Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Materials and Design 203 (2021) 109621

Contents lists available at ScienceDirect

Materials and Design

journal homepage: www.elsevier.com/locate/matdes

3D bioprinting modified autologous matrix-induced chondrogenesis


(AMIC) technique for repair of cartilage defects
Yang Zhou 1, Ran Qin 1, Tong Chen, Kaibin Zhang, Jianchao Gui ⁎
Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, PR China

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• Modified autologous matrix-induced


chondrogenesis to repair cartilage
defects.
• 3D printed biofilms combined
chondrogenic progenitor cells.
• Fibronectin was added to improve
the biological function of composite
hydrogel.
• A stable postoperative microenviron-
ment for bone mesenchymal stem cells.
• Reproducing layered structure similar
to natural cartilage.

a r t i c l e i n f o a b s t r a c t

Article history: Three-dimensional (3D) printing technology, has achieved good results in articular cartilage damage repair, yet
Received 21 October 2020 regeneration of hyaline cartilage similar to native cartilage and effectively performing clinical transformation re-
Received in revised form 26 January 2021 main challenging. In this study, we used 3D bioprinting technology to add chondrogenic progenitor cells (CPCs)
Accepted 27 February 2021
and fibronectin (FN) to an alginate/gelatin/hyaluronic acid (Alg/Gel/HA) composite hydrogel. We used this hy-
Available online 5 March 2021
drogel to prepare an active biofilm with uniform pores using modified autologous matrix-induced chondrogen-
Keywords:
esis (AMIC) technology to effectively repair cartilage defects. The Alg/Gel/HA composite hydrogel combined with
AMIC FN promoted the chondrogenic differentiation of CPCs by upregulating their gene expression of collagen II, SOX9,
3D bioprinting and especially PRG4. Adjacent biofilm provided adequate mechanical support and architectural integrity, offering
Cartilage repair a stable postoperative microenvironment for bone mesenchymal stem cells (BMSCs) released from subchondral
Chondrogenic progenitor cells bone, and ensured that a laminar structure similar to natural hyaline cartilage was regenerated in the rat cartilage
Fibronectin defect model. Characteristic cartilage-like lacuna structures and a gradient structure were observed in the AMIC
Gradient structure +FN + CPCs group. We developed an effective method to regenerate full-thickness cartilage defects, this
biofunctionalized cell-laden biofilm has great potential for application as a supplement to traditional AMIC tech-
nology to improve the quality of cartilage regeneration in a relatively feasible way.
© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://
creativecommons.org/licenses/by-nc-nd/4.0/).

Abbreviations: Three-dimensional, 3D; Autologous matrix induced chondrogenesis, AMIC; Chondrogenic progenitor cells, CPCs; Fibronectin, FN; Sodium alginate, Alg; Gelatin, Gel;
Hyaluronic acid, HA; Proteoglycan 4, PRG4; COL2, Type 2 collagen; Bone mesenchymal stem cells, BMSCs; Osteoarthritis, OA; Autologous chondrocyte implantation, ACI; Microfracture,
MF; Hydroxyapatite, HAP; Extracellular matrix, ECM; Scanning electron microscopy, SEM; ANOVA, Analysis of variance; Interpenetrating Polymer Network, IPN; Enzyme linked
immunosorbent assay, ELISA.
⁎ Corresponding author at: Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, PR China.
E-mail address: gui1997@126.com (J. Gui).
1
These authors contributed equally to this work and should be considered co-first authors.

https://doi.org/10.1016/j.matdes.2021.109621
0264-1275/© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

1. Introduction collagen fibers and exhibits homology with collagen [23]. It can not
only replace the collagen component in the cartilage extracellular ma-
Osteoarthritis (OA) is one of the most common diseases in middle- trix, but also enhance the hardness and viscosity of the Alg-based scaf-
aged and elderly people. It is a chronic and progressive degenerative fold and enhance its printability. HA is an acidic mucopolysaccharide
disease characterized by the degeneration and destruction of articular macromolecular substance and is widely present in connective tissues
cartilage [1]. Articular hyaline cartilage maintains very low metabolic of humans and animals. It can regulate various physiological functions
activity, and lacks distribution of blood vessels, nerves, and lymph ves- of cells as well as cell aggregation during tissue formation, and possesses
sels. Once damaged, it lacks the capacity to heal itself, and the damaged significant chondrogenic effects that show its application value in carti-
cartilage will accelerate degeneration of the affected joints and the lage tissue engineering [24]. Meanwhile,HA is widely present in carti-
onset of OA [2]. Therefore, effectively repairing early focal damage to ar- lage tissue and has a CD44 recognition site, which can specifically
ticular cartilage, controlling its degeneration, and rebuilding a durable recognize cartilage cells and maintain the chondrocyte phenotype
articular cartilage surface can effectively delay or prevent the occur- [25]. Fibrin-based bioinks also usually blend with other materials such
rence of OA [3]. Traditional articular cartilage repair techniques include as gelatin, collagen, and alginate [26]. Therefore, in this study, we pre-
osteochondral transplantation, autologous chondrocyte implantation pared a composite hydrogel based on Alg, Gel, and HA, which improves
(ACI), osteotomy, microfracture technology (MF), and autologous upon the defects of a single Alg hydrogel and has good printability. It can
matrix-induced chondrogenesis (AMIC) technology [4,5]. However, be used for extrusion 3D printing to prepare active biofilms for modified
the repair effects are limited and lack long-term efficacy. Although AMIC technology.
they can promote the formation of new articular surfaces, the newly Although 3D bioprinting is expected to lead to a new generation of
formed tissue is not necessarily hyaline cartilage [6]. cartilage regeneration therapy, it remains a huge challenge to regener-
The microfracture technique involves drilling a hole in the bone sur- ate hyaline cartilage similar to native cartilage and reconstruct a layered
face so that blood from the bone marrow containing bone mesenchymal structure with both mechanical and biological characteristics similar to
stem cells (BMSCs) will ooze out to repair cartilage [7]. Because the local native cartilage [27]. The regeneration is driven by cells encapsulated
microenvironment after surgery cannot recruit and guide the differenti- within the hydrogel. After implantation, through interaction with the
ation of BMSCs, this results in regeneration of essentially fibrous carti- host's biomechanical and biochemical environment, cartilage regenera-
lage, the strength of which is significantly lower than that of native tion is achieved over time. Therefore, it is very important to use cells
hyaline cartilage [8]. Therefore, improved microfracture technology with good regeneration ability in the bioink. Traditional seed cells
was proposed in the clinic, and it was named AMIC technology, which such as chondrocytes are associated with various problems such as lim-
provides a solution by combining microfracture with a collagen I/III ma- ited availability, low proliferative ability, and dedifferentiation during
trix applied to the cartilage defects. Compared with the microfracture culture expansion [28]. Mesenchymal stem cells can be obtained from
technique, this method can effectively and rapidly relieve pain, restore a wide range of sources and can be induced to differentiate into
joint function, successfully restore motor function, and has a relatively chondrocytes. They have strong reproductive capacity and good prolif-
long-term effect [9,10]. However, AMIC technology still cannot eration and differentiation activity, but they also have problems such
completely repair cartilage defects, and regeneration of hyaline cartilage as Chondrogenic progenitor cells (CPCs) are new seed cells for cartilage
with a gradient structure similar to native cartilage is still an urgent tissue engineering. CPCs can be separated by differential adhesion to fi-
problem to be solved [11]. bronectin (FN) [29]. They have similar characteristics to the self-cloning
In recent years, 3D printing technology has gradually spawned a and proliferation of stem cells. They are primitive cells located in carti-
new generation of cartilage repair techniques. 3D printing technology lage tissue and have the ability to self-proliferate as well as having the
can accurately control the internal structure of the scaffold, construct a potential to differentiate into chondrocytes by themselves [30,31]. FN
cartilage-like structure, and control the internal pore size to achieve is a high molecular weight (450 kDa) glycoprotein that promotes cell–
customization according to cartilage defects. 3D bioprinting can also cell and cell–substrate adhesion and cell migration, which are necessary
print hydrogel embedded with cells or active cytokines, and provide a to maintain cell structure and function [32]. In our previous study, we
new direction for constructing in vivo cartilage grafts [12]. In recent isolated CPCs through FN differential adhesion experiments and evalu-
years, there have been many advances in hydrogel-based 3D printing ated their stemness, proving that FN can promote the proliferation, mi-
for tissue engineering [13]. Recently, a direct coaxial 3D bioprinting sys- gration, and chondrogenesis of CPCs through integrin α5β1-dependent
tem was reported by Shao et al., who prepared a cell-laden structure signaling, thereby increasing the synthesis of both protein and RNA
with effective vascularized nutrient delivery channels [14]. Dalya et al. [33,34]. However, little is known about the applicability of CPCs in car-
prepared spatially organized tissues by directing the growth of cellular tilage tissue engineering, especially in combination with biomaterials
spheroids within 3D-printed polymeric microchambers to mimic the and 3D printing [35].
development process of native cartilage [15]. As the most widely used To address these issues, this research aimed to realize the co-
material for deposition 3D printing, hydrogel has many advantages, printing of FN, CPCs, and Alg/Gel/HA composite hydrogel through extru-
such as a similar structure to human soft tissues and high water content sion 3D bioprinting to prepare an active biofilm. This was then used to
[16]. Commonly used natural hydrogel materials, such as sodium algi- modify AMIC technology, by using this active biofilm combined with
nate (Alg), a natural polysaccharide, have advantages such as a wide va- microfracture technology to improve the quality of regenerated carti-
riety of sources, low price, and good histocompatibility and lage for the repair of articular cartilage damage. First, the Alg/Gel/HA
biodegradability [17]. It can quickly undergo ion exchange cross- composite hydrogel had a better cartilage-promoting effect compared
linking when encountering cations and has been widely used in carti- with AMIC technology using a single I/III collagen membrane. The uni-
lage tissue engineering [18]. However, Alg hydrogel does not contain form porosity and good structure of the active biofilm was conducive
any RGD molecules due to its natural sources, and consequently it to exchange of nutrients and communication between cells, effectively
lacks cell adhesion sites. In addition, its mechanical strength is poor, promoting growth and proliferation, and providing certain mechanical
and it is difficult to realize a complex three-dimensional structure. support before the generation of new cartilage [36]. The bioactive bio-
Therefore, Alg-based hydrogels have been used, supplemented with film effectively recruited BMSCs and provided a good microenviron-
an ECM component such as hyaluronic acid (HA), hydroxyapatite ment to guide the BMSCs to differentiate into chondrocytes through
(HAP) or gelatin (Gel) [19–21] . Krishna C. R. Kolan et al. investigated their interaction with biofilms [37,38]. FN can also promote the prolifer-
the alginate-gelatin composite bioink modified with bioactive glass ation and differentiation of chondrogenic progenitor cells through
and the feasibility of the solvent-based 3D bioprinting technique for tis- integrin α5β1-dependent signaling pathways. At the same time,
sue engineering applications [22]. Gel is taken from natural biological BMSCs stimulate CPCs through paracrine signals, promoting the

2
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

proliferation of CPCs and the production of total chondrocyte extracellu- CPCs were isolated in accordance with previously described method
lar matrix [15]. We evaluated the function of this biofilm in vitro and in [40]. Briefly, an aliquot of the freshly isolated cartilage cells was pelleted
rat animal models. In general, this active biofilm combined the excellent by centrifugation, suspended in serum-free DMEM/F12, and plated into
properties of Gel, Alg, and HA, had good biocompatibility, good mechan- FN-coated tissue culture plates, at a density of 500 cells cm−2. After
ical properties, and suitable uniform porosity. At the same time, the ad- 20 min, non-adherent cells were removed, and the attached cells were
dition of CPCs and FN improved its ability to regenerate cartilage. The cultured in DMEM containing 10% FBS and 1% penicillin/streptomycin
biofilm offered a stable microenvironment for BMSCs and newly formed at 37 °C with 5% CO2. Cells were expanded until passage two before
tissue, possessing the typical layered structure characteristic of natural being used for the study.
hyaline cartilage, was regenerated in the rat cartilage defect. The find-
ings of this study can be used to improve AMIC technology to repair car-
2.4. 3D bioprinting and characterization of cell encapsulated constructs
tilage defects, providing a theoretical basis for future clinical
transformation [39].
To prepare the cell-laden bio-ink for bioprinting, 2.5 g Gel, 1.25 g Alg,
and 0.1 g HA were each add to 30 mL DMEM basic medium and heated
2. Materials and methods and stirred at 37 °C until fully dissolved to form a 5% Gel +2.5%
Alg + 0.2% HA hydrogel precursor solution (w/v). Then, FN was added
2.1. Materials to the solution to a final concentration of 20 μg/mL. Finally, the cell sus-
pension and the pre-prepared hydrogel solution were evenly mixed at a
Alg from marine algae and FN were purchased from Sigma-Aldrich volume ratio of 2:3 to obtain a cell-laden bio-ink with a cell concentration
(St Louis, MO, USA). Gel was purchased from Shanghai Aladdin Bio- of 106 cells/mL. Gel, Alg, HA, and CaC2l powder were disinfected with UV
chemical Co. Ltd. (Shanghai, China). HA was purchased from Shanghai for more than 2 h, and all operations were performed on a clean bench.
Macklin Biochemical Co. Ltd. (Shanghai, China). Biofilm scaffolds were fabricated using a pneumatic 3D Bioprinter
(EFL-BP-6601, Yongqinquan Intelligent Equipment Co., Ltd., Suzhou,
2.2. Preparation and characterization of Alg/Gel/HA composite hydrogels China) using a direct extrusion-based technique. Temperature control-
lers were placed around the syringe and nozzle to ensure a homoge-
To prepare the composite hydrogel, 2.5 g Gel, 1.25 g Alg and 0.1 g HA neous plotting temperature, while a cooling system was also
were each added to 50 mL deionized water and heated and stirred at implemented under the receiving platform to maintain a stable temper-
37 °C until fully dissolved, forming a 5% Gel +2.5% Alg + 0.2% HA hydro- ature after the deposition of the cell-laden bioink. Cell-laden bioink was
gel precursor solution (w/v). Keeping other conditions constant, the loaded into the printing cartridge, and extruded onto the pre-cooling
2.5 g Gel was replaced with 4 g and 5 g respectively, and the above platform. The entire printing system was contained within a plexiglass
steps were repeated to obtain 8% Gel +2.5% Alg + 0.2% HA and 10% box to prevent temperature fluctuations. The specific printing parame-
Gel +2.5% Alg + 0.2% HA hydrogel precursor solutions (w /v). ters of hydrogels with different concentration ratios are given in
Table 1. Scaffolds for in vitro biological assessment had dimensions of
2.2.1. Rheological characterization 10 mm (length) × 10 mm (width) × 1 mm (height) (Fig. 1). Particularly
The rheological properties of the bioink were analyzed using a rhe- in animal experiments, biofilms were fabricated with a disk shape and a
ometer (DHR-2, TA Instruments, New Castle, DE, USA) with a 20 mm diameter of 2 mm to maximize the covered area in the cartilage defect.
aluminum fixture. For the thermosensitive process, the oscillation
model was adopted. The test temperature range was 5–37 °C, the tem- 2.4.1. Scanning electron microscopy
perature reduction rate was 5 °C/min, the strain was 1%, and the fre- The biofilm scaffolds were lyophilized, and the cross-section was ob-
quency was 1 rad/s. The gel point was determined as the time when served with a scanning electron microscope (JSM-IT100, Jeol Ltd.,
the storage modulus (G') surpassed the loss modulus (G"). The mea- Tokyo, Japan).
surements of shear stress were performed by varying the shear rate
from 1 to 600 s−1 with rotational tests at 37 °C. A frequency sweep 2.4.2. Viability evaluation of cells
was performed and by varying the angular frequency from 0.1 to To evaluate the viability of CPCs in biofilm, a live/dead staining kit
100 rad/s at 37 °C, the strain was 1%. (KeyGEN BioTECH Co., Ltd., Nanjing, China) was used according to the
manufacturer's instructions. After culturing the printed biofilm for 1,
2.2.2. Mechanical tests 7, and 14 days, the cells in biofilm were stained. Then, the cell-laden
Mechanical properties of the Alg/Gel/HA composite hydrogels with scaffolds were observed and imaged under a confocal fluorescence mi-
different concentration ratios were measured using a dynamic mechan- croscope (Olympus FV3000, Tokyo, Japan). Calcein AM-labeled live
ical analysis instrument (ElectroForce, TA Instruments). Briefly, the hy- cells emit green fluorescence, and propidium iodide (PI)-labeled dead
drogel was soaked in 4% CaCl2 solution for 10 min to undergo ionic cells emit red fluorescence. Under a confocal microscope, multi-layer
crosslinking into a round pie-like structure for compression testing. fluorescence images were captured, and the software Imaris x64 7.2.1
Three groups were tested, each sample was placed between two com-
pression plates, and the compressive speed was set at 1 mm/min. The
compressive modulus was calculated according to the slope of the linear Table 1
region in the 5–10% strain range of the stress−strain curve. Primers sequences used in qRT-PCR.

Gene Primer sequences


2.3. Isolation and expansion of BMSCs and CPCs GAPDH F:5′-CAACTCCCTCAAGATTGTCAGCAA- 3′
R: 5′-GGCATGGACTGTGGTCATGA- 3′
BMSCs were harvested from 2-week-old male Sprague Dawley (SD) Collagen II F: 5′-CAACACTGCCAACGTCCAGAT- 3′
R: 5′-TCTTGCAGTGGTAGGTGATGTTCT- 3′
rats. Specifically, bone marrow aspirates were collected from the femur
PRG4 F: 5′-CACAATGAAGTCAAAGTGAGCA- 3′
of each rat and mononuclear cells were separated by gradient centrifu- R: 5′-CTAATGTTGGGCAGTGTTATCG- 3′
gation and then plated into 100 mm dishes. Cells were cultured in SOX9 F: 5′-CTTCCGCGACGTGGACAT- 3′
DMEM (Hyclone, Logan, UT, USA) containing 10% FBS and 1% penicil- R: 5′-GTTGGGCGGCAGGTACTG- 3′
lin/streptomycin at 37 °C with 5% CO2. BMSCs grown to passage three Acan F: 5′- AACTCAGTGGCCAAACATCC-3′
R: 5′-TCAGGAATCCCAGATGTTCC-3′
were used in the present study.

3
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Fig. 1. (A) Schematic diagram of the hydrogel biofilm model and specific dimensions. (B) Biofilm printing effects of three different ratio hydrogels. (C) Demonstration of 3D printability of
5% Gel +2.5% Alg + 0.2% HA hydrogel and photograph of the printed scaffolds with various shapes: branched vessel and meniscus.

was used for three-dimensional reconstruction. Cell viability was de- were fixed in 4% paraformaldehyde at room temperature for 30 min and
tected at 1, 7 and 14 days, with 3 specimens at each time point. Image then stained with tissue-specific markers. Briefly, after fixation and
J software was used to count the number of living/dead cells. For the blocking of non-specific antigen, bioprinted constructs were incubated
case where multiple cells overlap and affect the statistical accuracy, overnight at 4 °C with one of the following primary antibodies: rabbit
the watershed method was adopted in this experiment to improve the monoclonal anti-Lubrin/PRG4 (1:500, Abcam, Cambridge, UK) and
statistical accuracy. Click the statistical particles under the analysis to anti-SOX9 (1:500, Abcam), and rabbit polyclonal anti-Collagen II
get the number of live/dead cells, and 4 non-overlapping visual fields (1:500, Bioss Antibodies, Woburn, MA, USA). After washing, they were
were randomly selected for each experimental specimen for detection. then incubated with a red fluorophore-labeled mouse anti-rabbit sec-
ondary antibody (1:500) for 2 h in the dark at room temperature. Fi-
2.4.3. Cell morphological observations nally, incubated sections were mounted in DAPI (Servicebio) and
Cells were seeded onto the biofilm and cultured for 1 and 14 days pictures were taken with a scanning laser confocal microscope (Olym-
and then rinsed using PBS and fixed in 4% paraformaldehyde solution pus FV3000) within 24 h.
for 30 min. Subsequently, after washing three times with PBS, the cells The 3D-printed biofilms were cultured in DMEM, and after 7 days of
were permeabilized with 0.1% Triton X-100 for 10 min and stained culture, chondrogenic medium (Cyagen Biosciences Inc., Guangzhou,
with TRITC phalloidin (Yeasen Biotech Co., Ltd., Shanghai, China) at a di- China) was added for continuous induction and culture for 21 days. Sul-
lution of 1:1000 in PBS for 30 min in the dark. After washing three times fated glycosaminoglycans (GAGs) were stained with 0.5% Alcian blue
with PBS, DAPI (1:500) (Solarbio, Beijing, China) was applied to stain (Sigma, pH 1) for 10 min at room temperature.
nuclei for 10 min. Finally, the cell-laden scaffolds were washed with
PBS and imaged under a confocal fluorescence microscope (Olympus 2.6. qRT-PCR determination of gene expression levels in vitro
FV3000), and the software Imaris x64 7.2.1 was used for three-
dimensional reconstruction. The gene expression levels of collagen II, PGR4 and SOX9 were ana-
lyzed by qRT-PCR. Briefly, biofilms were cultured for 3, 7, or 14 days,
2.4.4. Fibronectin release kinetics then lysis solution containing 1.62% sodium citrate, 0.585% EDTA and
To assess the release profile of FN from printed biofilm, a rat FN 0.88% sodium chloride was used to lyse the biofilm to release cells.
ELISA kit was used (Cusabio, Wuhan, China). Briefly, printed biofilms Total RNA was extracted from cartilage using Trizol reagent (Invitrogen,
were placed into a 12-well plate, followed by addition of 200 μL of Carlsbad, CA, USA) and reverse-transcribed with PrimeScript RT Master
PBS. The PBS was collected and replaced with fresh solution every Mix (Perfect Real Time; TaKaRa Bio, Japan) following the manufacturer's
2 days for 2 weeks. The collected PBS was stored at −80 °C. Finally, protocol. Quantitative real-time RT-PCR amplifications were performed
the content of FN was quantified using a rat-specific ELISA kit according as described. The primers used in this study are listed in Table 2.
to the manufacturer's instructions.
2.6.1. Effects of bioprinted biofilm on differentiation of BMSCs
2.5. In vitro biological assessment An indirect contact co-culture model was applied to study the im-
pact of Alg/Gel/HA + CPCs+FN biofilm on differentiation of BMSCs re-
2.5.1. Immunofluorescence and Alcian blue staining leased by microfracture, using Transwell culture plates (Corning, NY,
To detect secretion of cartilage-specific extracellular matrix by em- USA). In short, rat BMSCs were seeded at a density of 5 × 105 cells per
bedded CPCs, after culturing for 7 or 14 days, the bioprinted constructs well into wells of 6-well culture plates. Then, Transwells culture plates

4
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Table 2
The specific printing parameters of hydrogels with different concentration ratios.

Printing speed Printing pressure Nozzle temperature Platform temperature Inter layer increment Nozzle diameter Inter filament distance

5%Gel+2.5%Alg+0.2%HA 360 mm/min 18 ~ 19kpa 37 °C 5 °C 0.18 mm*2 260 μm 1.1 mm


8%Gel+2.5%Alg+0.2%HA 360 mm/min 37 ~ 38kpa 37 °C 5 °C 0.18 mm*2 260 μm 1.1 mm
10%Gel+2.5%Alg+0.2% 360 mm/min 41 ~ 42kpa 37 °C 5 °C 0.18 mm*2 260 μm 1.1 mm
HA

loaded with printed biofilm were suspended over the wells seeded with using Student's t-test or two-way ANOVA for comparing different
BMSCs. The BMSCs and biofilm were fed with 5 mL DMEM containing groups. Values of P < 0.05 were considered statistically significant.
10% FBS and 1% penicillin/streptomycin. BMSCs were seeded at the
same density in six-well culture plates without biofilm as the control 3. Results and discussion
group.
To assess the impact of biofilm on chondrogenic differentiation of In this study, we developed a bioactive cell-laden biofilm combined
BMSCs, Alcian blue staining was performed on BMSCs after 7 days. with FN to improve AMIC technology, accelerating cartilage regenera-
qRT-PCR was then utilized to test the gene expression of chondrogenic tion, and confirmed its repair effect of cartilage defects. Previous studies
differentiation markers (COLII, aggrecan (Acan), and SOX9) in the have demonstrated that covering a microfractured area could result in
BMSCs cultured with biofilm. After cultured for 7 or 14 days, total RNA better cartilage regeneration [41]. Our combination AMIC technique
was extracted from the cell layer using Trizol reagent (Invitrogen, Carls- promoted cartilage regeneration and accumulation of the cartilage-
bad, CA, USA) and reverse-transcribed with PrimeScript RT Master Mix specific extracellular matrix, showing well integration with the sur-
(Perfect Real Time; TaKaRa Bio Inc., Shiga, Japan) following the manu- rounding normal cartilage, as compared to the traditional MF and
facturer's protocol. Quantitative RT-PCR amplifications were performed AMIC technique. In the development and application of composite hy-
as described above. drogel scaffolds, based on 3D printing technology, it is of great signifi-
cance to integrate the complementary properties of materials and
2.7. Articular cartilage defect models have a good ability to induce cartilage differentiation. As a natural poly-
saccharide, Alg has good biocompatibility and biodegradability, and can
SD rats were provided by the Animal Center of Nanjing Medical Uni- quickly gel by chelating with Ca2+. However, it lacks the corresponding
versity. All animal protocols were approved by the animal ethics com- cell adhesion sites and the ability to induce cartilage differentiation [42].
mittee of Nanjing Medical University. In this experiment, 50 male SD Gel, as a degradation product of collagen widely present in cartilage tis-
rats aged 3 months were utilized. A total of five groups were evaluated, sue, has a large number of RGD binding sites and has strong adhesion to
ten rats per group: microfracture only, AMIC+, AMIC+FN, AMIC+CPCs, cells. Therefore, Alg/Gel-based hydrogels have been widely used in car-
and AMIC+FN + CPCs. A full-thickness cylindrical cartilage defect tilage tissue engineering [43]. Hyaluronic acid is widely present in carti-
(2 mm in diameter and 1 mm in depth) was created on the trochlear lage tissue and has been widely used in the treatment of osteoarthritis
groove of each distal femur using a drill. In MF groups, a 0.5 mm K- in clinic.
wire was used to drill three approximately 3 mm holes. In other four
AMIC-modified biofilm groups, 3D-printed Alg/Gel/HA biofilm only 3.1. Synthesis and characterization of composite hydrogel
(AMIC+) or containing FN (AMIC+FN), chondroprogenitor cells
(AMIC+CPCs), or FN and CPCs (AMIC+FN + CPCs), was placed over To develop a suitable biofilm to replace collagen membrane and im-
the cartilage defect site and secured with fibrin glue (Guangzhou Beixiu prove the cartilage repair effect of AMIC technology, it is necessary to
Biotechnology Co., Ltd., Guangzhou, China). After the operation, rats develop a material which provides both a soft and stimulatory environ-
were allowed to move freely in their cages and fed with standard food ment for cells (both CPCs and BMSCs) to maintain their chondrogenic
and water. 6 or 12 weeks later, rats were sacrificed for further study. phenotype, while also providing a certain mechanical support before
After sacrifice, damaged cartilage repair site of femoral bottom was neocartilage regeneration, after which the biofilm will be completely
collected and then general observation and photograph were per- degraded and eventually replaced by new cartilage. In this work, in
formed. The quality of cartilage repair was assessed by three blinded in- order to improve the biological activity and achieve a good cartilage re-
dependent researchers using Wakitani's histological scoring standard. generation effect, HA was added to Alg/Gel-based hydrogel to prepare
All specimens were fixed with 4% paraformaldehyde, embedded in par- an Alg/Gel/HA composite hydrogel. Using an Alg, Gel and HA
affin and sectioned for histological and immunohistochemical analysis. prepolymer mixture, a multimaterial Interpenetrating Polymer Net-
Sections were stained according to previously established methods work (IPN) hydrogel was fabricated through temperature change and
with hematoxylin and eosin (HE), Safranin-O, and anti-type II collagen Ca2+ mediated dual-crosslinking (Scheme 1). To optimize the polymer
to evaluate the histological structure and cartilage ECM deposition in blend for use as a bioink for depositional 3D bioprinting, Gel concentra-
the regenerated cartilage. tions of 5, 8, and 10% were investigated, while the Alg concentration was
Besides, qRT-PCR was used to analyze the gene expression of specific fixed at 2.5% and the HA concentration was fixed at 0.2%. Both concen-
differentiation makers in each groups. Total RNA was extracted from trations were previously reported as being ideal for the fabrication of
cartilage using Trizol reagent (Invitrogen) and reverse-transcribed hydrogels with suitable toughness and enhanced promotion of
with PrimeScript RT Master Mix (Perfect Real Time; TaKaRa Bio) follow- chondrogenic differentiation for cartilage engineering. Bioink was pre-
ing the manufacturer's protocol. Quantitative real-time RT-PCR amplifi- pared by compounding Alg, Gel and HA, which improved the disadvan-
cations were performed as described in Section 2.5.2. tages of poor cell adhesion of Alg hydrogel and inhibited the rapid
degradation of HA to a certain extent. We tested the effect of different
2.8. Statistical analysis Gel concentrations on the performance of the composite hydrogel, and
the results showed that the 3D-printed biofilm prepared from the hy-
All analyses were carried out using GraphPad Prism 6.0 software drogel containing 5% Gel has the best formability.
(GraphPad Software, Inc., La Jolla, CA, USA). All data are shown as the In the process of cooling from 37 °C to 5 °C, the sample storage mod-
mean ± standard deviation (SD) and comparisons were carried out ulus (G′) was smaller than its loss modulus (G″) before the phase

5
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Scheme 1. Schematic illustration of 3D printing of chondrogenic progenitor cells (CPCs) and fibronectin (FN) encapsulated within biofilm for repair of cartilage defects. The experimental
process can be divided into three main steps: 1) The composition and preparation of cell-laden bioink; 2) Preparation of biofilm scaffolds by extrusion-based 3D bioprinting; 3) Validation
of the cartilage damage repair effect of biofilm on a rat model.

transition temperature, and it was in the sol state. After cooling down to 0.40 kPa. The difference in elastic modulus between the 5% and 10%
the phase transition temperature, the storage modulus (G′) suddenly Gel hydrogels was statistically significant.
increased, and when it was greater than the loss modulus (G″), it ap- The Alg/Gel/HA composite hydrogel has shear thinning and a suit-
peared as a gel state (Fig. 2A). It can be seen from Fig. 2A that the sol– able sol–gel phase transition temperature (Fig. 2A–C) [16]. A hydrogel
gel phase transition temperatures of the three hydrogels were 10.6 °C, scaffold with a high degree of discrimination is prepared by extrusion-
12.1 °C, and 14.8 °C, for Gel concentrations of 5, 8, and 10%, respectively. based 3D printing. It has uniform porosity, a good structure (Fig. 2C)
As the concentration of Gel increased, the sol–gel phase transition tem- and suitable mechanical properties (Fig. 2D, E). Consequently, 5%Gel
perature of the composite hydrogel increased accordingly. Under condi- +2.5% Alg + 0.2%HA hydrogel was chosen for subsequent experiments.
tions of 1% strain, the angular frequency gradually increased from This scaffold can provide cells with appropriate mechanical properties
0.1 rad/s to 100 rad/s, the average storage modulus (elastic modulus) in the early stage of cartilage regeneration to support the structure of
of 5, 8, and 10% Gel hydrogels were 7736 Pa, 8575 Pa, and 13,698 Pa, re- the microenvironment, but will degrade over time, eventually being re-
spectively (Fig. 2B). Moreover, the three hydrogels showed low depen- placed by newly formed cartilage tissue.
dence on frequency from 0.1–100 rad/s, indicating that the hydrogels
had relatively high elasticity. The initial viscosities of the three 3.2. 3D bioprinting and evaluation of cell encapsulated biofilm
hydrogels were 3.45 Pa.s, 4.59 Pa.s, and 8.76 Pa.s. The hydrogels of
three concentrations all showed shear thinning. As the shear rate in- 3D bioprinting is a kind of rapid prototyping technology, and indi-
creased, the viscosity of the three composite hydrogels decreased and vidual precision medical treatment can be performed according to the
gradually approached (Fig. 2C). shape of the patient's cartilage defect [43]. In this work, a pneumatic
Creating a soft microenvironment that simulates chondrocyte pro- extrusion-based 3D bioprinter was used to deposit the Alg/Gel/HA
duction of pericellular matrix and allows fast diffusion of nutrients is bioinks and Table 1 shows the specific printing parameters of hydrogels
fundamental to scaffolds used for repairing joint cartilage damage. with different concentration ratios. A schematic diagram of hydrogel
Meanwhile, it is also essential to provide good mechanical support in biofilm model and specific dimensions are shown in Fig. 1A. The print-
the early stages of joint cartilage regeneration [44]. Fig. 2D and E sum- ing effects of the three hydrogel biofilms are presented in Fig. 1B. The
marizes the mechanical properties of the three hydrogels with pore width ranged from 200 to 400 μm, 150–300 μm, and 560–
different Gel concentrations. When the Alg/HA content was fixed, the 740 μm. During the printing process, 8% Gel +2.5% Alg + 0.2% HA and
mechanical strength was slightly increased with the increment in the 10% Gel +2.5% Alg + 0.2% HA hydrogels were prone to obvious filament
content of Gel. And the compression modulus of the three different con- breakage and uneven filament production, while the biofilm prepared
centrations of hydrogel were 7.07 ± 1.30, 8.68 ± 1.52, and 11.20 ± form 5% Gel +2.5% Alg + 0.2% HA hydrogel had a clear grid structure,

6
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Fig. 2. Rheological properties and mechanical characterizations of Alg/Gel/HA composite hydrogels with different gelatin concentrations. (A) Effect of temperature on storage modulus (G′)
and loss modulus (G″). (B) Storage modulus (G′) and loss modulus (G″) through frequency sweep analysis. (C) Shear rate dependencies of viscosities of the three formulated hydrogels.
(D) Compressive stress−strain curves. (E) Compressive modulus of the three hydrogels.(F)SEM (scanning electron microscopy) image of biofilm scaffold made from 5% Gel +2.5%
Alg + 0.2% HA. (Whole and local enlarged view.)

uniform pores and uniform filament distance. Because of the nature of treat cartilage defects. Therefore, the 5% Gel +2.5% Alg + 0.2% HA hy-
the shear thinning and viscosity changes, 5% Gel +2.5% Alg + 0.2% HA drogel was chosen for subsequent experiments. As observed by SEM,
gel precursors achieved smooth extrusion and rapid covalent the 3D-printed biofilm scaffold reproduced the predesigned structures
crosslinking by Ca2+. The thermo-responsive properties of the compos- very well (Fig. 2C).
ite hydrogel provided mechanical support during bioprinting before To evaluate the compatibility of Alg/Gel/HA hydrogel and any ad-
Ca2+ covalent crosslinking, while at the same time, the ideal structure verse effects that may have been caused by the printing process, cell vi-
(200–400 μm pore size) and certain mechanical strength (elastic mod- ability and morphology were monitored. The cytocompatibility of
ulus: 7.07 ± 1.30 kPa) make it an ideal bioink for printing scaffolds to composite hydrogels was evaluated by the live/dead assay of CPCs.

7
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

CPCs maintained a high survival rate (> 90%) after 1, 7, and 14 days of for the chondrogenic differentiation of CPCs. In addition, the cells began
culture (Fig. 3A,E). Three-dimensional reconstruction of live and dead to connect with each other and some aggregates were found. According
staining also revealed the even distribution of CPCs throughout the bio- to our research and previous studies, FN enhances proliferation, migra-
film (Fig. 3B). The cell growth process was continuously observed for tion, and the chondrogenic differentiation capacity of CPCs through the
two weeks to monitor cell proliferation and morphology. Compared to integrin α5β1-dependent signaling pathway, with a FN concentration
the first day, the number of cells increased significantly after two of 20 μg/mL having the best effect [33]. Therefore, we prepared a com-
weeks (Fig. 3C). It has been proven that when MSCs spread on stiff sub- posite hydrogel combined with FN (20 μg /mL) to improve the biological
strates, and with high cytoskeletal tension, they express high levels of function of the hydrogel. ELISA was performed to test the release kinet-
osteogenic cell makers, while when they maintain a round shape, ics of FN; a burst release was observed within the first eight days, after
chondrogenic differentiation is favored [45,46]. As shown in Fig. 3C, which the release of FN significantly slowed down. The results showed
CPCs remained round in shape in the biofilm instead of visibly that FN can be effectively retained at the site of a cartilage defect for a
stretching during culture. These findings indicate that the composite period of time after implantation, and that this enhances the prolifera-
hydrogels provide an ideal chemical and mechanical microenvironment tion and migration of CPCs. At the same time, the slow release of FN

Fig. 3. (A) Fluorescent microscopic images of CPCs after the live/dead assay on days 1, 7, and 14. (B) Three-dimensional reconstruction of live and dead staining of CPCs used the software
Imaris x64 7.2.1. (C) Cytoskeletal staining of CPCs cultured in the biofilm scaffold for 1 and 14 days. (D) Local magnification of cytoskeletal staining of CPCs at day 14. (E) Quantitative
analysis of live/dead staining. (F) Cumulative fibronectin release from printed biofilm. Alcian blue staining of printed CPCs grown in normal or chondrogenic medium in biofilm after
21 days in culture. Representative images were captured using (G) inverted microscope (H) stereo microscope. (I\ \K) Relative quantification of mRNA levels of chondrogenic makers
(PRG4, COLII, and SOX9) expressed by CPCs in each group by qRT-PCR cultured for 3, 7, or 14 days. (* indicates P <0.05, ** indicates P < 0.01, *** indicates P < 0.001). (2D: two-
dimensional control culture of CPCs in Petri dish, 3D: three-dimensional culture of CPCs in biofilm, 3D + FN: three-dimensional culture of CPCs in biofilm combined with fibronectin).

8
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

from the biofilm can effectively recruit BMSCs released from plates showed higher levels of COL2 and SOX9 than BMSCs cultured
subchondral bone. under control environment (Fig. 4C-E).
Extrusion-based 3D bioprinting can realize the co-printing of FN, The production of cartilage-specific extracellular matrix and in-
CPCs, and Alg/Gel/HA composite hydrogel to prepare a bioactive biofilm. creased expression of chondrogenic genes (COL II, PRG4, and especially
In this study, we used chondrogenic progenitor cells as seed cells in- SOX9) during culture was confirmed by immunofluorescence staining
stead of traditional chondrocytes or mesenchymal stem cells, thus (Fig. 4A) and qRT-PCR (Fig. 3I–K) . Previous studies always used TGF-β
avoiding the problems of dedifferentiation (chondrocytes), ossification, throughout the culture period, which is costly. The Alcian blue staining
and hypertrophy (MSCs) during the culture process. The introduction of of the control group and the induction group were both positive on the
FN into a composite hydrogel improves the biological activity of the bio- biofilm (Fig. 3G, H). These results suggest that exposure of CPCs to TGF-
film. ELISA results confirmed the slow release of FN from the biofilm β is not necessary in this designed biofilm and that CPCs embedded
after printing (Fig. 3F). FN retained within the biofilm can effectively en- within the biofilm possess the ability to spontaneously differentiate
hance the proliferation, migration, and chondrogenic differentiation ca- into chondrocytes [15]. Therefore, this method can shorten the required
pacity of CPCs through the integrin α5β1-dependent signaling pathway. time in vitro, save costs, and avoid adverse effects caused by exposure to
Some of the FN released from the biofilm was able to effectively recruit TGF-β [45]. In addition, in cartilage defects, plenty of BMSCs are released
BMSCs and promote their proliferation and differentiation. from sites of microfracture surrounding the biofilm. As the results
in vitro demonstrated, the bioactive biofilm encapsulated with CPCs
and FN stimulated BMSCs to undergo chondrogenic differentiation cul-
3.3. In vitro biological assessment of biofilm function tured under the biofilm (Fig. 4) [47]. It's worth noting that the interac-
tions of BMSCs and CPCs can maintain the chondrogenic phenotype of
Due to the lack of cell adhesion sites and poor mechanical strength of CPCs, which can promote hyaline cartilage formation [48]. Besides, the
single-material hydrogel, in this study, we prepared an Alg/Gel/HA BMSCs produce Multiple cytokines and thus induce CPCs to undergo
composite hydrogel biofilm. HA has a CD44 recognition site, which chondrogenic differentiation and proliferation through paracrine
can specifically recognize chondrocytes and maintain the chondrocyte signals.
phenotype. Gel, as a degradation product of collagen widely present in
cartilage tissue, has a large number of RGD binding sites, which can sig- 3.4. In vivo assessment of cartilage defect repair
nificantly improve the ability of composite hydrogels to support cell ad-
hesion. To assess the chondrogenic differentiation of CPCs in biofilm The ability of CPCs and FN encapsulated in Alg/Gel/HA biofilm for
in vitro, the GAG secretion of CPCs cultured in biofilm was measured cartilage damage repair was evaluated in a rat cartilage defect model.
using Alcian blue staining. The Alcian blue staining of the control The experiments were divided into five groups as follows: MF
group and the induction group were both positive on biofilm, and the (microfracture only), AMIC + (microfracture +3D-printed Alg/Gel/HA
experimental group was slightly darker (Fig. 3G, H). The results show biofilm), AMIC+FN (microfracture +3D-printed Alg/Gel/HA biofilm
that CPCs embedded in biofilm prepared from Alg/Gel/HA composite combined with FN), AMIC+CPCs (microfracture +3D-printed Alg/Gel/
hydrogel possess the ability to spontaneously differentiate into HA biofilm containing FN) and AMIC+FN + CPCs (microfracture
chondrocytes. The mRNA levels of COL2, PRG4, and SOX9 were evalu- +3D-printed Alg/Gel/HA biofilm containing FN and CPCs). A full-
ated by qRT-PCR for 3,7, and 14 days (Fig. 3I–K). CPCs encapsulated thickness cylindrical cartilage defect (2 mm in diameter) was created
within the biofilm exhibited higher mRNA expression for every protein on the trochlear groove of the distal femur, and then microfracture sur-
expressed by native CPCs within 14 days. The addition of FN increased gery was performed and biofilms were implanted in the injured area
the expression levels of chondrogenesis-related genes to different ex- (Fig. 5A). General observation of the cartilage defects at 6 weeks and
tents, in particular, the expression of PRG4. Interestingly, CPCs in biofilm 12 weeks post-operation were executed to assess the cartilage regener-
combined with FN had the highest expression of PRG4, a key factor in ation ability of the biofilm. According to Wakitani's histological scoring
joint lubrication [35], with the 3D + FN group displaying approximately standard, the scores for each group are shown in Fig. 5C. 6 and
a 5.5-fold (day14) increase compared to the 3D group without FN (2.6- 12 weeks after surgery, the scores were 14.5 and 12.5, 13.5 and 11, 11
fold). Furthermore, at day 14, CPCs in biofilm combined with FN re- and 7.75, 10.25 and 6.5, and 6.5 and 2.5 for the MF, AMIC+, AMIC
vealed higher mRNA levels with 2.4-fold increases in COLII, 5.5-fold in +FN, AMIC+CPCs, and AMIC+FN + CPCs groups, respectively. There
PRG4, and 1.7-fold in SOX9, compared with CPCs cultured in plates was no significant inflammatory response or synovitis in any of the
(Fig. 3K). The results indicated that Alg/Gel/HA composite hydrogel groups at any time point (Fig. 5B). At 6 weeks post-implantation, the
stimulates the chondrogenic differentiation of CPCs. newly formed tissues had an irregular surface, and the interface be-
Immunofluorescence analysis of COL2, PRG4 (cartilage-specific tween newly formed cartilage and normal cartilage was still noticeable.
markers) and SOX9 (a master regulatory gene of chondrogenesis), The neocartilage in the AMIC+CPCs group was solid and well integrated
showed a similar tendency (Fig. 4A). CPCs secrete cartilage-specific ex- with the surrounding normal cartilage, especially in the AMIC
tracellular matrix under uninduced conditions. CPCs in the biofilm+FN +FN + CPCs group, while the neocartilage in the AMIC+ and AMIC
group showed a markedly enhanced green fluorescence signal for COL2, +FN groups were baggy. In the MF group, only a small amount of regen-
compared the fluorescence signal of CPCs in the biofilm group. Specially, erated tissue was observed in the defect and this resembled fibrous tis-
strongly positive staining for PRG4 was present in the biofilm+FN sue (Fig. 5B). The colour of new tissue was similar to the surrounding
group after 14 days. The green signal from the stained chondrogenic tissue, indicating that the biofilm had been well degraded. At 8 weeks
transcription factor SOX9 was detected in all groups. post-implantation, the regenerated cartilage were still not well inte-
The indirect contact co-culture model was applied to study the im- grated with the nearby native cartilage. Nonetheless, the newly formed
pact of Alg/Gel/HA + CPCs+FN biofilm on chondrogenic differentiation tissue in the AMIC+CPCs groups had become smoother and well inte-
of BMSCs, simulating the BMSCs released by microfracture (Fig. 4B). grated with the surrounding normal cartilage. Especially, the interface
After 14 days, positive Alcian blue staining of BMSCs on the same well between neocartilage and normal cartilage could hardly be distin-
below the Transwell was observed (Fig. 4B), and was increased com- guished in the AMIC+FN + CPCs groups. It's worth noting that both
pared to that in BMSCs cultured on normal culture plates (control) AMIC+CPCs and AMIC+FN + CPCs groups displayed pregnant cartilage
(Fig. 4B). This proved that the Alg/Gel/HA + CPCs+FN biofilm can pos- regeneration at 6 weeks and 12 weeks, compared with three other
itively stimulate chondrogenic differentiation of BMSCs released by groups. Especially, the AMIC+FN + CPCs groups exhibited the best
microfracture. Further qRT-PCR results confirmed that BMSCs co- macroscopic cartilage regeneration covering the entire cartilage lesion
cultured with Alg/Gel/HA + CPCs+FN biofilm under the Transwell modeling area.

9
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Fig. 4. (A) COLII, PRG4, and SOX9 immunostaining of CPCs in each group. (green: COLII, PRG4, and SOX9, blue: nuclei) (B) Schematic diagram of evaluation method for investigating the
effects of Alg/Gel/HA + CPCs+FN biofilm on differentiation of BMSCs outside the construct. BMSCs were stained with Alcian blue. (C-E) Relative quantification of mRNA levels for COLII,
Acan and SOX9 expressed by BMSCs co-cultured with Alg/Gel/HA + CPCs+FN biofilm by qRT-PCR at 7 and 14 days (* indicates P < 0.05, ** indicates P < 0.01, *** indicates P < 0.001).

Images of HE-stained tissue are shown in Fig. 6. At week 6, the most little neocartilage regeneration was verified. However, the structures
obvious cartilage damage in the MF group was observed in all five of neocartilage were different from native cartilage and the articular
groups, where little tissue was regenerated and the neocartilage lacked surface was irregular. For the defects filled with Alg/Gel/HA biofilm
characteristic cartilage-like lacunae structures. Then improvement in combined with FN or CPCs, some cartilage regeneration was observed,
the cartilage layer was observed after implantation of biofilm, and a but the neocartilage was thinner than normal, and might not be well

Fig. 5. (A) Images of cartilage injury model during surgery. (B) Gross morphology of regenerated cartilage tissues at 6 and 12 weeks after surgery. (C) Histological scoring for the repaired
tissues in the different groups. * indicates that the histological scores in these groups were significantly lower than that in the MF group (P < 0.005).

10
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

integrated with the surrounding normal cartilage tissue, as well as lack- addition of CPCs or FN-functionalized biofilm further increased the ex-
ing well-organized maturation layers. On the other hand, although the pression of type II collagen in the regenerated cartilage tissue. Similar
cartilage matrix was unevenly distributed to a small extent, the to the results of HE and Safranin O staining, COL II was more positively
chondrocytes proliferated and were arranged neatly. Interestingly, the and uniformly stained in the AMIC+FN + CPCs group than any other
defects filled with biofilm combined with FN and CPCs showed regener- group, and the protein expressions were increased with time. It's
ated hyaline cartilage, similar to native cartilage. Furthermore, no worth noticing that the newly formed cartilage tissue showed clustered
boundary could be observed between neocartilage and the adjacent na- and spherical lacunae, that is, characteristic cartilage-like lacunae
tive cartilage. At 12 weeks post-implantation, area of cartilage defects structures.
were almost completely covered by the regenerated cartilage tissue. A To further quantitatively evaluate the repair efficacy for cartilage de-
distinct boundary between the neocartilage and adjacent normal carti- fects in vivo, qRT-PCR was used to detected chondrogenic difference
lage could still be observed in the MF group while it was almost invisible gene expression in the newly formed tissue (Fig. 8B-C). Compared
in the other AMIC+ groups. The AMIC+FN + CPCs groups still with that in the MF group, the gene levels of SOX9 and COL2 were up-
displayed the best performance in cartilage regeneration in all five regulated in the AMIC+CPCs groups at 6 weeks and 12 weeks and
groups. They also had characteristic cartilage-like lacunae and gradient were significantly increased after the synergies between CPCs and FN
structures similar to native cartilage tissue. in the AMIC+FN + CPCs group. The AMIC+ and AMIC+FN groups
Moreover, the repair effect of cartilage defects was further evaluated also tended to promote expression of chondrogenic difference markers,
by Safranin O, which stained the proteoglycan of newly formed cartilage although the increase was not significant.
red (Fig. 7). At 12 weeks postoperation, on the surface of the regener- Because the local microenvironment after surgery cannot recruit
ated cartilage tissue, a light red-stained layer was observed in all groups. and guide the differentiation of BMSCs, this results in the regenera-
There was almost on difference in neocartilage between AMIC tion of essentially fibrous cartilage with significantly lower strength
+FN + CPCs groups and the surrounding normal cartilage tissue, the than native cartilage. AMIC technology fixes the released cells onto
red-stained layer was the most compact, and the staining was the the cartilage defect using a collagen membrane and improves the
most uniform. Compared to the MF group, although the biofilm im- cartilage repair effect to a certain extent, but it still cannot regenerate
planted group stimulated cartilage formation, the structure of the re- natural hyaline cartilage. To overcome the lack of chemical cues in
generated cartilage in the AMIC+FN + CPCs groups was more similar local microenvironment after microfracture surgery for the released
to normal cartilage tissue. Likewise, a distinct boundary was seen be- cellular component, we incorporated a CPC-laden biofilm based on
tween the neocartilage and surrounding native cartilage. By contrast, an Alg/Gel/HA composite hydrogel combined with FN into the defect
the defects were filled with hyaline cartilage-like tissue in the AMIC area [49]. When the biofilm was implanted in vivo, the cartilage de-
+FN + CPCs groups at 6 weeks, which was positively stained by fects showed the best cartilage regeneration with a gradient struc-
Safranin O. ture among all groups in the AMIC+FN + CPCs group. The cell
The results of type II collagen immunohistochemical staining are morphology of the regenerated cartilage from the AMIC+FN + CPCs
shown in Fig. 8A. All AMIC-modified groups showed significant in- group was similar to native hyaline cartilage with round
creases in type II collagen expression compared to the MF control. The chondrocytes within lacunae (Figs. 6, 7).

Fig. 6. HE staining images of the regenerated tissues in the different groups at 6 and 12 weeks after surgery.

11
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

Fig. 7. Safranin-O staining of the regenerated tissues in the different groups at 6 and 12 weeks after surgery.

The traditional AMIC technology lacks a good postoperative micro- culture of CPCs and BMSCs. According to the literature, when MSCs
environment, and cannot effectively induce the chondrogenic differen- are co-cultured with CPCs, interactions between the two cell types
tiation of BMSCs released from subchondral bone. The regenerated maintained the chondrogenic phenotype of CPCs and stimulated the
cartilage tissue is basically fibrocartilage instead of natural hyaline carti- MSCs to differentiate into chondrocytes, which contributed to chondro-
lage, and lacks long-term efficacy. In this study, a 3D printed active bio- genesis [48,50]. On the other hand, thanks to the effective controlled re-
film was used to replace the collagen membrane, which effectively lease of FN in biofilms, BMSCs were effectively recruited. At the same
adhered to cartilage defects. The active biofilm as a whole provides a time, CPCs present in biofilms can spontaneously differentiate into
good microenvironment and can effectively induce BMSCs to differenti- chondrocytes. The high expression of PRG4 and COLII in vitro and
ate into cartilage. For one thing, this effect may benefit from the co- in vivo experiments confirms the superficial localization of PRG4 and

Fig. 8. (A) Immunohistochemical staining for Type II collagen of the regenerated tissues in the different groups at 6 and 12 weeks after surgery. Expression levels of the chondrogenic-
specific genes COLII, PRG4 and SOX9, in newly formed cartilage with biofilm implantation for 6 (A) and 12 weeks (B). *,# indicate P < 0.05, **, ## indicate P < 0.01, and ***,### indicate
P < 0.001.

12
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

COLII. Under the recruitment and action of the slowly released FN, References
BMSCs adhering to the lower layer of the biofilm differentiated into hy-
[1] D.J. Huey, J.C. Hu, K.A. Athanasiou, Unlike bone, cartilage regeneration remains elu-
pertrophic chondrocytes, and the abundant presence of type X collagen sive, Science (New York, N.Y.) 338 (6109) (2012) 917–921, https://doi.org/10.1126/
in the deep zone was confirmed, indicative of regenerated superficial science.1222454.
zone articular cartilage and deep zone hypertrophic cartilage in newly [2] S.C. Mastbergen, D.B. Saris, F.P. Lafeber, Functional articular cartilage repair: here,
near, or is the best approach not yet clear? Nat. Rev. Rheumatol. 9 (5) (2013)
formed cartilage. 277–290, https://doi.org/10.1038/nrrheum.2013.29.
The layered structure of natural cartilage, such as arrangement of [3] J. Martel-Pelletier, A.J. Barr, F.M. Cicuttini, P.G. Conaghan, C. Cooper, M.B. Goldring,
collagenous fibers or density distribution of chondrocytes, also should S.R. Goldring, G. Jones, A.J. Teichtahl, J.P. Pelletier, Osteoarthritis, Nat. Rev. Dis.
Primers 2 (2016) 16072, https://doi.org/10.1038/nrdp.2016.72.
be taken into account in future studies to achieve hyaline cartilage re-
[4] C. Becher, M.A. Malahias, M.M. Ali, N. Maffulli, H. Thermann, Arthroscopic
generation [46]. Although the thickness of cartilage in the rat limited microfracture vs. arthroscopic autologous matrix-induced chondrogenesis for the
the possibility of applying a layered scaffold in this study, it is very sig- treatment of articular cartilage defects of the talus, Knee Surg. Sports Traumatol.
nificative to evaluate the possibility of using layered cartilage scaffolds Arthrosc. 27 (9) (2019) 2731–2736, https://doi.org/10.1007/s00167-018-5278-7.
[5] D.L. Richter, R.C. Schenck Jr., D.C. Wascher, G. Treme, Knee articular cartilage repair
in large animals. In addition, an intact subchondral bed is essential to and restoration techniques: a review of the literature, Sports Health 8 (2) (2016)
the treatment of the chondral lesion [48]. We also need to improve 153–160, https://doi.org/10.1177/1941738115611350.
the preparation process of the scaffold to enhance the resolution of [6] E.A. Makris, A.H. Gomoll, K.N. Malizos, J.C. Hu, K.A. Athanasiou, Repair and tissue en-
gineering techniques for articular cartilage, Nat. Rev. Rheumatol. 11 (1) (2015)
the printed structure. For example, low-viscosity hydrogels can be fab- 21–34, https://doi.org/10.1038/nrrheum.2014.157.
ricated with high resolution via model-support bioink interaction [51]. [7] J.M. Hurst, J.R. Steadman, L. O’Brien, W.G. Rodkey, K.K. Briggs, Rehabilitation follow-
Consequently, regeneration of the osteochondral area as a whole need ing microfracture for chondral injury in the knee, Clin. Sports Med. 29 (2) (2010)
257–265 , viii https://doi.org/10.1016/j.csm.2009.12.009.
be noticed instead of the articular cartilage alone [48]. Although some [8] T. Guo, M. Noshin, H.B. Baker, E. Taskoy, S.J. Meredith, Q. Tang, J.P. Ringel, M.J.
cartilage repair results are promising, remodeling a consistent and sta- Lerman, Y. Chen, J.D. Packer, J.P. Fisher, 3D printed biofunctionalized scaffolds for
ble osteochondral interface to facilitate good graft-to-bone attachment microfracture repair of cartilage defects, Biomaterials 185 (2018) 219–231,
https://doi.org/10.1016/j.biomaterials.2018.09.022.
is still an unsolved problem [52].
[9] L. Weigelt, R. Hartmann, C. Pfirrmann, N. Espinosa, S.H. Wirth, Autologous matrix-
induced Chondrogenesis for Osteochondral lesions of the talus: a clinical and radio-
4. Conclusion logical 2- to 8-year follow-up study, Am. J. Sports Med. 47 (7) (2019) 1679–1686,
https://doi.org/10.1177/0363546519841574.
[10] A. Schiavone Panni, C. Del Regno, G. Mazzitelli, R. D’Apolito, K. Corona, M. Vasso,
In summary, we generated a 3D-printed bioactive biofilm to modify Good clinical results with autologous matrix-induced chondrogenesis (Amic) tech-
AMIC technology. Covering a defect with biofilm provides adequate me- nique in large knee chondral defects, Knee Surg. Sports Traumatol. Arthrosc. 26 (4)
chanical support and architectural integrity, offering a stable microenvi- (2018) 1130–1136, https://doi.org/10.1007/s00167-017-4503-0.
[11] L. de Girolamo, H. Schonhuber, M. Vigano, C. Bait, A. Quaglia, G. Thiebat, P. Volpi, Au-
ronment for BMSCs released from subchondral bone. We verified that a tologous matrix-induced Chondrogenesis (AMIC) and AMIC enhanced by autolo-
laminar structure similar to natural hyaline cartilage was regenerated in gous concentrated bone marrow aspirate (BMAC) allow for stable clinical and
the rat cartilage defect model. By introducing FN into CPC-laden biofilm, functional improvements at up to 9 years follow-up: results from a randomized
controlled study, J. Clin. Med. 8 (3) (2019)https://doi.org/10.3390/jcm8030392.
we showed that CPCs embedded in biofilm possess excellent potential [12] L.G. Bracaglia, B.T. Smith, E. Watson, N. Arumugasaamy, A.G. Mikos, J.P. Fisher, 3D
to undergo chondrogenic differentiation under uninduced conditions. printing for the design and fabrication of polymer-based gradient scaffolds, Acta
Alg/Gel/HA + CPCs+FN biofilm can positively stimulate chondrogenic Biomater. 56 (2017) 3–13, https://doi.org/10.1016/j.actbio.2017.03.030.
[13] J. Yang, Y.S. Zhang, K. Yue, A. Khademhosseini, Cell-laden hydrogels for
differentiation of BMSCs released by microfracture. It can provide cells
osteochondral and cartilage tissue engineering, Acta Biomater. 57 (2017) 1–25,
with certain mechanical properties in the early stage of cartilage regen- https://doi.org/10.1016/j.actbio.2017.01.036.
eration to support the structure of the microenvironment, and will de- [14] L. Shao, Q. Gao, C. Xie, J. Fu, M. Xiang, Y. He, Directly coaxial 3D bioprinting of large-
grade over time, eventually being replaced by newly formed cartilage scale vascularized tissue constructs, Biofabrication 12 (3) (2020), 035014, https://
doi.org/10.1088/1758-5090/ab7e76.
tissue. With this bioactive biofilm, the AMIC outcome was significantly [15] H. Rogan, F. Ilagan, X. Tong, C.R. Chu, F. Yang, Microribbon-hydrogel composite scaf-
enhanced by interaction between the CPC-laden biofilm and BMSCs re- fold accelerates cartilage regeneration in vivo with enhanced mechanical properties
leased from bone marrow during the healing process. Replacing the tra- using mixed stem cells and chondrocytes, Biomaterials 228 (2020) 119579, https://
doi.org/10.1016/j.biomaterials.2019.119579.
ditional collagen membrane with active biofilm resulted in the [16] Q. Gao, X. Niu, L. Shao, L. Zhou, Z. Lin, A. Sun, J. Fu, Z. Chen, J. Hu, Y. Liu, Y. He, 3D
regeneration of a thicker and healthier cartilage. For clinical translation, printing of complex GelMA-based scaffolds with nanoclay, Biofabrication 11 (3)
we could incorporate AMIC surgery and 3D bioprinting by performing (2019), 035006, https://doi.org/10.1088/1758-5090/ab0cf6.
[17] P. Rastogi, B. Kandasubramanian, Review of alginate-based hydrogel bioprinting for
an arthroscopy procedure to replace the damaged cartilage with 3D- application in tissue engineering, Biofabrication 11 (4) (2019), 042001, https://doi.
printed biofilm after microfracture surgery. org/10.1088/1758-5090/ab331e.
[18] K. Markstedt, A. Mantas, I. Tournier, H. Martínez Ávila, D. H?gg, P.J.B. Gatenholm, 3D
Bioprinting Human Chondrocytes with Nanocellulose-Alginate Bioink for Cartilage
Author statement
Tissue Engineering Applications, 16(5), 2015 1489–1496.
[19] J. Schiavi, L. Reppel, N. Charif, N. de Isla, D. Mainard, N. Benkirane-Jessel, J.F. Stoltz, R.
Yang Zhou: Methodology, Investigation, Writing - original draft. Ran Rahouadj, C. Huselstein, Mechanical stimulations on human bone marrow mesen-
Qin: Investigation, Writing - original draft. Tong Chen: Methodology, chymal stem cells enhance cells differentiation in a three-dimensional layered scaf-
fold, J. Tissue Eng. Regen. Med. 12 (2) (2018) 360–369, https://doi.org/10.1002/
Validation. Kaibin Zhang: Formal analysis. Jianchao Gui: Conceptualiza- term.2461.
tion, Resources, Supervision, Writing - review & editing [20] F. You, X. Chen, D.M.L. Cooper, T. Chang, B.F. Eames, Homogeneous hydroxyapatite/
alginate composite hydrogel promotes calcified cartilage matrix deposition with po-
tential for three-dimensional bioprinting, Biofabrication 11 (1) (2018) 015015,
https://doi.org/10.1088/1758-5090/aaf44a.
Declaration of Competing Interest [21] C. Antich, J. de Vicente, G. Jimenez, C. Chocarro, E. Carrillo, E. Montanez, P. Galvez-
Martin, J.A. Marchal, Bio-inspired hydrogel composed of hyaluronic acid and algi-
nate as a potential bioink for 3D bioprinting of articular cartilage engineering con-
The authors declare that they have no known competing financial structs, Acta Biomater. 106 (2020) 114–123, https://doi.org/10.1016/j.actbio.2020.
interests or personal relationships that could have appeared to influ- 01.046.
ence the work reported in this paper. [22] K.C.R. Kolan, J.A. Semon, B. Bromet, D.E. Day, M.C. Leu, Bioprinting with human stem
cell-laden alginate-gelatin bioink and bioactive glass for tissue engineering, Int. J.
Bioprint. 5 (2.2) (2019) 204, https://doi.org/10.18063/ijb.v5i2.2.204.
Acknowledgements [23] P. Dubruel, R. Unger, S.V. Vlierberghe, V. Cnudde, P.J. Jacobs, E. Schacht, C.J.
Kirkpatrick, Porous gelatin hydrogels: 2. In vitro cell interaction study,
Biomacromolecules 8 (2) (2007) 338–344, https://doi.org/10.1021/bm0606869.
This work is financially supported by the National Natural Science
[24] S.H. Jeong, M. Kim, T.Y. Kim, H. Kim, J.H. Ju, S.K. Hahn, Supramolecular injectable
Foundation of China (No. 81601954, No. 81672210 and No.81673995). hyaluronate hydrogels for cartilage tissue regeneration, ACS Appl. Bio Mater. 3 (8)
(2020) 5040–5047, https://doi.org/10.1021/acsabm.0c00537.

13
Y. Zhou, R. Qin, T. Chen et al. Materials and Design 203 (2021) 109621

[25] S. Stichler, T. Bock, N. Paxton, S. Bertlein, R. Levato, V. Schill, W. Smolan, J. Malda, J. [39] A. Dhawan, P.M. Kennedy, E.B. Rizk, I.T. Ozbolat, Three-dimensional bioprinting for
Tessmar, T. Blunk, J. Groll, Double printing of hyaluronic acid/poly(glycidol) hybrid bone and cartilage restoration in Orthopaedic surgery, J Am Acad Orthop Surg 27
hydrogels with poly(epsilon-caprolactone) for MSC chondrogenesis, Biofabrication (5) (2019) e215–e226, https://doi.org/10.5435/JAAOS-D-17-00632.
9 (4) (2017), 044108, https://doi.org/10.1088/1758-5090/aa8cb7. [40] G.P. Dowthwaite, J.C. Bishop, S.N. Redman, I.M. Khan, P. Rooney, D.J. Evans, L.
[26] A. Shpichka, D. Osipova, Y. Efremov, P. Bikmulina, N. Kosheleva, M. Lipina, E.A. Haughton, Z. Bayram, S. Boyer, B. Thomson, M.S. Wolfe, C.W. Archer, The surface
Bezrukov, R.B. Sukhanov, A.B. Solovieva, M. Vosough, P. Timashev, Fibrin-based of articular cartilage contains a progenitor cell population, J. Cell Sci. 117 (Pt 6)
Bioinks: new tricks from an old dog, Int. J. Bioprint. 6 (3) (2020) 269, https://doi. (2004) 889–897, https://doi.org/10.1242/jcs.00912.
org/10.18063/ijb.v6i3.269. [41] C. Erggelet, M. Endres, K. Neumann, L. Morawietz, J. Ringe, K. Haberstroh, M.
[27] A.C. Daly, F.E. Freeman, T. Gonzalez-Fernandez, S.E. Critchley, J. Nulty, D.J. Kelly, 3D Sittinger, C. Kaps, Formation of cartilage repair tissue in articular cartilage defects
bioprinting for cartilage and Osteochondral tissue engineering, Adv Healthc Mater pretreated with microfracture and covered with cell-free polymer-based implants,
6 (22) (2017)https://doi.org/10.1002/adhm.201700298. J. Orthop. Res. 27 (10) (2009) 1353–1360, https://doi.org/10.1002/jor.20879.
[28] D. Cinats, S. Miller, Z. Abusara, S.M. Heard, C. Hutchison, N. Schachar, S. [42] E. Axpe, M.L. Oyen, Applications of alginate-based bioinks in 3D bioprinting, Int. J.
Timmermann, Evolution of a novel tissue preservation protocol to optimize Mol. Sci. 17 (12) (2016)https://doi.org/10.3390/ijms17121976.
osteochondral transplantation outcomes, Cartilage (2018)https://doi.org/10.1177/ [43] J.H.Y. Chung, S. Naficy, Z. Yue, R. Kapsa, A. Quigley, S.E. Moulton, G.G. Wallace, Bio-
1947603518812557 1947603518812557. ink properties and printability for extrusion printing living cells, Biomater Sci 1
[29] Y. Jiang, R.S. Tuan, Origin and function of cartilage stem/progenitor cells in osteoar- (7) (2013) 763–773, https://doi.org/10.1039/c3bm00012e.
thritis, Nat. Rev. Rheumatol. 11 (4) (2015) 206–212, https://doi.org/10.1038/ [44] Y.A.J. Bruna, A.G. de Melo, Shreya Mehrotra, Michelle A. Calabrese, Tom Kamperman,
nrrheum.2014.200. Biman B. Mandal, Maria H.A. Santana, Eben Alsberg, Jeroen Leijten*, Su Ryon Shin*,
[30] K. Brady, S.C. Dickinson, A.P. Hollander, Changes in chondrogenic progenitor popu- 3D printed cartilage-like tissue constructs with spatially controlled mechanical
lations associated with aging and osteoarthritis, Cartilage 6 (2 Suppl) (2015) properties %J advanced functional materials, Adv. Funct. Mater. 29 (51) (2019).
https://doi.org/10.1177/1947603515574838 30s–5s. [45] M. Costantini, J. Idaszek, K. Szoke, J. Jaroszewicz, M. Dentini, A. Barbetta, J.E.
Brinchmann, W. Swieszkowski, 3D bioprinting of BM-MSCs-loaded ECM biomi-
[31] K. Xue, X. Zhang, Z. Gao, W. Xia, L. Qi, K. Liu, Cartilage progenitor cells combined
metic hydrogels for in vitro neocartilage formation, Biofabrication 8 (3) (2016),
with PHBV in cartilage tissue engineering, J. Transl. Med. 17 (1) (2019) 104,
035002, https://doi.org/10.1088/1758-5090/8/3/035002.
https://doi.org/10.1186/s12967-019-1855-x.
[46] A.C. Daly, D.J. Kelly, Biofabrication of spatially organised tissues by directing the
[32] P. Singh, C. Carraher, J.E. Schwarzbauer, Assembly of fibronectin extracellular matrix,
growth of cellular spheroids within 3D printed polymeric microchambers, Biomate-
Annu. Rev. Cell Dev. Biol. 26 (2010) 397–419, https://doi.org/10.1146/annurev-
rials 197 (2019) 194–206, https://doi.org/10.1016/j.biomaterials.2018.12.028.
cellbio-100109-104020.
[47] C.D. Hoemann, J. Sun, M.D. McKee, A. Chevrier, E. Rossomacha, G.E. Rivard, M.
[33] T. Tao, Y. Li, C. Gui, Y. Ma, Y. Ge, H. Dai, K. Zhang, J. Du, Y. Guo, Y. Jiang, J. Gui, Fibro- Hurtig, M.D. Buschmann, Chitosan-glycerol phosphate/blood implants elicit hyaline
nectin enhances cartilage repair by activating progenitor cells through integrin cartilage repair integrated with porous subchondral bone in microdrilled rabbit de-
alpha5beta1 receptor, Tissue Eng Part A 24 (13–14) (2018) 1112–1124, https:// fects, Osteoarthr. Cartil. 15 (1) (2007) 78–89, https://doi.org/10.1016/j.joca.2006.
doi.org/10.1089/ten.TEA.2017.0322. 06.015.
[34] Y. Li, J. Zhou, X. Yang, Y. Jiang, J. Gui, Intermittent hydrostatic pressure maintains and [48] Y. Zhu, L. Kong, F. Farhadi, W. Xia, J. Chang, Y. He, H. Li, An injectable continuous
enhances the chondrogenic differentiation of cartilage progenitor cells cultivated in stratified structurally and functionally biomimetic construct for enhancing
alginate beads, Develop. Growth Differ. 58 (2) (2016) 180–193, https://doi.org/10. osteochondral regeneration, Biomaterials 192 (2019) 149–158, https://doi.org/10.
1111/dgd.12261. 1016/j.biomaterials.2018.11.017.
[35] R. Levato, W.R. Webb, I.A. Otto, A. Mensinga, Y. Zhang, M. van Rijen, R. van Weeren, [49] W. Shi, M. Sun, X. Hu, B. Ren, J. Cheng, C. Li, X. Duan, X. Fu, J. Zhang, H. Chen, Y. Ao,
I.M. Khan, J. Malda, The bio in the ink: cartilage regeneration with bioprintable Structurally and functionally optimized silk-fibroin-gelatin scaffold using 3D print-
hydrogels and articular cartilage-derived progenitor cells, Acta Biomater. 61 ing to repair cartilage injury in vitro and in vivo, Adv. Mater. 29 (29) (2017)
(2017) 41–53, https://doi.org/10.1016/j.actbio.2017.08.005. https://doi.org/10.1002/adma.201701089.
[36] L. Shao, Q. Gao, C. Xie, J. Fu, M. Xiang, Z. Liu, L. Xiang, Y. He, Sacrificial microgel-laden [50] V.V. Meretoja, R.L. Dahlin, F.K. Kasper, A.G. Mikos, Enhanced chondrogenesis in co-
bioink-enabled 3D bioprinting of mesoscale pore networks, Bio-Design Manufact. 3 cultures with articular chondrocytes and mesenchymal stem cells, Biomaterials 33
(1) (2020) 30–39, https://doi.org/10.1007/s42242-020-00062-y. (27) (2012) 6362–6369, https://doi.org/10.1016/j.biomaterials.2012.05.042.
[37] S. Jordahl, L. Solorio, D.B. Neale, S. McDermott, J.H. Jordahl, A. Fox, C. Dunlay, A. Xiao, [51] E.Y. Tan, R. Suntornnond, W.Y.J.D. P, A. Yeong, Manufacturing, High-Resolution
M. Brown, M. Wicha, G.D. Luker, J. Lahann, Engineered Fibrillar Fibronectin net- Novel Indirect Bioprinting of Low-Viscosity Cell-Laden Hydrogels via Model-
works as three-dimensional tissue scaffolds, Adv. Mater. 31 (46) (2019), Support Bioink Interaction, 2020.
e1904580, https://doi.org/10.1002/adma.201904580. [52] L. Chen, C. Deng, J. Li, Q. Yao, J. Chang, L. Wang, C. Wu, 3D printing of a lithium-
[38] J. Zhang, X. Ma, D. Lin, H. Shi, Y. Yuan, W. Tang, H. Zhou, H. Guo, J. Qian, C. Liu, Mag- calcium-silicate crystal bioscaffold with dual bioactivities for osteochondral inter-
nesium modification of a calcium phosphate cement alters bone marrow stromal face reconstruction, Biomaterials 196 (2019) 138–150, https://doi.org/10.1016/j.
cell behavior via an integrin-mediated mechanism, Biomaterials 53 (2015) biomaterials.2018.04.005.
251–264, https://doi.org/10.1016/j.biomaterials.2015.02.097.

14

You might also like