
Rev. Methods Primers, 2022, 2, 94.
100 E. Y. Jeon, L. Sorrells and H. E. Abaci, Front. Bioeng.
Biotechnol., 2022, 10, 1038277.
101 E. Yoshihara, C. O’Connor, E. Gasser, Z. Wei, T. G. Oh,
T. W. Tseng, D. Wang, F. Cayabyab, Y. Dai, R. T. Yu,
C. Liddle, A. R. Atkins, M. Downes and R. M. Evans, Nature,
2020, 586, 606.
102 R. Nguyen, S. Da Won Bae, L. Qiao and J. George, Cancer
Lett., 2021, 508, 13.
103 J. Huang, L. Zhang, A. Lu and C. Liang, Cells, 2023,
12, 1590.
104 S. Chen, X. Chen, Z. Geng and J. Su, Bioact. Mater., 2022,
18, 15.
105 H. Clevers, Cell, 2016, 165, 1586.
106 D. Vyas, P. M. Baptista, M. Brovold, E. Moran, B. Gaston,
C. Booth, M. Samuel, A. Atala and S. Soker, Hepatology,
2018, 67, 750.
107 S. J. Mun, J. S. Ryu, M. O. Lee, Y. S. Son, S. J. Oh, H. S. Cho,
M. Y. Son, D. S. Kim, S. J. Kim, H. J. Yoo, H. J. Lee, J. Kim,
C. R. Jung, K. S. Chung and M. J. Son, J. Hepatol., 2019,
71, 970.
108 N. Tanimizu, N. Ichinohe, Y. Sasaki, T. Itoh, R. Sudo,
T. Yamaguchi, T. Katsuda, T. Ninomiya, T. Tokino,
T. Ochiya, A. Miyajima and T. Mitaka, Nat. Commun.,
2021, 12, 3390.
109 F. Wu, D. Wu, Y. Ren, Y. Huang, B. Feng, N. Zhao, T. Zhang,
X. Chen, S. Chen and A. Xu, J. Hepatol., 2019, 70, 1145.
110 B. R. Dye, R. L. Youngblood, R. S. Oakes, T. Kasputis,
D. W. Clough, J. R. Spence and L. D. Shea, Biomaterials,
2020, 234, 119757.
111 T. G. Gu
¨ney, A. M. Herranz, S. Mumby, I. E. Dunlop and
I. M. Adcock, iScience, 2021, 24, 103061.
112 C. Lee, J. H. Song, Y. E. Cha, D. K. Chang, Y. H. Kim and
S. N. Hong, J. Crohns Colitis, 2022, 16, 1911.
113 G. W. He, L. Lin, J. DeMartino, X. Zheng, N. Staliarova,
T. Dayton, H. Begthel, W. J. van de Wetering, E. Bodewes,
J. van Zon, S. Tans, C. Lopez-Iglesias, P. J. Peters, W. Wu,
D. Kotlarz, C. Klein, T. Margaritis, F. Holstege and
H. Clevers, Cell Stem Cell, 2022, 29, 1333.
114 J. H. Low, P. Li, E. G. Y. Chew, B. Zhou, K. Suzuki, T. Zhang,
M. M. Lian, M. Liu, E. Aizawa, C. Rodriguez Esteban,
K. S. M. Yong, Q. Chen, J. M. Campistol, M. Fang,
C. C. Khor, J. N. Foo, J. C. Izpisua Belmonte and Y. Xia,
Cell Stem Cell, 2019, 25, 373.
115 K. T. Lawlor, J. M. Vanslambrouck, J. W. Higgins,
A. Chambon, K. Bishard, D. Arndt, P. X. Er, S. B. Wilson,
S. E. Howden, K. S. Tan, F. Li, L. J. Hale, B. Shepherd,
S. Pentoney, S. C. Presnell, A. E. Chen and M. H. Little, Nat.
Mater., 2021, 20, 260.
116 J. W. Kim, S. A. Nam, J. Yi, J. Y. Kim, J. Y. Lee, S.-Y. Park,
T. Sen, Y.-M. Choi, J. Y. Lee, H. L. Kim, H. W. Kim, J. Park,
D.-W. Cho and Y. K. Kim, Adv. Sci., 2022, 9, 2103526.
117 P. A. Mollica, E. N. Booth-Creech, J. A. Reid, M. Zamponi,
S. M. Sullivan, X.-L. Palmer, P. C. Sachs and R. D. Bruno,
Acta Biomater., 2019, 95, 201.
118 Y. Park, E. Cheong, J. G. Kwak, R. Carpenter, J. H. Shim
and J. Lee, Sci. Adv., 2021, 7, eabd6495.
119 W. L. Tam, L. Freitas Mendes, X. Chen, R. Lesage, I. Van
Hoven, E. Leysen, G. Kerckhofs, K. Bosmans, Y. C. Chai,
A. Yamashita, N. Tsumaki, L. Geris, S. J. Roberts and
F. P. Luyten, Stem Cell Res. Ther., 2021, 12, 513.
120 C. Xie, R. Liang, J. Ye, Z. Peng, H. Sun, Q. Zhu, X. Shen,
Y. Hong, H. Wu, W. Sun, X. Yao, J. Li, S. Zhang, X. Zhang
and H. Ouyang, Biomaterials, 2022, 288, 121741.
121 A. Akiva, J. Melke, S. Ansari, N. Liv, R. van der Meijden,
M. van Erp, F. Zhao, M. Stout, W. H. Nijhuis, C. de Heus,
C. Mun
˜iz Ortera, J. Fermie, J. Klumperman, K. Ito,
N. Sommerdijk and S. Hofmann, Adv. Funct. Mater.,
2021, 31, 2010524.
122 Q. Vallmajo-Martin, N. Broguiere, C. Millan, M. Zenobi-Wong
and M. Ehrbar, Adv. Funct. Mater., 2020, 30, 1910282.
123 X. Sun, J. Xiang, R. Chen, Z. Geng, L. Wang, Y. Liu, S. Ji,
H. Chen, Y. Li, C. Zhang, P. Liu, T. Yue, L. Dong and X. Fu,
Adv. Sci., 2021, 8, 2103079.
124 J. Ma, J. liu, D. Gao, X. Li, Q. Zhang, L. Lv, Y. Wang, J. Li,
Y. Zhu, Z. Wu, H. Hu, Y. Li, L. Ma, Q. Liu, Z. Hu, S. Zhang,
Y. Zhou, M. Wang and L. Leng, Adv. Sci., 2022, 9, 2104192.
125 J. Lee, W. H. van der Valk, S. A. Serdy, C. Deakin, J. Kim,
A. P. Le and K. R. Koehler, Nat. Protoc., 2022, 17, 1266.
126 J. A. Bagley, D. Reumann, S. Bian, J. Le
´vi-Strauss and
J. A. Knoblich, Nat. Methods, 2017, 14, 743.
127 P. R. Ormel, R. Vieira de Sa
´, E. J. van Bodegraven, H. Karst,
O. Harschnitz, M. A. M. Sneeboer, L. E. Johansen, R. E. van
Dijk, N. Scheefhals, A. Berdenis van Berlekom, E. Ribes
Martı
´nez, S. Kling, H. D. MacGillavry, L. H. van den Berg,
R. S. Kahn, E. M. Hol, L. D. de Witte and R. J. Pasterkamp,
Nat. Commun., 2018, 9, 4167.
128 S. Velasco, A. J. Kedaigle, S. K. Simmons, A. Nash, M. Rocha,
G.Quadrato,B.Paulsen,L.Nguyen,X.Adiconis,A.Regev,
J. Z. Levin and P. Arlotta, Nature, 2019, 570,523.
129 S. P. Pas-ca, P. Arlotta, H. S. Bateup, J. G. Camp, S. Cappello,
F. H. Gage, J. A. Knoblich, A. R. Kriegstein, M. A. Lancaster,
G.-L. Ming, A. R. Muotri, I.-H. Park, O. Reiner, H. Song,
L. Studer, S. Temple, G. Testa, B. Treutlein and
F. M. Vaccarino, Nature, 2022, 609, 907.
130 N. Vogt, Nat. Methods, 2021, 18, 27.
131 S. Kanton and S. P. Pas-ca, Development, 2022,
149, dev201120.
132 J. A. Brassard, M. Nikolaev, T. Hu
¨bscher, M. Hofer and
M. P. Lutolf, Nat. Mater., 2021, 20, 22.
133 Y. S. Zhang, A. Arneri, S. Bersini, S.-R. Shin, K. Zhu, Z. Goli-
Malekabadi, J. Aleman, C. Colosi, F. Busignani,
V. Dell’Erba, C. Bishop, T. Shupe, D. Demarchi, M. Moretti,
M. Rasponi, M. R. Dokmeci, A. Atala and A. Khademhosseini,
Biomaterials, 2016, 110,45.
134 Y. R. Lewis-Israeli, A. H. Wasserman and A. Aguirre, Bio-
molecules, 2021, 11, 1277.
135 Y. Miura, M. Y. Li, O. Revah, S. J. Yoon, G. Narazaki and
S. P. Pas-ca, Nat. Protoc., 2022, 17, 15.
136 P. Hofbauer, S. M. Jahnel, N. Papai, M. Giesshammer,
A. Deyett, C. Schmidt, M. Penc, K. Tavernini, N. Grdseloff,
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4683
C. Meledeth, L. C. Ginistrelli, C. Ctortecka, S
ˇ.S
ˇalic,
M. Novatchkova and S. Mendjan, Cell, 2021, 184, 3299.
137 P. Augsornworawat, L. Velazco-Cruz, J. Song and
J. R. Millman, Acta Biomater., 2019, 97, 272.
138 J. Candiello, T. S. P. Grandhi, S. K. Goh, V. Vaidya,
M. Lemmon-Kishi, K. R. Eliato, R. Ros, P. N. Kumta,
K. Rege and I. Banerjee, Biomaterials, 2018, 177, 27.
139 A. Linkous, D. Balamatsias, M. Snuderl, L. Edwards,
K. Miyaguchi, T. Milner, B. Reich, L. Cohen-Gould,
A. Storaska, Y. Nakayama, E. Schenkein, R. Singhania,
S. Cirigliano, T. Magdeldin, Y. Lin, G. Nanjangud,
K. Chadalavada, D. Pisapia, C. Liston and H. A. Fine, Cell
Rep., 2019, 26, 3203.
140 Y. Zhu, X. Zhang, L. Sun, Y. Wang and Y. Zhao, Adv. Mater.,
2023, 35, e2210083.
141 J. Andersen, O. Revah, Y. Miura, N. Thom, N. D. Amin,
K. W. Kelley, M. Singh, X. Chen, M. V. Thete,
E. M. Walczak, H. Vogel, H. C. Fan and S. P. Pas-ca, Cell,
2020, 183, 1913.
142 V. Velasco, S. A. Shariati and R. Esfandyarpour, Microsyst.
Nanoeng., 2020, 6, 76.
143 C. Mandrycky, Z. Wang, K. Kim and D. H. Kim, Biotechnol.
Adv., 2016, 34, 422.
144 E. Moradi, S. Jalili-Firoozinezhad and M. Solati-Hashjin,
Acta Biomater., 2020, 116, 67.
145 Z. Fu, L. Ouyang, R. Xu, Y. Yang and W. Sun, Mater. Today,
2022, 52, 112.
146 X. Li, B. Liu, B. Pei, J. Chen, D. Zhou, J. Peng, X. Zhang,
W. Jia and T. Xu, Chem. Rev., 2020, 120, 10793.
147 R. Suntornnond, W. L. Ng, X. Huang, C. H. E. Yeow and
W. Y. Yeong, J. Mater. Chem. B, 2022, 10, 5989.
148 E. Masaeli, V. Forster, S. Picaud, F. Karamali, M. H. Nasr-
Esfahani and C. Marquette, Biofabrication, 2020, 12,
025006.
149 C. Dou, V. Perez, J. Qu, A. Tsin, B. Xu and J. Li, ChemBioEng
Rev., 2021, 8, 517.
150 N. Ashammakhi, A. Hasan, O. Kaarela, B. Byambaa,
A. Sheikhi, A. K. Gaharwar and A. Khademhosseini, Adv.
Healthcare Mater., 2019, 8, 1801048.
151 K. Song, D. Zhang, J. Yin and Y. Huang, Addit. Manuf.,
2021, 41, 101963.
152 Z. Fu, S. Naghieh, C. Xu, C. Wang, W. Sun and X. Chen,
Biofabrication, 2021, 13, 033001.
153 Q. Guo, X. Su, X. Zhang, M. Shao, H. Yu and D. Li, Soft
Matter, 2021, 17, 3010.
154 K. Chen, E. Jiang, X. Wei, Y. Xia, Z. Wu, Z. Gong, Z. Shang
and S. Guo, Lab Chip, 2021, 21, 1604.
155 H. Cai, Z. Wu, Z. Ao, A. Nunez, B. Chen, L. Jiang,
M. Bondesson and F. Guo, Biofabrication, 2020, 12, 035025.
156 M. A. S. R. Saadi, A. Maguire, N. T. Pottackal, M. S. H.
Thakur, M. M. Ikram, A. J. Hart, P. M. Ajayan and
M. M. Rahman, Adv. Mater., 2022, 34, 2108855.
157 S. Park, W. Shou, L. Makatura, W. Matusik and K. Fu,
Matter, 2022, 5, 43.
158 Z. Wang, R. Abdulla, B. Parker, R. Samanipour, S. Ghosh
and K. Kim, Biofabrication, 2015, 7, 045009.
159 H. Goodarzi Hosseinabadi, E. Dogan, A. K. Miri and
L. Ionov, ACS Biomater. Sci. Eng., 2022, 8, 1381.
160 J. Spangenberg, D. Kilian, C. Czichy, T. Ahlfeld, A. Lode,
S. Gu
¨nther, S. Odenbach and M. Gelinsky, ACS Biomater.
Sci. Eng., 2021, 7, 648.
161 H. Tseng, C. Ionescu-Zanetti, J. Jensen, J. A. Gage, W. L.
Haisler, P. K. Desai, A. Liao, R. Brobey, M. Dehghani,
G. R. Souza and R. J. Amato, J. Clin. Oncol., 2016, 34, 291.
162 J. J. Martin, B. E. Fiore and R. M. Erb, Nat. Commun., 2015,
6, 8641.
163 M. Hospodiuk, M. Dey, D. Sosnoski and I. T. Ozbolat,
Biotechnol. Adv., 2017, 35, 217.
164 Y. Huang, X. Li, A. J. Poudel, W. Zhang and L. Xiao, Appl.
Mater. Today, 2022, 29, 101668.
165 I. Matai, G. Kaur, A. Seyedsalehi, A. McClinton and
C. T. Laurencin, Biomaterials, 2020, 226, 119536.
166 A. M. Compaan, K. Christensen and Y. Huang, ACS Bio-
mater. Sci. Eng., 2017, 3, 1519.
167 A. Negro, T. Cherbuin and M. P. Lutolf, Sci. Rep., 2018,
8, 17099.
168 J. Liu, M. Shahriar, H. Xu and C. Xu, Biofabrication, 2022,
14,045020.
169 G. L. Duffy, H. Liang, R. L. Williams, D. A. Wellings and
K. Black, Mater. Sci. Eng., C, 2021, 131, 112476.
170 D. Kang, J. A. Park, W. Kim, S. Kim, H. R. Lee, W. J. Kim,
J. Y. Yoo and S. Jung, Adv. Sci., 2021, 8, 2004990.
171 N. Touya, M. Devun, C. Handschin, S. Casenave, N. Ahmed
Omar, A. Gaubert, N. Dusserre, H. De Oliveira, O.
Ke
´roure
´dan and R. Devillard, Biofabrication, 2022, 14, 024104.
172 F. Kawecki, W. P. Clafshenkel, F. A. Auger, J. M. Bourget,
J. Fradette and R. Devillard, Biofabrication, 2018, 10, 035006.
173 V. Keriquel, H. Oliveira, M. Re
´my, S. Ziane, S. Delmond,
B. Rousseau, S. Rey, S. Catros, J. Ame
´de
´e, F. Guillemot and
J.-C. Fricain, Sci. Rep., 2017, 7, 1778.
174 A. Sorkio, L. Koch, L. Koivusalo, A. Deiwick, S. Miettinen,
B. Chichkov and H. Skottman, Biomaterials, 2018, 171, 57.
175 Z. Ataie, S. Kheirabadi, J. W. Zhang, A. Kedzierski,
C. Petrosky, R. Jiang, C. Vollberg and A. Sheikhi, Small,
2022, 18, 2202390.
176 W. Liu, Z. Zhong, N. Hu, Y. Zhou, L. Maggio, A. K. Miri,
A. Fragasso, X. Jin, A. Khademhosseini and Y. S. Zhang,
Biofabrication, 2018, 10, 024102.
177 J. H. Kim, Y. J. Seol, I. K. Ko, H. W. Kang, Y. K. Lee, J. J. Yoo,
A. Atala and S. J. Lee, Sci. Rep., 2018, 8, 12307.
178 L. Lian, C. Zhou, G. Tang, M. Xie, Z. Wang, Z. Luo, J. Japo,
D. Wang, J. Zhou, M. Wang, W. Li, S. Maharjan, M. Ruelas,
J. Guo, X. Wu and Y. S. Zhang, Adv. Healthcare Mater., 2022,
11, 2102411.
179 H. Chen, L. Du, J. Li, Z. Wu, Z. Gong, Y. Xia, Z. Fan,
Q. Qian, Z. Ding, H. Hu and S. Guo, J. Mater. Chem. B, 2022,
10, 1843.
180 S. Jentsch, R. Nasehi, C. Kuckelkorn, B. Gundert, S. Aveic
and H. Fischer, Small Methods, 2021, 5, 2000971.
181 H. Chen, Z. Wu, Z. Gong, Y. Xia, J. Li, L. Du, Y. Zhang,
X. Gao, Z. Fan, H. Hu, Q. Qian, Z. Ding and S. Guo, Adv.
Healthcare Mater., 2022, 11, 2102784.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
4684 | Mater. Horiz., 2023, 10, 4662–4685 This journal is © The Royal Society of Chemistry 2023
182 D. Jeong, J. W. Seo, H.-G. Lee, W. K. Jung, Y. H. Park and
H. Bae, Adv. Sci., 2022, 9, 2202877.
183 Y. He, F. Wang, X. Wang, J. Zhang, D. Wang and X. Huang,
Mater. Des., 2021, 202, 109588.
184 K. Yu, X. Zhang, Y. Sun, Q. Gao, J. Fu, X. Cai and Y. He,
Bioact. Mater., 2022, 11, 254.
185 J. S. Lee, H. S. Park, H. Jung, H. Lee, H. Hong, Y. J. Lee,
Y. J. Suh, O. J. Lee, S. H. Kim and C. H. Park, Addit. Manuf.,
2020, 33, 101136.
186 P. Zhang, H. Wang, P. Wang, Y. Zheng, L. Liu, J. Hu, Y. Liu,
Q. Gao and Y. He, Bioact. Mater., 2021, 6, 1402.
187 X. Xie, S. Wu, S. Mou, N. Guo, Z. Wang and J. Sun, Adv.
Healthcare Mater., 2022, 11, 2201877.
188 P. N. Bernal, M. Bouwmeester, J. Madrid-Wolff, M. Falandt,
S. Florczak, N. G. Rodriguez, Y. Li, G. Gro
¨ßbacher, R. A.
Samsom, M. van Wolferen, L. J. W. van der Laan, P. Delrot,
D. Loterie, J. Malda, C. Moser, B. Spee and R. Levato, Adv.
Mater., 2022, 34, 2110054.
189 J. Simin
´ska-Stanny, M. Nizioł, P. Szymczyk-Zio
´łkowska,
M. Broz˙yna, A. Junka, A. Shavandi and D. Podstawczyk,
Addit. Manuf., 2022, 49, 102506.
190 C. Adine, K. K. Ng, S. Rungarunlert, G. R. Souza and
J. N. Ferreira, Biomaterials, 2018, 180, 52.
191 A. Yamasaki, Y. Kunitomi, D. Murata, T. Sunaga,
T. Kuramoto, T. Sogawa and K. Misumi, J. Orthop. Res.,
2019, 37, 1398.
192 H. Kizawa, E. Nagao, M. Shimamura, G. Zhang and
H. Torii, Biochem. Biophys. Rep., 2017, 10, 186.
193 Y. Nakanishi, T. Okada, N. Takeuchi, N. Kozono, T. Senju,
K. Nakayama and Y. Nakashima, Regener. Ther., 2019,
11, 47.
194 D. Taniguchi, K. Matsumoto, T. Tsuchiya, R. Machino,
Y. Takeoka, A. Elgalad, K. Gunge, K. Takagi, Y. Taura,
G. Hatachi, N. Matsuo, N. Yamasaki, K. Nakayama and
T. Nagayasu, Interact. Cardiovasc. Thorac. Surg., 2018,
26, 745.
195 Y. Takeoka, K. Matsumoto, D. Taniguchi, T. Tsuchiya,
R. Machino, M. Moriyama, S. Oyama, T. Tetsuo,
Y. Taura, K. Takagi, T. Yoshida, A. Elgalad, N. Matsuo,
M. Kunizaki, S. Tobinaga, T. Nonaka, S. Hidaka,
N. Yamasaki, K. Nakayama and T. Nagayasu, PLoS One,
2019, 14, e0211339.
196 D. N. Heo, B. Ayan, M. Dey, D. Banerjee, H. Wee, G. S.
Lewis and I. T. Ozbolat, Biofabrication, 2021, 13, 015013.
197 B. Ayan, Y. Wu, V. Karuppagounder, F. Kamal and
I. T. Ozbolat, Sci. Rep., 2020, 10, 13148.
198 Y. Jo, D. G. Hwang, M. Kim, U. Yong and J. Jang, Trends
Biotechnol., 2023, 41, 93.
199 P. D. Dalton, T. B. F. Woodfield, V. Mironov and J. Groll,
Adv. Sci., 2020, 7, 1902953.
200 W. Kim and G. Kim, Bioeng. Transl. Med., 2022, 8, e10397.
201 Y. Bin, Z. Dongzhen, C. Xiaoli, E. jirigala, S. Wei, L. Zhao,
H. Tian, Z. Ping, L. Jianjun, W. Yuzhen, Z. Yijie,
F. Xiaobing and H. Sha, Bioact. Mater., 2022, 10, 247.
202 D. L. L. Ho, S. Lee, J. Du, J. D. Weiss, T. Tam, S. Sinha,
D. Klinger, S. Devine, A. Hamfeldt, H. T. Leng,
J. E. Herrmann, M. He, L. G. Fradkin, T. K. Tan,
D. Standish, P. Tomasello, D. Traul, N. Dianat, R. Ladi,
Q. Vicard, K. Katikireddy and M. A. Skylar-Scott, Adv.
Healthcare Mater., 2022, 11, 2201138.
203 A.C.Daly,M.D.DavidsonandJ.A.Burdick,Nat. Commun.,
2021, 12,753.
204 P. Rawal, D. M. Tripathi, S. Ramakrishna and S. Kaur, Bio-
Des. Manuf., 2021, 4, 627.
205 L. Pang, J. Ding, X.-X. Liu and S.-K. Fan, TrAC, Trends Anal.
Chem., 2019, 117, 291.
206 E. K. Sackmann, A. L. Fulton and D. J. Beebe, Nature, 2014,
507, 181.
207 J. Tien and Y. W. Dance, Adv. Healthcare Mater., 2021,
10, 2001028.
208 K. O. Rojek, M. C
´wiklin
´ska, J. Kuczak and J. Guzowski,
Chem. Rev., 2022, 122, 16839.
209 N. Del Piccolo, V. S. Shirure, Y. Bi, S. P. Goedegebuure,
S. Gholami, C. C. W. Hughes, R. C. Fields and S. C. George,
Adv. Drug Delivery Rev., 2021, 175, 113798.
210 M. Chung, J. Ahn, K. Son, S. Kim and N. L. Jeon, Adv.
Healthcare Mater., 2017, 6, 1700196.
211 M. S. Freag, B. Namgung, M. E. Reyna Fernandez,
E. Gherardi, S. Sengupta and H. L. Jang, Hepatol. Commun.,
2021, 5, 217.
212 Y. Gao, G. Stybayeva and A. Revzin, Lab Chip, 2019, 19, 306.
213 Y. Zheng, Z. Wu, M. Khan, S. Mao, K. Manibalan, N. Li,
J.-M. Lin and L. Lin, Anal. Chem., 2019, 91, 12283.
214 Z. Zhao, R. Li, H. Ruan, Z. Cai, Y. Zhuang, Z. Han,
M. Zhang, W. Cui and M. Cai, Chem. Eng. J., 2022, 436,
135176.
215 J. Yu, E. Berthier, A. Craig, T. E. de Groot, S. Sparks,
P. N. Ingram, D. F. Jarrard, W. Huang, D. J. Beebe and
A. B. Theberge, Nat. Biomed. Eng., 2019, 3, 830.
216 A. Mansoorifar, R. Gordon, R. C. Bergan and L. E.
Bertassoni, Adv. Funct. Mater., 2021, 31, 2006796.
217 G. Vunjak-Novakovic, K. Ronaldson-Bouchard and M. Radisic,
Cell, 2021, 184, 4597.
218 W. M. Liu, M. L. Sun, B. X. Lu, M. M. Yan, K. Han and
J. Y. Wang, Sens. Actuators, B, 2019, 292, 111.
219 M. Zhang, P. Wang, R. Luo, Y. Wang, Z. Li, Y. Guo, Y. Yao,
M. Li, T. Tao, W. Chen, J. Han, H. Liu, K. Cui, X. Zhang,
Y. Zheng and J. Qin, Adv. Sci., 2021, 8, 2002928.
220 Y. Guo, R. Luo, Y. Wang, P. Deng, T. Song, M. Zhang,
P. Wang, X. Zhang, K. Cui, T. Tao, Z. Li, W. Chen, Y. Zheng
and J. Qin, Sci. Bull., 2021, 66, 783.
221 Y. Zhao, E. Y. Wang, F. B. L. Lai, K. Cheung and M. Radisic,
Trends Biotechnol., 2023, 41, 410.
222 W. Liu, J. Song, X. Du, Y. Zhou, Y. Li, R. Li, L. Lyu, Y. He,
J. Hao, J. Ben, W. Wang, H. Shi and Q. Wang, Acta
Biomater., 2019, 91, 195.
223 A. Skardal, S. V. Murphy, M. Devarasetty, I. Mead, H. W.
Kang, Y. J. Seol, Y. Shrike Zhang, S. R. Shin, L. Zhao,
J. Aleman, A. R. Hall, T. D. Shupe, A. Kleensang,
M. R. Dokmeci, S. Jin Lee, J. D. Jackson, J. J. Yoo,
T. Hartung, A. Khademhosseini, S. Soker, C. E. Bishop
and A. Atala, Sci. Rep., 2017, 7, 8837.
Materials Horizons Review
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
This journal is © The Royal Society of Chemistry 2023 Mater. Horiz., 2023, 10, 4662–4685 | 4685
224 F. Yin, X. Zhang, L. Wang, Y. Wang, Y. Zhu, Z. Li, T. Tao,
W. Chen, H. Yu and J. Qin, Lab Chip, 2021, 21, 571.
225 H. J. Chen, P. Miller and M. L. Shuler, Lab Chip, 2018,
18, 2036.
226 N. Picollet-D’hahan, A. Zuchowska, I. Lemeunier and S. Le
Gac, Trends Biotechnol., 2021, 39, 788.
227 N. Sachs, J. de Ligt, O. Kopper, E. Gogola, G. Bounova,
F. Weeber, A. V. Balgobind, K. Wind, A. Gracanin,
H. Begthel, J. Korving, R. van Boxtel, A. A. Duarte,
D. Lelieveld, A. van Hoeck, R. F. Ernst, F. Blokzijl,
I. J. Nijman, M. Hoogstraat, M. van de Ven, D. A. Egan,
V. Zinzalla, J. Moll, S. F. Boj, E. E. Voest, L. Wessels,
P. J. van Diest, S. Rottenberg, R. G. J. Vries, E. Cuppen
and H. Clevers, Cell, 2018, 172, 373.
228 H. H. N. Yan, H. C. Siu, S. Law, S. L. Ho, S. S. K. Yue,
W. Y. Tsui, D. Chan, A. S. Chan, S. Ma, K. O. Lam,
S. Bartfeld, A. H. Y. Man, B. C. H. Lee, A. S. Y. Chan,
J. W. H. Wong, P. S. W. Cheng, A. K. W. Chan, J. Zhang,
J. Shi, X. Fan, D. L. W. Kwong, T. W. Mak, S. T. Yuen,
H. Clevers and S. Y. Leung, Cell Stem Cell, 2018, 23, 882.
229 R. Shi, N. Radulovich, C. Ng, N. Liu, H. Notsuda,
M. Cabanero, S. N. Martins-Filho, V. Raghavan, Q. Li, A. S.
Mer, J. C. Rosen, M. Li, Y. H. Wang, L. Tamblyn,
N. A. Pham, B. Haibe-Kains, G. Liu, N. Moghal and
M. S. Tsao, Clin. Cancer Res., 2020, 26, 1162.
230 L. Lei, B. Ma, C. Xu and H. Liu, TrAC, Trends Anal. Chem.,
2022, 153, 116640.
231 I. Holland, W. Shu and J. A. Davies, Biofabrication, 2023,
15, 045003.
232 W. Wei, W. Liu, H. Kang, X. Zhang, R. Yu, J. Liu, K. Huang,
Y. Zhang, M. Xie, Y. Hu and H. Dai, Adv. Healthcare Mater.,
2023, 12, e2300108.
233 C. G. Hubert, M. Rivera, L. C. Spangler, Q. Wu, S. C. Mack,
B. C. Prager, M. Couce, R. E. McLendon, A. E. Sloan and
J. N. Rich, Cancer Res., 2016, 76, 2465.
234 X. Qian, Y. Su, C. D. Adam, A. U. Deutschmann,
S. R. Pather, E. M. Goldberg, K. Su, S. Li, L. Lu, F. Jacob,
P. T. T. Nguyen, S. Huh, A. Hoke, S. E. Swinford-Jackson,
Z. Wen, X. Gu, R. C. Pierce, H. Wu, L. A. Briand, H. I. Chen,
J. A. Wolf, H. Song and G.-l Ming, Cell Stem Cell, 2020,
26, 766.
235 Z. Wang, S.-N. Wang, T.-Y. Xu, Z.-W. Miao, D.-F. Su and
C.-Y. Miao, CNS Neurosci. Ther., 2017, 23, 771.
236 K. Gehling, S. Parekh, F. Schneider, M. Kirchner, V. Kondylis,
C. Nikopoulou and P. Tessarz, Life Sci. Alliance, 2022,
5, e202101340.
237 J. Gong, Y. Gong, T. Zou, Y. Zeng, C. Yang, L. Mo, J. Kang,
X. Fan, H. Xu and J. Yang, Lab Chip, 2023, 23, 3820–3836.
238 I. T. Whelan, R. Burdis, S. Shahreza, E. Moeendarbary,
D. A. Hoey and D. J. Kelly, Biofabrication, 2023, 15, 034103.
239 G. Fang, Y. C. Chen, H. Lu and D. Jin, Adv. Funct. Mater.,
2023, 33, 2215043.
240 Z. Fang, Y. Ding, Z. Zhang, F. Wang, Z. Wang, H. Wang and
T. Pan, Lab Chip,2020, 20, 722.
241 Y. Nashimoto, R. Mukomoto, T. Imaizumi, T. Terai,
S. Shishido, K. Ino, R. Yokokawa, T. Miura, K. Onuma,
M. Inoue and H. Shiku, Biosens. Bioelectron., 2022, 219,
114808.
242 L. E. Niklason and J. H. Lawson, Science, 2020, 370, eaaw8682.
243 B. L. LeSavage, R. A. Suhar, N. Broguiere, M. P. Lutolf and
S. C. Heilshorn, Nat. Mater., 2022, 21, 143.
244 J. M. Unagolla and A. C. Jayasuriya, Appl. Mater. Today,
2022, 29, 101582.
245 D. Hendriks, J. F. Brouwers, K. Hamer, M. H. Geurts,
L. Luciana, S. Massalini, C. Lo
´pez-Iglesias, P. J. Peters,
M. J. Rodrı
´guez-Colman, S. Chuva de Sousa Lopes,
B. Artegiani and H. Clevers, Nat. Biotechnol., 2023, DOI:
10.1038/s41587-023-01680-4.
246 C. Chen, W. Ji and Y. Niu, Trends Biotechnol., 2021, 39,
1332.
Review Materials Horizons
Published on 31 August 2023. Downloaded on 11/1/2023 11:25:34 AM.
View Article Online
Article
Stem cell-based therapy is a novel technique used to reconstruct the ocular surface of patients with limbal stem cell deficiency (LSCD). However, the reported stem cell therapies for LSCD are limited to two-dimensional (2D) culture systems, which cannot mimic the native microenvironment in which stem cells reside in vivo. Herein, an injectable photocrosslinkable hydrogel was developed based on porous gelatin methacryloyl (GelMA)/silk fibroin glycidyl methacrylate (SilMA), which encapsulated limbal stem cells (LSCs) to reconstruct ocular surfaces. The porous GelMA/SilMA hydrogels demonstrated excellent transparency and good adhesion. More importantly, this hydrogel maintained viability, supported the adhesion of encapsulated rabbit LSCs and promoted their migration in vitro in three-dimensional (3D) culture environments. Proteomic analysis confirmed that the porous GelMA/SilMA hydrogel 3D cell culture system maintained both the self-renewal and differentiation of the LSCs. Furthermore, histological analyses demonstrated that the porous GelMA/SilMA hydrogel encapsulating LSCs effectively promoted the re-epithelization of LSCD. Porous hydrogels were subjected to simplified serial cell culture operations to construct an ocular surface with a uniform cell distribution. These findings provide valuable insights into stem cell therapies and regenerative medicine for ocular surface reconstruction.
Article
Full-text available
The bioassembly of layered tissue that closely mimics human histology presents challenges for tissue engineering. Existing bioprinting technologies lack the resolution and cell densities necessary to form the microscale cell-width layers commonly observed in stratified tissue, particularly when using low-viscosity hydrogels, such as collagen. Here we present rotational internal flow layer engineering (RIFLE), a novel, low-cost biofabrication technology for assembling tuneable, multi-layered tissue-like structures. Using high-speed rotating tubular moulds, small volumes of cell-laden liquids added to the inner surface were transitioned into thin layers and gelled, progressively building macroscale tubes composed of discrete microscale strata with thicknesses a function of rotational speed. Cell encapsulation enabled the patterning of high-density layers (108cells/ml) into heterogenous constructs. RIFLE versatility was demonstrated through tunica media assembly, encapsulating human smooth muscle cells in cell-width (12.5µm) collagen layers. Such deposition of discrete microscale layers, facilitates the biofabrication of composite structures mimicking the nature of native stratified tissue. This enabling technology has the potential to allow researchers to economically create a range of representative layered tissue.&#xD.
Article
Full-text available
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state‐of‐the‐art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large‐scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life‐like human tissue models that provide a platform to answer health‐based scientific questions.
Article
Full-text available
Bone is one of the key components of the musculoskeletal system. Bone and joint disease are the fourth most widespread disease, in addition to cardiovascular disease, cancer, and diabetes, which seriously affect people’s quality of life. Bone organoids seem to be a great model by which to promote the research method, which further could improve the treatment of bone and joint disease in the future. Here, we introduce the various bone and joint diseases and their biology, and the conditions of organoid culture, comparing the in vitro models among 2D, 3D, and organoids. We summarize the differing potential methods for culturing bone-related organoids from pluripotent stem cells, adult stem cells, or progenitor cells, and discuss the current and promising bone disease organoids for drug screening and precision medicine. Lastly, we discuss the challenges and difficulties encountered in the application of bone organoids and look to the future in order to present potential methods via which bone organoids might advance organoid construction and application.
Article
Full-text available
Endochondral ossification (EO) is an essential biological process than underpins how human bones develop, grow, and heal in the event of a fracture. So much is unknown about this process, thus clinical manifestations of dysregulated EO cannot be adequately treated. This can be partially attributed to the absence of predictive in vitro models of musculoskeletal tissue development and healing, which are integral to the development and preclinical evaluation of novel therapeutics. Microphysiological systems, or organ-on-chip devices, are advanced in vitro models designed for improved biological relevance compared to traditional in vitro culture models. Here we develop a microphysiological model of vascular invasion into developing/regenerating bone, thereby mimicking the process of EO. This is achieved by integrating endothelial cells and organoids mimicking different stages of endochondral bone development within a microfluidic chip. This microphysiological model is able to recreate key events in EO, such as the changing angiogenic profile of a maturing cartilage analogue, and vascular induced expression of the pluripotent transcription factors SOX2 and OCT4 in the cartilage analogue. This system represents an advanced in vitro platform to further EO research, and may also serve as a modular unit to monitor drug responses on such processes as part of a multi-organ system.
Article
Full-text available
The lack of registered drugs for nonalcoholic fatty liver disease (NAFLD) is partly due to the paucity of human-relevant models for target discovery and compound screening. Here we use human fetal hepatocyte organoids to model the first stage of NAFLD, steatosis, representing three different triggers: free fatty acid loading, interindividual genetic variability (PNPLA3 I148M) and monogenic lipid disorders (APOB and MTTP mutations). Screening of drug candidates revealed compounds effective at resolving steatosis. Mechanistic evaluation of effective drugs uncovered repression of de novo lipogenesis as the convergent molecular pathway. We present FatTracer, a CRISPR screening platform to identify steatosis modulators and putative targets using APOB−/− and MTTP−/− organoids. From a screen targeting 35 genes implicated in lipid metabolism and/or NAFLD risk, FADS2 (fatty acid desaturase 2) emerged as an important determinant of hepatic steatosis. Enhancement of FADS2 expression increases polyunsaturated fatty acid abundancy which, in turn, reduces de novo lipogenesis. These organoid models facilitate study of steatosis etiology and drug targets.
Article
Full-text available
Multicellular spheroids and organoids are promising in vitro 3D models in personalized medicine and drug screening. They replicate the structural and functional characteristics of human organs in vivo. Microfluidic technology and micro‐nano fabrication can fulfill the high requirement of the engineering approach in the growing research interest in spheroids and organoids. In this review, spheroids and organoids are comparatively introduced. Then it is illustrated how spheroids‐ and organoids‐on‐a‐chip technology facilitates their establishment, expansion, and application through spatial‐temporal control, mechanical cues modeling, high‐throughput analysis, co‐culture, multi‐tissue interactions, biosensing, and bioimaging integration. The potential opportunities and challenges in developing spheroids‐ and organoids‐on‐a‐chip technology are finally outlooked.
Article
Full-text available
Osteochondral defect (OCD) regeneration remains challenging because of the hierarchy of the native tissue including both the articular cartilage and the subchondral bone. Constructing an osteochondral scaffold with biomimetic composition, structure, and biological functionality is the key to achieve its high‐quality repair. In the present study, an injectable and 3D printable bilayered osteochondral hydrogel based on compositional gradient of methacrylated sodium alginate, gelatin methacryloyl, and β‐tricalcium phosphate (β‐TCP), as well as the biochemical gradient of kartogenin (KGN) in the two well‐integrated zones of chondral layer hydrogel (CLH) and osseous layer hydrogel (OLH) is developed. In vitro and subcutaneous in vivo evaluations reveal that apart from the chondrogenesis of the embedded bone mesenchymal stem cells induced by CLH with a high concentration of KGN, a low concentration of KGN with β‐TCP in the OLH synergistically achieves superior osteogenic differentiation by endochondral ossification, instead of the intramembranous ossification using OLH with only β‐TCP. The biomimetic construct leveraging KGN as the only biochemical inducer can facilitate cartilage and subchondral bone restoration in the in vivo osteochondral defect. This one‐stone‐two‐birds strategy opens up a new facile approach for OCD regeneration by exploiting the biological functions of the bioactive drug molecule KGN.
Article
Tissue-engineered articular cartilage constructs are currently not able to equal native tissues in terms of mechanical and biological properties. A major cause lies in the deficiency in engineering the biomechanical microenvironment (BMME) of articular chondrocytes. In this work, to engineer the BMME of articular chondrocytes, heterogeneous hydrogel structures of gelatin methacrylated (GelMA) containing differential-stiffness domains were first fabricated, and then periodic dynamic mechanical stimulations were applied to the hydrogel structures. The chondrocyte phenotype of ATDC5 cells was enhanced as the spatial differentiation in stiffness was increased in the hydrogel structures and was further strengthened by dynamic mechanical stimulation. It was speculated that the mechanical signals generated by the engineered BMME were sensed by the cells through the integrin β1-FAK signaling pathway. This study revealed the key role of the combined effects of differential and dynamic BMME on the chondrocyte phenotype, which could provide theoretical guidance for highly active tissue-engineered articular cartilage.
Article
Retinal organoids (ROs) derived from human pluripotent stem cells (hPSCs) have become a promising model in vitro to recapitulate human retinal development, which can be further employed to explore the mechanisms of retinal diseases. However, the current culture systems for ROs lack physiologically relevant microenvironments, such as controllable mechano-physiological cues and dynamic feedback between cells and the extracellular matrix (ECM), which limits the accurate control of RO development. Therefore, we designed a controllable perfusion microfluidic chip (CPMC) with the advantages of precisely controlling fluidic shear stress (FSS) and oxygen concentration distribution in a human embryonic stem cell (hESC)-derived RO culture system. We found that ROs cultured under this system allow for expanding the retinal progenitor cell (RPC) pool, orchestrating the retinal ganglion cell (RGC) specification, and axon growth without disturbing the spatial and temporal patterning events at the early stage of RO development. Furthermore, RNA sequencing data revealed that the activation of voltage-gated ion channels and the increased expression of ECM components synergistically improve the growth of ROs and facilitate the differentiation of RGCs. This study elaborates on the advantages of the designed CPMC to promote RO growth and provide a controllable and reliable platform for the efficient maturity of RGCs in the ROs, promising applications in modeling RGC-related disorders, drug screening, and cell transplantation.
Article
We review the emergence of the new field of organ-on-a-chip (OOAC) engineering, from the parent fields of tissue engineering and microfluidics. We place into perspective the tools and capabilities brought into the OOAC field by early tissue engineers and microfluidics experts. Liver-on-a-chip and heart-on-a-chip are used as two case studies of systems that heavily relied on tissue engineering techniques and that were amongst the first OOAC models to be implemented, motivated by the need to better assess toxicity to human tissues in preclinical drug development. We review current challenges in OOAC that often stem from the same challenges in the parent fields, such as stable vascularization and drug absorption.