You are on page 1of 12

Bioprinting 24 (2021) e00163

Contents lists available at ScienceDirect

Bioprinting
journal homepage: www.sciencedirect.com/journal/bioprinting

Alginate-based tissue-specific bioinks for multi-material 3D-bioprinting of


pancreatic islets and blood vessels: A step towards vascularized
pancreas grafts
Joanna Idaszek a, *, 1, Marina Volpi a, 1, Alessia Paradiso a, Martyna Nguyen Quoc a,
Żaneta Górecka a, Marta Klak b, Grzegorz Tymicki b, Andrzej Berman b, Mateusz Wierzbicki c,
Sławomir Jaworski c, Marco Costantini a, d, Agnieszka Kępczyńska e, Ewa Sawosz Chwalibóg c,
Michał Wszoła b, Wojciech Święszkowski a, **
a
Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
b
Foundation of Research and Science Development, Warsaw, Poland
c
Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
d
Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
e
Laboratory of Electron Microscopy, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: Although allogeneic islet transplantation has been proposed as a therapy for type 1 diabetes, its success rate
Microfluidic-assisted multi-material 3D bio­ remains low. Disruption of both extracellular matrix (ECM) and dense vascular network during islets isolation are
printing referred to as some of the main causes of their poor engraftment. Therefore, the recapitulation of the native
Pancreatic islets
pancreatic microenvironment and its prompt revascularization should be beneficial for long-term islet survival.
Blood vessels
Bioink
In this study, we developed novel bioinks suitable for the microfluidic-assisted multi-material biofabrication of
ECM 3D porous pancreatic and vascular structures. The tissue-specific bioactivity was introduced by blending alginate
Fibrinogen either with pancreatic decellularized extracellular matrix powder (A_ECM) or fibrinogen (A_FBR). The formu­
Alginate lated bioinks, although they exhibited different rheological properties, were 3D-printed with high shape fidelity
using a multichannel microfluidic platform coupled with a co-axial needle system. The A_FBR scaffolds were
subjected to secondary crosslinking using thrombin, which significantly decreased their swelling and increased
mechanical properties. To investigate the bioactivity, the A_ECM and A_FBR bioinks were laden with, respec­
tively, porcine pancreatic islets and a mixture of vessel-forming cells (HUVEC and HMSC) and 3D-bioprinted
with high viability. Insulin secretion upon glucose stimulation and developed vessel-like structures composed
of CD31-positive cells validated the biomimetic potential of the two formulated bioinks. Moreover, we
demonstrated the feasibility of the secondary crosslinking to modulate cell behaviour and angiogenic potential
both in vitro and in vivo using a chick chorioallantoic membrane model. Finally, the successful 3D-printing of
heterogeneous 3D scaffolds with three different configurations demonstrated that our approach could be
considered as a potential step towards the biofabrication of a vascularized pancreas construct.

1. Introduction blood glucose level and leads to disorders of glucose homeostasis, which
may, in turn, result in long-term, and potentially life-threatening,
Type 1 diabetes mellitus (T1DM) is a metabolic disorder character­ co-morbidities such as retinopathy, cardiovascular diseases, and renal
ized by an autoimmune response causing the destruction of insulin- failure [2]. The most widespread therapy for T1DM treatment includes
secreting β-cells [1]. Thus, T1DM results in the inability to control the delivery of exogenous synthetic insulin by subcutaneous injections.

* Corresponding author. Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland.
** Corresponding author.
E-mail addresses: joanna.idaszek@pw.edu.pl (J. Idaszek), wojciech.swieszkowski@pw.edu.pl (W. Święszkowski).
1
These authors have equally contributed

https://doi.org/10.1016/j.bprint.2021.e00163
Received 23 February 2021; Received in revised form 23 July 2021; Accepted 26 July 2021
Available online 3 August 2021
2405-8866/© 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
J. Idaszek et al. Bioprinting 24 (2021) e00163

Even though it has been considered a life-saving procedure, such fabrication of complex heterogenous structures by simultaneously
treatment is associated with several limitations, mostly related to diffi­ extruding or rapidly switching between multiple bioinks [30–33].
culties in mimicking the physiological insulin release [3]. In this frame, Nevertheless, proper bioink formulations for microfluidic-assisted
allogeneic pancreatic islet transplantation has emerged as a promising 3D-BP still remain challenging, as they have to meet technological and
therapy for T1DM [4,5]. Although the implantation procedure of islets biological criteria imposed by the bioprinting window.
of Langerhans is less invasive than conventional pancreas trans­ In this study, we developed two biomimetic bioinks suitable for
plantation and allows the successful transplantation of lower quality microfluidics-assisted extrusion-based 3D-BP of heterogeneous scaffolds
tissues (e.g., autologous islets transplantation) [6,7], the therapeutic consisting of pancreatic and vascular compartments. The two tissue-
outcomes of such procedure are partially limited due to low efficiency of specific bioinks were formulated by incorporating pancreatic decellu­
islet engraftment [8]. larized ECM (dECM) powder and fibrinogen into alginate to reproduce
Several factors are indicated as the underlying cause of high post- pancreatic and vascular structure, respectively. The proposed bioinks
transplantation islet loss [9]. Among them, deprivation of interactions were successfully bioprinted into 3D high-resolution porous scaffolds
between the islets and extracellular matrix (ECM) molecules and and investigated separately through morphological, physicochemical,
disruption of the dense vascular network intertwining the islets are a and mechanical characterization to validate their capacity for engi­
direct consequence of the isolation from the donor pancreas [10,11]. neering the tissue-specific function. Moreover, biological evaluation was
Moreover, the destruction of the dense microvasculature not only im­ performed to further explore the biomimetic potential. In particular,
pairs efficient oxygenation, nutrient delivery, and metabolic waste dECM- and fibrinogen-based bioinks were combined with, respectively,
removal, but also disables the pancreatic islets from rapidly sensing porcine pancreatic islets and vessel-forming cells (i.e., a mixture of
blood glucose fluctuations and responding accordingly by secreting in­ human mesenchymal stem cells (HMSC) and human umbilical vein
sulin [12]. Although transplanted islets can survive in avascular condi­ endothelial cells (HUVEC)). As a result, an increased insulin secretion
tions for up to several days, further delays in tissue vascularization leads upon glucose stimulation was detected, and the formation of in vitro
to necrosis and loss of endocrine function [10]. Therefore, recapitulation vessel-like structures was obtained. Moreover, potential pro-angiogenic
of native pancreatic ECM-microenvironment and a dense vascular properties were evaluated in vivo in the chick chorioallantoic membrane
network is considered crucial to increase the rate of positive therapeutic model. Finally, as a proof of concept, the two developed bioink formu­
outcomes related to islet transplantation procedures. lations were co-printed at different spatial configurations. Thus, a bio­
To this end, pancreatic tissue engineering (TE) explored bio­ mimetic pancreatic-vascular 3D scaffold could be successfully
fabrication strategies focused on combining islet encapsulation into fabricated.
ECM-like hydrogels (e.g., fibrinogen [13], Matrigel [14], collagen [15])
with angiogenic factors (e.g., vascular endothelial growth factor (VEGF) 2. Results and discussion
[14] and platelet-derived growth factor (PDGF) [13], among others
[16], or porous scaffolds able to stimulate vascular ingrowth upon im­ 2.1. Ink formulations and rheological characterization
plantation [17]. In this frame, 3D-bioprinting (3D-BP) of pancreatic is­
lets has been proposed as a biofabrication method to engineer a suitable In this work, a co-axial microfluidic chip was coupled with a com­
biomimetic pancreatic microenvironment [18,19]. Indeed, 3D-BP offers mercial 3D bioprinter for the fabrication of bio-mimetic multi-material
the advantage of precisely depositing islet-laden ECM-like hydrogel scaffolds to simultaneously engineer both pancreatic and vessel-like
strands in a layer-by-layer fashion in order to fabricate 3D porous structures (Fig. 1A). Co-axial 3D-BP is a TE strategy that combines the
scaffolds with different geometries and pore sizes. For instance, microfluidic fabrication of cell-laden hydrogel-based microfibers with
Marchioli et al. used an extrusion-based system to 3D bioprint human layer-by-layer 3D deposition [34]. In the frame of microfluidic-assisted
islets in a wide range of alginate-based bioink formulations. They applications, alginate-based hydrogel formulations have been widely
demonstrated the positive effect of the introduction of porosity into employed due to their instantaneous gelation ability via calcium ions
hydrogel bulk on insulin secretion [20]. Duin and co-workers success­ crosslinking [35]. Indeed, the simultaneous delivery of both alginate
fully 3D bioprinted rat islets encapsulated in alginate blended with solution and calcium chloride allows the fabrication of highly-defined
methylcellulose [21]. Liu et al. further advanced the 3D-BP of islets by 3D porous structures [36]. However, despite its high suitability for
employing a co-axial system to fabricate 3D porous scaffolds by microfluidic applications, alginate lacks cell-binding sites. Thus, the
extruding murine islet-laden gelatin methacryoloyl (GelMA)/alginate maintenance of an appropriate micro-environment to support the
core-shell fibers, thus showing the control over islets spatial distribution long-term viability and functionality of encapsulated cells is limited.
[22]. Kim and co-workers demonstrated enhanced effect of liquefied Therefore, alginate has been combined with more biomimetic bio­
pancreatic dECM bioink on insulin secretion by islets; however, the polymers (i.e., chitosan, gelatin, GelMA) according to the specific tissue
powder dECM has not been applied in bioprinting of pancreatic islets yet engineering application [20,37,38]. In this work, alginate was used as a
[18]. templating agent, ensuring sufficient mechanical properties and high
Although it has been already reported that 3D porous scaffolds can shape fidelity, and tissue-specific bioactivity was introduced by blending
stimulate and promote efficient vascular ingrowth once such engineered with pancreatic dECM and fibrinogen-based tissue glue for engineering
tissues are implanted in vivo, this process requires up to 14 days to fully pancreatic and vessel-like structures, respectively (Fig. 1B).
develop [23,24]. To reduce the time required to achieve an appropriate DECM-powder was employed to closely mimic the complex molec­
vascular supply in close proximity of transplanted islets, several strate­ ular composition of the pancreatic microenvironment. The introduction
gies for co-culture and co-transplantation of vascular supporting cells of native-ECM components (e.g., collagen, laminin) into inert hydrogels
have been recently developed [16,25,26]. However, the integration of and the use of pristine solubilized pancreatic dECM improved the sur­
vascularized structures in islet-laden constructs by 3D-BP technologies vival endocrine function of pancreatic islets and insulin-producing cells
may be considered challenging, as different bioink formulations should [18,39,40]. However, liquid dECM is obtained by enzymatic solubili­
be designed to ensure a suitable tissue-specific microenvironment. zation of the decellularized tissue, thus yielding degradation of its native
Common approaches employed for the 3D-BP of multi-material and structure and mechanical strength [41,42]. Moreover, pepsin digestion
multi-cellular constructs involve the use of highly complex was reported to cause denaturation of some of the biochemical com­
multiple-printing heads apparatuses [27–29]. However, this results in ponents of dECM (e.g., proteins, growth factors) [43–45]. Therefore, in
an overall time-consuming fabrication process. To address this limita­ this study, pancreatic dECM in the form of powder was selected over
tion, recent advances in biofabrication strategies integrated 3D printing conventional liquid dECM bioink. The median of the volumetric particle
heads with a multi-inlet microfluidic platform, allowing a rapid size distribution of the dECM powder used in this study was equal 148 ±

2
J. Idaszek et al. Bioprinting 24 (2021) e00163

Fig. 1. A) Schematic of the experimental 3D-BP


setup. The microfluidic platform is adapted to fit
the bottom of the cartridge and connected with two
different digital pumps, loaded with pre-hydrogel
and crosslinking solution, respectively. The 3D-
printed fiber undergoes gelation as it is extruded
from the needle tip. B) Alginate-based tissue-spe­
cific pre-hydrogel formulations of cell-laden bio­
inks. C) Schematic model of a vascularized
pancreatic islet. D) Schematic of the microfluidic Y-
junction designed to simultaneously or alternatively
dispense the two formulated bioinks for the fabri­
cation of multi-layered heterogeneous structures.

10 μm. The DNA content was <50 ng per 1 mg of dECM powder, which and used as a control. After material and biological characterization,
corresponds to 1.25 ± 0.11% DNA content of the native pancreas. both bioink formulations were simultaneously 3D printed to reproduce a
Fibrinogen has been widely employed in vascular tissue engineering vascularized pancreatic microenvironment using a custom-made Y-chip
[46]. Indeed, in physiological conditions, fibrinogen is involved in the (Fig. 1C and D).
repairment of tissues and blood vessels after injury by forming fibrin Ink rheological characterization and 3D-printing of vascular and
clots upon thrombin-catalyzed enzymatic polymerization. Moreover, pancreatic scaffolds.
fibrin possesses an effective potential to induce the growth of blood Before 3D-printing, the rheological behaviour of the proposed bioink
vessels and the sprouting of newly formed capillaries (i.e., angiogenesis) formulations was tested by applying a shear rate ramp of 0.1–100 s− 1. As
in the proximity of in vivo implanted scaffolds [47–49]. shown in Fig. 2A, A_ECM showed a pronounced shear thinning behav­
Alginate-dECM powder and alginate-fibrinogen bioinks were deno­ iour, characterized by a decrease in viscosity over an increasing shear
ted as A_ECM and A_FBR, respectively. To further tailor physiochemical rate. Both A_PRS and A_FBR exhibited a Newtonian plateau below shear
properties of the A_FBR hydrogel, we performed secondary crosslinking rates of approximately 0.5 s− 1, followed by a slight shear thinning
with thrombin (A_FBR_T). Pristine alginate bioink was denoted as A_PRS behaviour. Moreover, it was observed that all bioink formulations

Fig. 2. A) Dynamic viscosity of the formulated bioinks employed in 3D-BP experiments as a function of shear rate. Viscosity of alginate is depicted as a reference
bioink. Effect of 3D-printing parameters on deposited fibers and morphological characterization of 3D-printed scaffolds: B) Effect of deposition speed (v) and C)
bioink flow rate (Q) on fiber diameter (D). D-F) Optical and macroscopic images of 3D-printed scaffolds: D) A_PRS, E) A_ECM, and F) A_FBR. G) Average fiber
diameter and H) average pore size of 3D printed scaffolds upon optimized 3D-printing parameters (Q = 15 μl min− 1, v = 2.5 mm s− 1); * – p < 0.05.

3
J. Idaszek et al. Bioprinting 24 (2021) e00163

exhibited high differences in terms of dynamic viscosity. Specifically, facilitated nutrients supply and waste removal of products within the
A_ECM presented the highest dynamic viscosity, revealing values 240- scaffold [50–52]. After the 3D-printing process, A_FBR scaffolds were
and 45-fold larger than A_PRS and A_FBR, respectively. A lower differ­ subjected to secondary crosslinking by immersing in thrombin solution
ence was detected comparing A_PRS to A_FBR, where A_FBR showed a (A_FBR_T). Not-crosslinked A_FBR scaffolds were used as a control
dynamic viscosity 3- to 6-fold larger than A_PRS. Despite those differ­ (A_FBR_C).
ences, all bioink formulations were easily extrudable by applying the Taken together, the aforementioned data clearly shows that our
same flow rate. Hence, well-defined hydrogel filaments could be formed. approach may be a good strategy for relatively fast and convenient
The effect of the deposition speed (v) and bioink flow rate (Q) on the fabrication of high-resolution 3D porous structures using bioinks char­
morphology of 3D-printed scaffolds was investigated for all bioinks acterized by inherently low printability and poor structural integrity (e.
formulations (Fig. 2B and C). In particular, fiber diameter was modu­ g., ECM and fibrinogen), further advancing the 3D-BP of soft hydrogels
lated by increasing v (i.e., 1–6 mm s− 1) and Q (i.e., 5, 10, 15, 20, 25 and [53–56].
30 μl min− 1). A linear increase (R2 > 0.9) of fiber diameter following the
increase of Q was observed for all bioink formulations. Conversely, a 2.2. Swelling test
linear decrease (R2 > 0.9) of fiber diameter was measured as the v
increased. These findings are in accordance with the volumetric flow A swelling test was performed to investigate the water uptake ca­
rate equation, i.e., Q = Av, where A is the fiber cross-section area. The pacity of the 3D printed scaffolds. The swelling ratio, expressed as a
diameter of the 3D printed fibers is directly dependent on the ink flow difference in weight of wet and dry samples normalized to the weight of
rate and inversely dependent on the deposition speed. Continuous and dry samples (ΔW%), is reported in Fig. 3A. A significantly lower
well-defined fibers were also obtained for most of the tested printing swelling ratio was observed for A_ECM (546.2 ± 55.7 %) compared to
parameters. The only exception was found for the lowest and the highest A_PRS (1180.2 ± 66.5 %). This finding is in contrast with results re­
deposition speeds (1 mm s− 1 and 6 mm s− 1, respectively). In the first ported in the literature, where liquid dECM increased the water ab­
case, filaments appeared undulating rather than fully stretched, while sorption capacity when combined with pristine hydrogels [57]. We
the highest speed reduced the chance of contact between 3D-printed hypothesize that the dECM powder incorporated in this study could
microfibers and the deposition plate. 3D scaffolds have been success­ interfere with alginate polymer chains resulting in a packed network
fully printed using the same optimized printing parameters. However, with limited polymer chain mobility, leading to a decrease in the overall
3D-printing of A_ECM presented more technical challenges when water penetration. Another possible explanation is the effect of
compared to the other ink formulations. Specifically, nozzle-clogging nano-topography of dECM powder obtained during the freeze-drying
episodes occurred more frequently due to the relevant viscosity of process, increasing its hydrophobicity despite its chemical composi­
A_ECM and the presence of dispersed ECM particles. For all other for­ tion [58]. This could be supported by the fact that we measured lower
mulations, the 3D-printing process moved forward without any major water uptake by freeze-dried alginate and alginate-based samples upon
issues over the long printing sessions (i.e., 2–3 h). incubation in DI water, suggesting permanent structural changes
3D-printed scaffolds were structurally stable and could be easily occurring during the lyophilization process (unpublished data). A
manipulated by means of a spatula. Moreover, macroscopic images different water absorption effect was observed when fibrinogen was
(Fig. 2D–F) revealed overall high printing accuracy, as confirmed by a incorporated into pristine alginate. Results showed a significantly higher
distinct 0◦ –90◦ square grid pattern. In addition, 3D-printed scaffolds A_FBR_C swelling ratio (1493.5 ± 80.7 %) compared to A_PRS. This
presented a fully interconnected structure characterized by well-defined could be attributed to the fibrinogen molecules disturbing the formation
square-shaped open pores. The investigation of fiber diameter (Fig. 2G) of intramolecular bonds between alginate chains and Ca2+ ions and the
showed a significantly higher fiber diameter for A_ECM scaffold (584.3 clearance of fibrinogen from the samples upon incubation in DI water
± 25.5 μm) compared to both A_PRS (445.2 μm ± 52.4) and A_FBR due to lack of thrombin crosslinking. This finding is confirmed by a
(406.3 ± 28.9 μm). The increase in fiber dimension may be likely significantly higher swelling ratio of A_FBR_C compared to values ob­
attributed to lower crosslinking efficiency upon Ca2+ due to the high tained for secondary crosslinked A_FBR_T (1013.1 ± 81.7 %), in which
concentration of ECM powder. Consequently, as shown in Fig. 2H, fibrinogen was enzymatically converted into fibrin. Moreover, 40 mM
thicker fibers resulted in a significant decrease in the pore size of A_ECM CaCl2 present in thrombin solution might induce a secondary cross­
scaffolds (360.8 ± 29.3 μm) compared to A_PRS (557.2 ± 52.4 μm) and linking effect also on the alginate network, thus further decreasing the
A_FBR (494.2 ± 29.9 μm). The obtained pore sizes and open porosity water uptake, as confirmed by significantly higher swelling ratio of
may be beneficial in terms of in vivo vascularization since it has been A_PRS compared to A_FRB_T [59].
reported that pore sizes >200 μm increase vascular cell invasion and
ultimately lead to vessel ingrowth. Moreover, pore size >500 μm

Fig. 3. Physical and mechanical characterization of 3D-printed scaffolds (A_PRS, A_ECM, A_FBR_C, A_FBR_T). A) Averaged swelling ratio (ΔW%) of 3D-printed
scaffolds after 3 days of immersion in DI water determined by measuring the mass of both wet and freeze-dried samples. B) Average elastic modulus (E) ob­
tained from mechanical compression test. The stiffness of A_ECM and A_FBR_T scaffolds was significantly higher than A_PRS (p < 0.05).

4
J. Idaszek et al. Bioprinting 24 (2021) e00163

2.3. Mechanical characterization 2.4. 3D-bioprinting of the pancreatic islets

Mechanical properties of all bioink formulations were investigated After thorough characterization of the developed ink in terms of
by a static mechanical compression test on samples hydrated for 3 days printability and the investigation of physicochemical properties of the
in DI water. Elastic modulus (E) values are reported in Fig. 3B. A_ECM 3D scaffolds, 3D-BP experiments with pancreatic islet-laden bioink were
exhibited significantly higher stiffness (2.3 ± 0.1 kPa) compared to performed. One of the major concerns related to the 3D-BP technique
A_PRS (1.0 ± 0.1 kPa). This increase could be induced by incorporating was the effect of shear stress on islet viability during extrusion of the
high concentration dECM powder, which created a denser and more A_ECM bioink. In Fig. 4A, results of live/dead (L/D) staining are pre­
compact structure than the A_PRS. Our finding is in accordance with the sented. Despite the overall difficulties in distinguishing islets from flake-
results obtained by Kim et al., where E of the formulated gelatin-based like dECM particles due to their green autofluorescence, the absence of
bioink was higher after dECM powder was incorporated [42]. On the red fluorescence led us to conclude that all islets survived the 3D-BP
other hand, the incorporation of fibrinogen did not increase mechanical process. This result was further confirmed by the evaluation of the
properties. Indeed, A_FBR_C showed a similar E value (1.2 ± 0.4 kPa) response of the islets to glucose fluctuations in a culture medium, as
compared to A_PRS. This is probably due to the dual effect introduced by shown in Fig. 4C. The GSIS assay was performed 24 h and 48 h after the
the possible partial clearance of fibrinogen due to the absence of 3D-BP. The difference in secretion of insulin by the islets encapsulated in
thrombin secondary crosslinking and a lower crosslinking of alginate A_PRS and A_ECM bioinks and the corresponding controls (i.e., the free-
chains resulting from lack of incubation with 40 mM CaCl2. This was floating islets designated as C_A_PRS and C_A_ECM) could be due to
confirmed by A_FBR_T, which showed a significantly higher E value (1.6 biological diversity, as the islets were isolated from two different ani­
± 0.3 kPa) compared to A_PRS. Indeed, secondary crosslinking intro­ mals in two independent isolation procedures. Interestingly, both types
duced by immersion in thrombin could keep fibrinogen inside the 3D of islets encapsulated in the bioinks produced in the low-glucose me­
printed scaffold, thus creating a packed polymeric network which may dium (LG) more insulin than the free-floating controls. Similar results
in turn increase E values under mechanical compression. Results are in were obtained by Marchioli and co-workers [9]. Although the islets
accordance with Duong et al., where higher E values were detected by encapsulated in the A_ECM bioink secreted in the LG over 3 times less
increasing thrombin concentration [60]. Moreover, the synergistic effect insulin than those bioprinted in A_PRS, both groups secreted a similar
of thrombin and 40 mM CaCl2 crosslinking was also reflected on algi­ concentration of insulin upon stimulation with high-glucose medium
nate, as confirmed by the significantly higher stiffness of A_FBR_T (HG). Therefore, the stimulation index (SI, i.e., the ratio of insulin
compared to A_PRS. Finally, A_ECM, A_FBR_C, and A_FBR_T showed secreted in a high glucose condition to the concentration secreted in a
mechanical properties similar to those of the native tissues they inten­ low glucose condition) was approximately 2-fold higher for the former
ded to mimic. In particular, elastic moduli of A_ECM, A_FBR_C, and (SI equal 2,8 ± 1,4 vs. 1,3 ± 0,2 respectively; see Fig. 4D). Those results
A_FBR_T are respectively in the range of the values found for pancreatic suggest loss of functionality by the islets bioprinted in the pristine
(1–5 kPa) [61] and endothelium native tissue (1.2–2 kPa) [62]. alginate. Unfortunately, after additional 24 h of culture, the

Fig. 4. Biological evaluation of the A_PRS and


A_ECM bioinks. A, B) Viability of islets after 3D-BP
in A_PRS (A) and A_ECM (B) bioinks determined by
means of L/D staining. The islets encapsulated in
both bioinks were viable (green) after the 3D-BP
process; no dead islets were detected (lack of red
fluorescence). Scale bar 200 μm. C) Secretion of
insulin upon stimulation with high-glucose medium
(HG, 16.7 mM of glucose) 24 h after the 3D-BP.
Both 3D bioprinted constructs and controls (free-
floating islets, C_A_PRS, C_A_ECM) contained
approximately 3000 IEQ isolated from two different
pancreases in two independent isolation proced­
ures. LG – low-glucose medium (2.5 mM of
glucose). D) Stimulation index (SI) measured for
islets encapsulated in controls, A_PRS and A_ECM
24 h after 3D-BP. E) Diffusion of glucose through
A_PRS and A_ECM disc; the diffusion followed
Fick’s law of diffusion. F) Diffusion of insulin
through A_PRS and A_ECM membranes; after 1 h,
the diffusion was significantly higher in the case of
A_ECM membrane.

5
J. Idaszek et al. Bioprinting 24 (2021) e00163

glucose-stimulated insulin secretion was significantly reduced for both needed for the diffusion of glucose molecules through the whole thick­
the 3D bioprinted and the free-floating islets, indicating loss of their ness of the hydrogel membrane (l), could not be determined from the
functionality (data not shown). Loss of functionality after 24 h of in vitro diffusion curve, as it was below 15 min. Based on extrapolation of the
culture, i.e., a decrease in SI below 2, has been reported for the linear region of the curve representing constant glucose diffusion flux,
free-floating islets by Wang and co-workers [63]. the lag time was estimated to be approximately 5 min and 11 min for the
To further investigate the enhanced endocrine function of the 3D A_PRS and A_ECM disc, respectively. Those values correspond to diffu­
bioprinted porcine islets, we measured diffusion of glucose and insulin sion coefficients D equal around 6.6 × 10− 6 cm2 s− 1 for A_PRS (3 % w/v)
through A_PRS and A_ECM membranes. The working hypothesis of this and 3.2 × 10− 6 cm2 s− 1 for A_ECM hydrogels. Both values are in the
experiment was that, although dECM particles increase the overall range of D determined for a blend of alginate (4 % w/v) with gelatin (5
density of the hydrogel, they could disrupt the alginate network, thus % w/v) equal 1.13 × 10− 6 cm2 s− 1 and water equal 6.7 × 10− 6 cm2 s− 1
increase its diffusivity. However, obtained results of glucose diffusion [20,64]. Moreover, the D of the A_PRS membrane, i.e., containing 3 %
led us to a different conclusion (see Fig. 4E). The lag time, i.e., the time (w/v) of alginate, was higher than the coefficient reported for

Fig. 5. Biological evaluation of A_FBR bio­


ink. After bioprinting, viability of cells was
visualized by means of L/D staining (A – all
cells, B – dead cells) and quantified using
MTS assay over 1 week of culture (C). The
MTS was performed for samples bioprinted
at two cell densities, i.e., 12 × 106 and 24 ×
106 cells per ml, which were either submit­
ted to secondary crosslinking with thrombin
(A_FBR_T) or left unmodified (A_FBR_C).
Morphology (D–F) and expression of CD31
marker by HUVEC (G–I) after 1 (D, E, G, H)
and 2 weeks (F, I) of in vitro culture. D and G
– A_FBR_C; E, F, H, I – A_FBR_T. Sprouting of
vessels (blue) in vitro from the 3D bioprinted
constructs after 2 weeks of culture in fibrin
hydrogel: J) A_FBR_C, K) A_FBR_T. Scale bar
100 μm.

6
J. Idaszek et al. Bioprinting 24 (2021) e00163

membrane made of 4 % (w/v; D = 5 × 10− 6 cm2 s− 1) [65]. On the other formation of EC intracellular junctions (indicated by an arrow on the
hand, the increased insulin diffusion through A_ECM discs (depicted in insert in Fig. 5H) [69]. This result could be a consequence of adhesion
Fig. 4F) cannot be explained by Fick’s law of diffusion, as it resembles motives presented by both fibrinogen and fibrin layer. Moreover, in case
the super-diffusion variant of anomalous diffusion. Increased insulin of scaffolds crosslinked with thrombin, a significant presence of
uptake was measured by Duin and coworkers in the case of alginate and HUVEC-bridging points between adjacent fibers was detected (see
methylcellulose discs, although no difference was observed for the Fig. 5H). This is likely due to the mature intercellular junction stabili­
glucose [21]. However, having taken into account the higher molecular zation occurring between adjacent cells and to the pro-angiogenic signal
weight and bigger hydrodynamic radius of an insulin molecule, the transduction resulting from the secreted ECM. Indeed, ECs acquired a tip
result obtained in the present study is surprising, and the underlying cell’s phenotype and form filopodia, leading to preliminary motility
mechanism of this phenomenon should be better understood. through the fibers [70,71]. 3D bioprinted scaffolds crosslinked with
Because the presence of dECM powder in the alginate-based hydrogel CaCl2 did not show HUVEC-bridging points, suggesting a
significantly reduced the diffusion of glucose, the enhanced function­ thrombin-mediated angiogenic feedback at the cell-surface interface.
ality of the 3D-printed islets could likely be attributed to the recapitu­ At week 2, much more CD31-positive cells were observed on the
lation of native niches and matrix-islet interactions provided by dECM surface of A_FBR_T fibers (Fig. 5G). Moreover, such cells revealed typical
powder. Similar results were obtained by Weber et al., who demon­ tight junctions-patterns at the intercellular level, which plays a pivotal
strated that incorporating biomolecules derived from pancreatic ECM (e. role in the adhesion of endothelial cells [72]. In this frame, confluent
g., collagen type IV and laminin) significantly improved secretion of HUVEC cells formed a monolayer on the hydrogel surface and guided
insulin compared to the bioinert hydrogel [39]. A similar trend was characteristic endothelial morphogenesis. In addition, established filo­
reported by Kim and colleagues, who showed improved functionality of podia contacts started tip cell anastomosis by fusion of endothelial
insulin-producing cells for increasing degrees of tissue biomimicry [18]. sprouts, and a microvascular network was formed (Fig. 5I). However,
Moreover, the results suggest that the small fiber diameter obtained in CD31-positive cells did not form a homogenous monolayer on the entire
this study (i.e., ~580 μm), combined with high open porosity, was suf­ surface of the constructs. This may be attributed to the different cell
ficient to overcome the mass transfer limitation observed by others in 3D attachment occurred over the whole cell-laden scaffolds [41,73,74]. In
plotted scaffolds (fiber diameter of 1.55 ± 0.13 mm) [20]. Improved case of A_FBR_C 3D bioprinted scaffolds, the loss of mechanical integrity
functionality of rat islets encapsulated in a small-diameter (<250 μm) occurred after 2 weeks of culture, making the confocal imaging of the
alginate-collagen fibers was demonstrated both in vitro and in vivo by constructs virtually impossible.
Jun and coworkers [66]. In vitro sprouting: After implanting in vivo, the scaffolds would be
covered by fibrin clots due to bleeding induced by the surgical proced­
2.5. 3D-bioprinting of vessel-like structures ure. Therefore, we wanted to investigate the sprouting of blood vessels
in such environment. To investigate this phenomenon, A_FBR_C and
Cell viability: LD staining of the HMSC/HUVEC-laden A_FBR scaffolds A_FBR_T 3D bioprinted constructs were embedded in fibrin hydrogel and
was performed after the 3D-BP process. Due to the high cell density, it incubated in co-culture medium for 2 weeks. Representative images of
was difficult to precisely number the overall amount of cells (green cells with fluorescently-labeled cell membranes are presented in Fig. 5J
stain, Fig. 5A). However, the low number of dead cells (depicted in and K, where consistent differences in cell density can be appreciated.
Fig. 5B) suggested relatively high cell viability, which we estimated to Secondary crosslinking significantly reduced the blood vessel sprouting
be around 80 % (based on the ratio of live cells (green stain - red stain from 3D constructs. This could be explained by the higher crosslinking
(G-R)) and all cells (i.e., live and dead cells, green stain (G)). To quan­ density of the A_FBR_T hydrogels due to incubation in thrombin/Ca2+
titatively investigate cell viability, an MTS assay was performed on day 1 solution. The secondary crosslinking induced the formation of fibrin
and day 7 of culture. Cell metabolic activity increased with the initial shells around the fibers and increased the crosslinking of alginate mol­
cell density. However, it did not change significantly over the culture ecules, both likely contributing to hindering the cell escape. Similar
period (see Fig. 5C). The thrombin-catalyzed fibrin polymerization did correlation between crosslinking density and cell release from alginate
not affect the MTS conversion except for the 3D scaffolds bioprinted at a fibers was observed in another study (unpublished data). Our findings
density of 24 × 106 cells⋅ml− 1 at day 7 of culture. This result could be suggest the potential of the secondary crosslinking to modulate cell
explained by significantly lower swelling of the samples crosslinked release kinetics from 3D bioprinted constructs.
with thrombin in the presence of Ca2+ ions due to a higher crosslinking
degree, which led to reduced diffusivity and lower cell metabolic ac­ 2.7. In ovo CAM assay
tivity [67,68]. Another reason for the reduced diffusion could be a layer
of cells that covered the surface of A_FBR_T fibers and thus reduced In ovo CAM assay on A_FBR scaffolds was performed to investigate
diffusion of nutrients into the fibers (see Fig. 5E). Of note, the cell sheet the potential in vivo angiogenic response induced by the transplanted
could also limit the accessibility of the MTS substrate to cells residing heterogeneous scaffolds. The chick embryo chorioallantoic membrane
inside the fibers; some of the MTS molecules could not diffuse through (CAM) is known to possess many advantages [75]. Compared to in vivo
the cell layer because they were irreversibly reduced to formazan upon experiments, CAM studies are cost-effective and allow immediate visu­
interacting with the cells. To overcome this issue in future, we will use alization. In addition, it can be considered an intermediate stage be­
an assay based on detection of ATP to measure cell viability. tween the in vitro and in vivo models, which enables effective screening
of novel biomaterial configurations. In this frame, CAM assay also pro­
2.6. CD31 expression vides reliable outcomes in terms of material-tissue biocompatibility at
the material interface, as it is commonly considered a potential target for
The early endothelialization of scaffolds containing pro-angiogenic the angiogenesis response [76].
cells was evaluated by means of immunocytochemical staining against In this study, the CAM assay was used to test the effect of the
CD31, a specific marker for endothelial cells (ECs). Moreover, staining hydrogel-based formulations on vascular sprouting and angiogenesis in
against F-actin was performed to visualize the cytoskeleton. To inves­ a physiological environment. Herein, cell-free and cell-laden scaffolds
tigate the angiogenesis of the constructs, long-culture experiments (up were investigated. Results showed the effect of secondary crosslinking
to 14 days) were performed. Confocal images of the cell-laden constructs and incorporation of cells on A_FBR scaffolds stability, as well as their
after 7 and 14 days of culture are depicted in Fig. 5. At week 1 (Fig. 5D, interaction with the membrane (Fig. 6). Evidence-based differences in
E, G, H), there was evidence of cell migration towards the fiber surface. scaffold integrity were observed. This was likely due to the different
Angiogenic activity on spreading cells was visible, as well as the crosslinking conditions. Indeed, secondary crosslinking with thrombin

7
J. Idaszek et al. Bioprinting 24 (2021) e00163

Fig. 6. In vivo evaluation of the samples in CAM assay. Representative images of A_FBR_C (A, B) and A_FBR_T (C, D) samples without cells (A, C) and with
encapsulated cells (B, D). Histological evaluation of the explanted 3D bioprinted constructs. H&E (E, G) and Sirius red (F, H) stainings were performed to visualize the
overall structure of the explanted samples, CAM, and the presence of blood vessels. Scale bar 200 μm (E–G) and 100 μm (H). SC – acellular A_FBR_T hydrogel scaffold;
SCC – cell-laden A_FBR_T hydrogel scaffolds; black arrows indicate vessels and grey arrow the interface between cell-laden hydrogel fiber and CAM.

reduced the swelling behaviour and increased the stability of the 3D Interestingly, the study showed no effective angiogenesis on neither
structures. This was confirmed by the dissolution of some A_FBR_C A_FBR_C nor A_FBR_T acellular scaffolds, suggesting the poor influence
samples during the incubation time in CAM (almost 20% of samples), of crosslinking methods at this stage of the investigation.
furtherly enhanced by the incorporation of cells (almost 40 % of dis­ The obtained results highlight the potential of the proposed bioinks
solved scaffolds). It could also be speculated that the higher presence of and hydrogel-based scaffolds for engineering heterogeneous tissues.
Ca2+ - alginate ionic bonds formed during the secondary crosslinking However, further investigation on the angiogenic potential and the
caused the overall increased stability of A_FBR_T scaffolds, as well as the ability of the encapsulated cells to anastomose with host vasculature
presence of fibrin shell surrounding the alginate-fibrinogen core (see should be undertaken.
swelling test, Fig. 3A). Moreover, secondary crosslinking with thrombin
likely improved the overall interactions between samples and CAM, as
highlighted in Fig. 6H. On the other hand, acellular scaffolds showed 2.8. Multi-material 3D-printing: a proof-of-concept
poor integration with the chorioallantoic membrane (Fig. 6E and F).
Interestingly, it was not possible to obtain histological sections of CAM After A_ECM and A_FBR were thoroughly investigated to ultimately
incubated with cell-laden A_FBR_C scaffolds due to the disintegration of validate the respective TE applications, the two developed bioinks were
the membrane. This finding suggested that increased dissolution of co-printed. As a proof-of-concept, this approach was addressed to
A_FBR_C samples led to the weakening of the CAM structure. Hence, the determine the potentiality of our 3D-printing method to recreate a
underlying cause should be investigated in future studies. Cell-laden bioartificial vascularized pancreatic microenvironment (Fig. 7A). In this
A_FBR_T scaffolds revealed a dense capillary network at the CAM frame, a microfluidic device bearing a Y-junction was employed to
interface (depicted in Fig. 6D), suggesting increased angiogenesis. The simultaneously or alternatively deliver A_ECM and A_FBR bioinks
macroscopic observation was confirmed by histological analysis (Fig. 6G (Fig. 7B and C). Three different scaffold configurations were 3D printed:
and H). The improved angiogenesis could be a result of the paracrine i) Janus-scaffold, obtained by the simultaneous delivery of A_ECM and
action of encapsulated cells. Indeed, HMSC secrete pro-angiogenic fac­ A_FBR; ii) alternating-layer scaffold, obtained by the alternative delivery
tors (i.e., FGF-2, VEGF, and IL-6) [77,78]. Thus, they could promote of either A_ECM or A_FBR bioinks, and iii) hybrid scaffold, which is a
tissue vascularization upon incorporation in engineered constructs. hybrid of Janus- and alternating-layer geometries.
Although all three scaffold configurations were successfully 3D

Fig. 7. Proof of concept of the multi-


material 3D printed scaffolds by using the
two developed hydrogel-based inks (A_FBR
and A_ECM). Geometries and representative
optical images of multi-material 3D printed
scaffolds, where material “A” indicates
A_FBR bioink and material “B” refers to
A_ECM formulation. Herein, AB/AB (A), A/B
(B), and AB/B (C) 2-layer scaffolds are dis­
played. (A) In the Janus scaffold geometry,
two filaments of both bioinks are simulta­
neously compartmentalized in the same 3D-
printed fiber, while in the alternating-layer
scaffold (B) the geometry was obtained by
alternating a layer of A_FBR with a layer of
A_ECM (A/B). The hybrid scaffold (C) was
obtained by alternating a Janus-layer with a
layer of A_ECM fibers.

8
J. Idaszek et al. Bioprinting 24 (2021) e00163

printed, the fabrication of alternating-layer scaffolds was more chal­ 150 rpm. The solution of the detergent was changed every 4 h. The ef­
lenging due to the inertial forces present in the A_ECM bioink, which did ficiency of the decellularization was evaluated by measuring the total
not allow the precise and controlled switching of A_ECM and A_FBR DNA content with a DNA quantification assay (Quant-iT PicoGreen
bioinks. This might lead to the fabrication of multiple layers not entirely dsDNA Assay kit, Invitrogen). The obtained dECM was frozen in liquid
composed of a single specific ink formulation. To overcome this issue, nitrogen, ground into fragments of about 0.5 cm, and subjected to the
higher flow rates up to 50 μl min− 1 were applied for a very short time (i. lyophilization at − 32 ◦ C. The obtained lyophilizate was subsequently
e., 10 s) prior to the 3D printing of the layer. However, an increase of ground in a cryogenic mill. The volumetric particle size distribution was
bioink flow rate induces higher shear stress, especially for high-viscous determined by means of diffraction aerosol spectrometer Spraytec
A_ECM, which may hamper cell viability and functionality. Therefore, (Malvern Instruments, UK) operating at a flow rate of 100 dm3 min− 1.
simultaneous delivery of A_ECM and A_FBR for 3D-printing of Janus-
microfiber scaffolds has been selected as a potential method to fabri­ 4.3. Ink preparation
cate vascularized-pancreatic bio-hybrid structures. Moreover, the Janus-
fiber configuration may provide a biological advantage by increasing the A_PRS was prepared by dissolving 3 % (w/v) alginate (2 % (w/v)
paracrine communication between pancreatic islets and HMSC/HUVEC high molecular weight, 1 % (w/v) low molecular weight) in HEPES
due to higher physical proximity than the other scaffold configuration. buffer (25 mM) containing 10 % (v/v) of fetal bovine serum (FBS) and 1
% of antibiotic-antimycotic solution (AA; 10 000 units⋅ml− 1 penicillin,
3. Conclusion 10 mg ml− 1 streptomycin and 25 μg ml− 1 amphotericin B).
A_ECM was prepared by dissolving 15 % (w/v) dECM porcine pow­
In this study, we have presented a novel approach for microfluidics- der in A_PRS. A_FBR was prepared by dissolving 9 % (w/v) fibrinogen in
assisted multi-material 3D bioprinting of vascularized pancreatic sub­ a solution of 3000 KIU aprotinin at 35 ◦ C. After complete dissolution, 1
stitutes. The incorporation of pancreatic dECM and fibrinogen into ml of fibrinogen solution was added under stirring condition to 1 ml of a
alginate enabled the development of two distinct tissue-specific bioinks 2-times concentrated solution of A_PRS to obtain a final concentration of
and 3D bioprinting of hydrogel scaffolds characterized with high shape A_PRS equal 3 % (w/v) and 4.5 % (w/v) of fibrinogen. The thrombin
fidelity and open porosity. The bioinks were suitable for the 3D bio­ solution used for the post-process crosslinking of A_FBR was prepared by
printing of islets- and HMSC/HUVEC-laden constructs and supported dissolving 500 U/ml of thrombin in a solution of 40 mM CaCl2.
endocrine function of porcine pancreatic islets (secretion of insulin upon
glucose stimulation) and vascularization (formation of intercellular 4.4. Rheology
junctions and vessel-like structures by CD31-positive cells). Secondary
crosslinking with thrombin in the presence of Ca2+ increased the sta­ The rheological properties of A_PRS, A_FB, and A_ECM were inves­
bility of the A_FBR hydrogels and enabled the modulation of in vitro tigated by using an ARES rheometer (TA Instruments, Delaware, USA)
vessel sprouting, which was higher in the absence of secondary cross­ with cone and plate geometry, setting the gap at 51 μm. Shear viscosity
linking. On the other hand, the higher angiogenic potential in vivo was behaviour was evaluated by applying shear rates ranging from 0.1 to
demonstrated by more stable hydrogel scaffolds in CAM assay. 100 s− 1. All measurements were performed at a fixed temperature of
To better understand the effect of secondary crosslinking on angio­ 25 ◦ C, controlled by a Peltier system on the plate.
genesis, further work will include evaluation of the A_FBR scaffolds by
means of prolonged CAM assay. Moreover, we will investigate the 4.5. 3D-bioprinting set up
impact of paracrine activity of HMSC/HUVEC on islets functionality by
co-printing the islets and cells within one scaffold. Another aspect to be Design of the microfluidic printing head: 3D-bioprinting was performed
pursued in the future study is the effect of different sizes of dECM par­ by coupling a microfluidic platform with a 3D-printer (3D-Bioplotter™
ticles on the 3D-BP and behaviour of pancreatic islets. System, EnvisionTEC, Germany). The microfluidic platform was
In conclusion, the presented results can be considered a significant designed with a Y-junction (2 inlets, 1 outlet) to be purposely compat­
advancement both in the field of biofabrication of vascularized ible with simultaneous or alternative multi-material deposition. The
pancreatic substitutes and microfluidic-assisted multi-material/multi- microfluidic device was fabricated as previously reported [34]. The
cellular 3D bioprinting, as they expand the biofabrication window by outlet of the microfluidic platform was fluidically connected to a
application of alginate-based biomimetic bioinks. co-axial nozzle system formed by inner needle (21G) and outer needle
(16G), as already described in previous studies [32,36,79]. Finally, the
4. Experimental section dispensing co-axial system was screwed at the bottom of a
low-temperature cartridge of 3D-Bioplotter.
4.1. Materials Dispensation of the bioink: In multi-material 3D-printing, both bioinks
were loaded into 3 ml syringes subsequently placed into programmable
Sodium alginate with high molecular weight (75–200 kDa, PRO­ microfluidic syringe pumps (neMESYS low pressure, CETONI GmbH),
NOVA UP LVG) and low molecular weight (<75 kDa, PRONOVA UP and the CaCl2 solution (0.2 M) was accommodated on a single-channel
VLVG) were purchased from Novamatrix (Norway). Fibrinogen and microfluidic pump (New Era Pump Systems, Inc). Inks and CaCl2 were
thrombin were purchased from Baxter (Austria) as a part of tissue glue connected, respectively, to the inlets of the microfluidic system and to
kit Tisseel Lyo. Calcium chloride (CaCl2) was purchased from Eurochem the outer of the co-axial nozzle system by polyethylene (PE) microfluidic
BGD (Poland). tubings. In case of single-bioink 3D-printing, only one inlet of the Y-
junction was used and connected to the system. Once either A_ECM or
4.2. Decellularization of porcine pancreas A_FBR came in contact with CaCl2 at the tip of the co-axial system, the
instantaneous gelation process occurred.
Pig pancreases (total weight of 5 kg) were collected at a local
slaughterhouse. After harvesting, all organs were frozen at − 20 ◦ C. Prior 4.6. 3D-bioprinting
the decellularization process, the pancreases were thawed, manually
cleaned of fat and rinsed with streptomycin solution (0.1 mg ml− 1). The The 3D-printed fiber diameter was investigated by increasing the
decellularization process was carried out by immersing the pancreases deposition speed (i.e., 1–6 mm s− 1) and ink flow rate (i.e., 5, 10, 15, 20,
in a 1 % (v/v) solution of Triton X-100 in PBS and placing it in an 25, and 30 μl min− 1). Microfibers were printed on cover glasses, and
incubator shaker (Eppendorf Innova) maintained at 4 ◦ C and mixed at microfiber diameter was measured using a confocal microscope (Leica

9
J. Idaszek et al. Bioprinting 24 (2021) e00163

TCS SP8). Five microfibers were measured for each combination of pa­ entire isolation procedure was done using Ricordi’s chamber. The iso­
rameters and each ink formulation. lated pancreatic islets were pooled and stored at 4 ◦ C until the start of
3D printed scaffolds were designed to have dimensions correspond­ the 3D-BP process.
ing to 1 × 1 × 0.3 cm and 0◦ –90◦ orientation. Inks and CaCl2 were
dispensed at flow rates of 15 and 8.5 μl min− 1, respectively. The printing 4.11. Cell culture
speed was set to 2.5 mm s− 1. Hydrogel microfibers were printed as
continuous filaments with a fiber distance of 900 μm and 190 μm layer Human bone marrow-derived mesenchymal stem cells (HMSC) were
thickness. Immediately after 3D-printing, A_FBR_T was subjected to obtained from Dr. Jan E. Brinchman of Oslo University Hospital. The
secondary crosslinked by incubation in thrombin solution at 37 ◦ C for cells were isolated and validated as HMSC according to protocols
15 min. Scaffolds without secondary crosslinking (A_FBR_C) were used developed in the past [80–82] and expanded in a medium consisting of
as a control. Fiber diameter and pore size of the 3D printed scaffolds α-MEM (Gibco, UK) supplemented with 10 % fetal bovine serum (FBS,
were measured using a confocal microscope. Five different samples (n = Biowest, South America origin), 1 % of antibiotics (10′ 000 U ml− 1
5) were analyzed for each ink formulation. penicillin, 10 μg ml− 1 streptomycin; Gibco, USA) and 1 ng ml− 1 human
Multi-material 3D printing: As a proof of concept, the fabrication of a basic fibroblast growth factor 2 (both Sigma-Aldrich). Human umbilical
multi-material 3D printed scaffold was also performed. To obtain Janus- vein endothelial cells (HUVEC) were purchased from Lonza and cultured
fiber constructs, A_FBR and A_ECM were simultaneously delivered at in a dedicated medium (EGM-2 Endothelial Medium Bullet Kit, Lonza).
flow rates of 10 and 5 μl min− 1, respectively. To obtain 3D scaffolds Cells used in the 3D-BP experiments were from passages 4 and 5. For the
formed by alternating layers of A_ECM and A_FBR, inks were alterna­ co-culture experiments, a mixture of EGM-2 and supplemented α-MEM
tively delivered at 15 μl min− 1. at ratio 4:1 was used.

4.7. Swelling test 4.12. 3D-bioprinting process of islets- and cell-laden scaffolds

To investigate water absorption capacity, 3D printed scaffolds were For the bioinks preparation, A_PRS and CaCl2 (0.2 M) were sterile-
weighed after 72 h of immersion in DI water at 37 ◦ C to obtain swollen filtered (pore Ø = 0.22 μm). The other bioink components (i.e., 2×
weight (Ws). Samples were then frozen at − 80 ◦ C overnight, lyophilized A_PRS, fibrinogen, and dECM powder) were provided already sterile.
for 1 day, and weighed again to obtain dry weight (WD). Water ab­ Islets-laden A_PRS and A_ECM bioinks were prepared by mixing A_PRS
sorption capacity (ΔW%) was obtained as: ((Ws- WD)/WD) × 100. and A_ECM with the islets at density, ensuring approximately 3000 IEQ
per construct. Cell-laden A_FBR bioink was prepared by mixing A_FBR
4.8. Mechanical characterization with HUVECs and HMSC at 1:2 ratio [83,84], respectively, to achieve a
final density of 2.4 × 107 cells mL− 1. The as-prepared bioinks were
The compressive mechanical test was performed on 3D-printed loaded into syringes and then 3D-bioprinted with the same printing
scaffolds (n = 4 per ink formulation) after 72 h-incubation in DI water parameters used for acellular inks.
using a Dynamic Mechanical Analyzer (DMA Q800, TA instruments).
Mechanical tests were conducted at 37 ◦ C with a pre-load of 0.001 N, by 4.13. Evaluation of viability
applying a compressive strain ramp at 2.5 % min− 1 until 30 % strain.
From stress-strain (σ–ε) curves, elastic modulus (E) was calculated as the Live/dead (L/D) staining: Islets and cell viability was evaluated post-
slope in the 0–5% strain range, R2 > 0.9). printing using live and dead staining. Briefly, the printed islets and cell-
laden constructs were incubated in a solution of acridine orange (Sigma,
4.9. Diffusion of glucose and insulin 3 μg mL− 1, green fluorescence, all cells (G)) and propidium iodide
(Sigma, 10 μg mL− 1, red fluorescence, dead cells (R)) dissolved in 25 mM
Solid hydrogel discs with a diameter of 15 mm were fabricated using HEPES for approximately 5 min and analyzed with a fluorescent mi­
ion eluting molds. Briefly, 200 μl of either A_PRS or A_ECM bioinks was croscope (Leica TCS SP8) at wavelengths corresponding to fluorophores
cast into molds made of agarose (1 % (w/v) in deionized (DI) water) and of interest. The cells visible in green and red were counted, and the
immersed in 0.2 M CaCl2 for 25 min. The surface of the mold was viability was calculated according to the formula: % viability = 100 %⋅
covered with MCE membrane (MF-Millipore). The samples were washed (G-R)/G.
with DI water and placed between 2 rubber seals in an in-house made MTS assay: The HMSC/HUVEC-laden bioprinted constructs were
diffusion chamber. Porcine insulin (Sigma) was dissolved in a RPMI washed with α-MEM w/o FBS and moved to new wells containing 1 ml of
1640 (Gibco, Thermo Fisher) medium at a concentration of 2.5 μg ml− 1 pre-incubated α-MEM w/o FBS. Subsequently, 200 μL of MTS (Cell Titer
and pipetted into one of the chambers. RPMI 1640 medium without 96 Aqueous One Solution Cell Proliferation Assay, Promega, USA) was
glucose (Gibco, Thermo Fisher) was added to the other chamber; to added, and the samples were incubated at 37 ◦ C and 5 % CO2 for 1 h,
measure the diffusion of glucose and insulin, 0.5 ml of the medium was followed by the addition of 250 μL of 10 % SDS and shaking for 30 min
removed at 15 min intervals for up to 1 h. According to the manufac­ 100 μL, aliquots of the supernatant were transferred quadruplicates into
turer’s protocols, glucose concentration was determined using glucose a 96-well plate, and the absorbance was measured at λ = 490 nm.
colorimetric detection assay (Invitrogen) and insulin by means of
porcine insulin ELISA assay (Invitrogen). 4.14. Evaluation of islets functionality

4.10. Isolation of pancreatic islets Glucose-stimulated insulin secretion (GSIS): The solution of 2.5- and
16.7 mM glucose was prepared by diluting glucose in sterile Creb’s
Pancreatic islets were isolated from the pancreases of breeding pigs, buffer. Before use, the solutions were heated to 37 ◦ C, and the time zero
which died as a result of bleeding out within 2 h of the organs being samples were taken from both solutions. Islets-laden scaffolds were
harvested. The entire process of preparing the organs for isolation was placed on the inserts and submerged in 2.5 mM glucose solution. After
carried out under hypothermic conditions. After cleansing of the fatty 30 min, inserts were dried on tissue paper and placed in 16.7 mM
tissue, the pancreas was injected with a solution of NB8 collagenase at a glucose solution for 60 min. This was followed by drying inserts on
concentration of 2.73 mg of collagenase per 1 g of pancreas and neutral tissue paper and placing them in 2.5 mM glucose solution for another 60
protease (100 DMC-U). The solution was administered directly into the min. During the entire assay, the plates were incubated in the cell cul­
Wirsung’s duct. Then, the organ was mechanically fragmented. The ture incubator at 37 ◦ C, and 100 μL of supernatant was taken every 15

10
J. Idaszek et al. Bioprinting 24 (2021) e00163

min and frozen at − 80 ◦ C for further analysis by means of ELISA assay CRediT authorship contribution statement
(Demeditec Diagnostics GmbH (Insulin ELISA ref: DE2935) according to
manufacturer’s protocol. Joanna Idaszek: Conceptualization, Methodology, Investigation,
Writing – original draft, Writing – review & editing, Visualization, Su­
4.15. Evaluation of cell morphology and formation of endothelial cell pervision. Marina Volpi: Methodology, Investigation, Formal analysis,
intercellular junctions Writing – original draft, Writing – review & editing, Visualization.
Alessia Paradiso: Methodology, Investigation, Formal analysis, Writing
The cytoskeleton was visualized by staining of F-Actin (Alexa Fluor – original draft, Writing – review & editing, Visualization. Martyna
Phalloidin). Scaffolds were fixed with 4 % paraformaldehyde for 30 min Nguyen Quoc: Investigation. Żaneta Górecka: Investigation. Marta
and then washed with HEPES thrice, followed by permeabilization with Klak: Conceptualization, Investigation, Methodology. Grzegorz
0.2 % Triton X-100 (v/v) in HEPES for 15 min. In order to block un­ Tymicki: Investigation, Methodology. Andrzej Berman: Investigation,
specific binding, the constructs were incubated in 10 % (v/v) goat serum Methodology. Mateusz Wierzbicki: Investigation, Methodology,
(GS) for 30 min at RT. Subsequently, the samples were incubated with Writing – original draft, (CAM model). Sławomir Jaworski: Investiga­
rabbit polyclonal anti-CD31 primary antibody (1:20, Abcam ab28364) tion, Methodology. Marco Costantini: Methodology. Agnieszka
and Alexa Fluor 546 phalloidin (1:40, Invitrogen) overnight at 4 ◦ C. Kępczyńska: Methodology, Investigation. Ewa Sawosz Chwalibóg:
After washing thrice with HEPES, scaffolds were furtherly incubated Supervision, Resources. Michał Wszoła: Conceptualization, Investiga­
with Alexa Fluor 488 anti-rabbit secondary antibody produced in goat tion, Supervision, Resources, Funding acquisition. Wojciech
(1:300) for 3 h in the darkness at RT. Finally, cell nuclei were stained Święszkowski: Conceptualization, Supervision, Resources, Funding
with Draq5 solution (1:1000, Thermo Scientific) and incubated for 15 acquisition.
min in the dark and washed thrice with HEPES before imaging by
confocal microscope (Nikon A1R).
Declaration of competing interest

4.16. In vitro vessel sprouting


The authors declare that they have no known competing financial
interests or personal relationships that could have appeared to influence
The 3D-bioprinted scaffolds were embedded in fibrin matrices
the work reported in this paper.
(TISSEEL, Baxter) with a final concentration of 2.5 mg/ml fibrinogen
and crosslinked with 0.2 IU/ml thrombin for 30 min. The cell-laden
Acknowledgments
scaffolds were cultured in co-culture medium and fixed with 4 % para­
formaldehyde buffered with HEPES after 2 weeks. Prior to the obser­
This work was supported by the National Centre for Research and
vations, the cells were labeled with DiD dye (Molecular Probes) and
Developments in the frame of the STRATEGMED program (contract no.
washed extensively with HEPES. Due to the high opacity of the samples,
STRATEGMED3/305813/2/NCBR/2017). The authors would like to
glycerine was applied to improve the visibility of the embedded cells
acknowledge Katarzyna Kosowska, Tomasz Bryniarski, Piotr Cywoniuk,
[85]. The constructs were analyzed with a confocal microscope (Leica
Paweł Turowski, and Magdalena Gomółka, all of Foundation of Research
TCS SP8) at excitation wavelengths of 633 nm.
and Science Development, for their assistance in preparation of
pancreatic dECM, isolation of pancreatic islets, and GSIS assay. We
4.17. Evaluation of angiogenic properties of the bioinks by means of in
would also like to thank Mariusz Nyc of the Faculty of Materials Science
ovo CAM assay
and Engineering, Warsaw University of Technology, for his assistance in
rheological measurements.
Fertilized eggs from Ross line 308 hens were obtained from a certi­
fied hatchery. The eggs were cleaned, sterilized with UVC light and
divided into four groups (4 × 15 eggs). Embryos were incubated at References
standard conditions (temperature 37 ◦ C, humidity 60 %, and turned
[1] D.W. Scharp, P. Marchetti, Adv. Drug Deliv. Rev. 67–68 (2014) 35.
once per hour). At day 9 of embryonic development, scaffolds were [2] J.D. Weaver, D.M. Headen, M.M. Coronel, M.D. Hunckler, H. Shirwan, A.J. García,
placed on the chorioallantoic membrane (CAM). A hole of approxi­ Am. J. Transplant. 19 (2019) 1315.
[3] V. Vaithilingam, B.E. Tuch, Rev. Diabet. Stud. 8 (2011) 51.
mately 1.5 cm was made in the shell and, subsequently, the inner
[4] E.A. Ryan, J.R.T. Lakey, R.V. Rajotte, G.S. Korbutt, T. Kin, S. Imes, A. Rabinovitch,
membrane was gently stripped off, and a scaffold was placed on CAM. J.F. Elliott, D. Bigam, N.M. Kneteman, et al., Diabetes 50 (2001) 710.
Chicken embryos were incubated to day 11 of embryonic development [5] A.M.J. Shapiro, J.R.T. Lakey, E.A. Ryan, G.S. Korbutt, E. Toth, G.L. Warnock, N.
when scaffolds with CAM were prefixed with 1 ml of 4 % para­ M. Kneteman, R.V. Rajotte, N. Engl. J. Med. 343 (2000) 230.
[6] M. Wszola, A. Berman, L. Gorski, A. Ostaszewska, M. Serwanska-Swietek,
formaldehyde. After 5 min of incubation, CAM with implants were cut M. Krajewska, A. Lipinska, A. Chmura, A. Kwiatkowski, Transplant. Proc. 50
out and fixed at 4 ◦ C in 4 % paraformaldehyde for 60 min. Scaffolds with (2018) 2119.
CAM were imaged using stereomicroscope under a 6.3-fold magnifica­ [7] A. Berman, M. Wszola, L. Gorski, M. Serwanska-Swietek, A. Ostaszewska,
A. Lipinska, M. Durlik, A. Chmura, A. Kwiatkowski, Transplant. Proc. 51 (2019)
tion (SZ×10, CellD software version 3.1; Olympus Corporation, Tokyo, 2775.
Japan). Subsequently, the samples were incubated in a gradient of su­ [8] J. Beck, R. Angus, B. Madsen, D. Britt, B. Vernon, K.T. Nguyen, Tissue Eng. 13
crose (10, 20, and 30 w/v%), embedded in Shandon Cryomatrix (2007) 589.
[9] G. Marchioli, L. Van Gurp, P.P. Van Krieken, D. Stamatialis, M. Engelse, C.A. Van
(Thermo Scientific), and cut with a cryostat (Microm HM 550) to yield Blitterswijk, M.B.J. Karperien, E. De Koning, J. Alblas, L. Moroni, et al.,
10 μm sections. The histological sections were stained with hematoxylin Biofabrication 7 (2015), https://doi.org/10.1088/1758-5090/7/2/025009.
and eosin (H&E) or Sirius red, followed by examination using light [10] M. Brissova, A.C. Powers, Diabetes 57 (2008) 2269.
[11] S. Paraskevas, D. Maysinger, R. Wang, W.P. Duguid, L. Rosenberg, Pancreas 20
microscope (Zeiss Axio Scope A.1).
(2000) 270.
[12] A.R. Pepper, B. Gala-Lopez, O. Ziff, A.M.J. Shapiro, Clin. Dev. Immunol. 2013
4.18. Statistical analysis (2013) 352315.
[13] M. Najjar, V. Manzoli, M. Abreu, C. Villa, M.M. Martino, R.D. Molano, Y. Torrente,
A. Pileggi, L. Inverardi, C. Ricordi, et al., Biotechnol. Bioeng. 112 (2015) 1916.
Results are expressed as means ± standard deviation. Two-way [14] A. Golocheikine, V. Tiriveedhi, N. Angaswamy, N. Benshoff, R. Sabarinathan,
ANOVA with Tukey’s multiple comparison tests was performed using T. Mohanakumar, Transplantation 90 (2010) 725.
GraphPad Prism ® analysis software (GraphPad Software, La Jolla, CA) [15] X. Wang, K. Wang, W. Zhang, M. Qiang, Y. Luo, Biomaterials 138 (2017) 80.
[16] D.T. Bowers, W. Song, L.H. Wang, M. Ma, Acta Biomater. 95 (2019) 131.
to investigate statistical differences between data populations. Differ­ [17] Y.C. Chiu, M.H. Cheng, H. Engel, S.W. Kao, J.C. Larson, S. Gupta, E.M. Brey,
ences are displayed as statistically significant when p < 0.05. Biomaterials 32 (2011) 6045.

11
J. Idaszek et al. Bioprinting 24 (2021) e00163

[18] J. Kim, I.K. Shim, D.G. Hwang, Y.N. Lee, M. Kim, H. Kim, S.W. Kim, S. Lee, S. [50] J.X. Lu, B. Flautre, K. Anselme, P. Hardouin, A. Gallur, M. Descamps, B. Thierry,
C. Kim, D.W. Cho, et al., J. Mater. Chem. B 7 (2019) 1773. J. Mater. Sci. Mater. Med. 10 (1999) 111.
[19] D.J. Ravnic, A.N. Leberfinger, I.T. Ozbolat, Trends Biotechnol. 35 (2017) 1025. [51] Y. Kuboki, Q. Jin, M. Kikuchi, J. Mamood, H. Takita, Connect. Tissue Res. 43
[20] G. Marchioli, L. Van Gurp, P.P. Van Krieken, D. Stamatialis, M. Engelse, C.A. Van (2002) 529.
Blitterswijk, M.B.J. Karperien, E. De Koning, J. Alblas, L. Moroni, et al., [52] T.M.G. Chu, D.G. Orton, S.J. Hollister, S.E. Feinberg, J.W. Halloran, Biomaterials
Biofabrication 7 (2015), https://doi.org/10.1088/1758-5090/7/2/025009. 23 (2002) 1283.
[21] S. Duin, K. Schütz, T. Ahlfeld, S. Lehmann, A. Lode, B. Ludwig, M. Gelinsky, Adv. [53] F. Pati, J. Jang, D.H. Ha, S.W. Kim, J.W. Rhie, J.H. Shim, D.H. Kim, D.W. Cho, Nat.
Healthc. Mater. 8 (2019) 1. Commun. 5 (2014), https://doi.org/10.1038/ncomms4935.
[22] X. Liu, S.S.D. Carter, M.J. Renes, J. Kim, D.M. Rojas-Canales, D. Penko, C. Angus, [54] Y.J. Choi, T.G. Kim, J. Jeong, H.G. Yi, J.W. Park, W. Hwang, D.W. Cho, Adv.
S. Beirne, C.J. Drogemuller, Z. Yue, et al., Adv. Healthc. Mater. 8 (2019) 1. Healthc. Mater. 5 (2016) 2636.
[23] E.C. Novosel, C. Kleinhans, P.J. Kluger, Adv. Drug Deliv. Rev. 63 (2011) 300. [55] D. Choudhury, H.W. Tun, T. Wang, M.W. Naing, Trends Biotechnol. 36 (2018) 787.
[24] J. Gilbert-Honick, W. Grayson, Adv. Healthc. Mater. 9 (2020) 1. [56] W.J. Kim, H. Lee, J.U. Lee, A. Atala, J.J. Yoo, S.J. Lee, G.H. Kim, Biomaterials 230
[25] B.J. Oh, S.M. Jin, J.M. Choi, S.H. Oh, W. Shim, M.S. Lee, M.K. Lee, J.H. Kim, Am. J. (2020) 119632.
Transplant. 15 (2015) 1543. [57] B. Nasiri, S. Mashayekhan, Biologicals 48 (2017) 39.
[26] J.P.M.C.M. Cunha, G. Leuckx, P. Sterkendries, H. Korf, G. Bomfim-Ferreira, [58] D.M. Spori, T. Drobek, S. Zürcher, M. Ochsner, C. Sprecher, A. Mühlebach, N.
L. Overbergh, B. Vaes, H. Heimberg, C. Gysemans, C. Mathieu, Diabetologia 60 D. Spencer, Langmuir 24 (2008) 5411.
(2017) 134. [59] C.E. Vorwald, T. Gonzalez-Fernandez, S. Joshee, P. Sikorski, J.K. Leach, Acta
[27] N. Ashammakhi, S. Ahadian, C. Xu, H. Montazerian, H. Ko, R. Nasiri, N. Barros, Biomater. 108 (2020) 142.
A. Khademhosseini, Mater. Today Bio 1 (2019), https://doi.org/10.1016/j. [60] H. Duong, B. Wu, B. Tawil, Tissue Eng. 15 (2009) 1865.
mtbio.2019.100008. [61] J. Liu, H. Zheng, P. Poh, H.-G. Machens, A. Schilling, Int. J. Mol. Sci. 16 (2015)
[28] T.K. Merceron, M. Burt, Y.J. Seol, H.W. Kang, S.J. Lee, J.J. Yoo, A. Atala, 15997.
Biofabrication 7 (2015), https://doi.org/10.1088/1758-5090/7/3/035003. [62] T.R. Cox, J.T. Erler, DMM Dis. Model. Mech. 4 (2011) 165.
[29] J.H. Kim, Y.J. Seol, I.K. Ko, H.W. Kang, Y.K. Lee, J.J. Yoo, A. Atala, S.J. Lee, Sci. [63] X. Wang, K. Wang, W. Zhang, M. Qiang, Y. Luo, Biomaterials 138 (2017) 80.
Rep. 8 (2018) 1. [64] L.G. Longsworth, J. Am. Chem. Soc. 75 (1953) 5705.
[30] F. Maiullari, M. Costantini, M. Milan, V. Pace, M. Chirivì, S. Maiullari, A. Rainer, [65] B.J.M. Hannoun, G. Stephanopoulos, Biotechnol. Bioeng. 28 (1986) 829.
D. Baci, H.E.S. Marei, D. Seliktar, et al., Sci. Rep. 8 (2018) 1. [66] Y. Jun, M.J. Kim, Y.H. Hwang, E.A. Jeon, A.R. Kang, S.H. Lee, D.Y. Lee,
[31] M. Costantini, S. Testa, P. Mozetic, A. Barbetta, C. Fuoco, E. Fornetti, F. Tamiro, Biomaterials 34 (2013) 8122.
S. Bernardini, J. Jaroszewicz, W. Święszkowski, et al., Biomaterials 131 (2017) 98. [67] W.S. Toh, T.C. Lim, M. Kurisawa, M. Spector, Biomaterials 33 (2012) 3835.
[32] C. Colosi, S.R. Shin, V. Manoharan, S. Massa, M. Costantini, A. Barbetta, M. [68] L. Bian, C. Hou, E. Tous, R. Rai, R.L. Mauck, J.A. Burdick, Biomaterials 34 (2013)
R. Dokmeci, M. Dentini, A. Khademhosseini, Adv. Mater. 28 (2016) 677. 413.
[33] J. Idaszek, M. Costantini, T.A. Karlsen, J. Jaroszewicz, C. Colosi, S. Testa, [69] J.R. Privratsky, P.J. Newman, Cell Tissue Res. 355 (2014) 607.
E. Fornetti, S. Bernardini, M. Seta, K. Kasarełło, et al., Biofabrication 11 (2019), [70] L.K. Phng, H. Gerhardt, Dev. Cell 16 (2009) 196.
https://doi.org/10.1088/1758-5090/ab2622. [71] L. Jakobsson, C.A. Franco, K. Bentley, R.T. Collins, B. Ponsioen, I.M. Aspalter,
[34] M. Costantini, S. Testa, P. Mozetic, A. Barbetta, C. Fuoco, E. Fornetti, F. Tamiro, I. Rosewell, M. Busse, G. Thurston, A. Medvinsky, et al., Nat. Cell Biol. 12 (2010)
S. Bernardini, J. Jaroszewicz, W. Święszkowski, et al., Biomaterials 131 (2017) 98. 943.
[35] X.Y. Du, Q. Li, G. Wu, S. Chen, Adv. Mater. 31 (2019) 1. [72] L. Liu, G.P. Shi, Cardiovasc. Res. 94 (2012) 3.
[36] M. Costantini, J. Idaszek, K. Szöke, J. Jaroszewicz, M. Dentini, A. Barbetta, J. [73] N.E. Fedorovich, W. Schuurman, H.M. Wijnberg, H.J. Prins, P.R. Van Weeren,
E. Brinchmann, W. Świȩszkowski, Biofabrication 8 (2016), https://doi.org/ J. Malda, J. Alblas, W.J.A. Dhert, Tissue Eng. C Methods 18 (2012) 33.
10.1088/1758-5090/8/3/035002. [74] H.W. Kang, S.J. Lee, I.K. Ko, C. Kengla, J.J. Yoo, A. Atala, Nat. Biotechnol. 34
[37] T. Fischetti, N. Celikkin, N. Contessi Negrini, S. Farè, W. Swieszkowski, Front. (2016) 312.
Bioeng. Biotechnol. 8 (2020) 1. [75] K.H. Kain, J.W.I. Miller, C.R. Jones-Paris, R.T. Thomason, J.D. Lewis, D.M. Bader,
[38] C. Rinoldi, M. Costantini, E. Kijeńska-Gawrońska, S. Testa, E. Fornetti, M. Heljak, J.V. Barnett, A. Zijlstra, Dev. Dynam. 243 (2014) 216.
M. Ćwiklińska, R. Buda, J. Baldi, S. Cannata, et al., Adv. Healthc. Mater. 8 (2019) [76] P. Nowak-Sliwinska, T. Segura, M.L. Iruela-Arispe, Angiogenesis 17 (2014) 779.
1. [77] H. Tao, Z. Han, Z.C. Han, Z. Li, Stem Cell. Int. (2016) 2016, https://doi.org/
[39] L.M. Weber, K.N. Hayda, K.S. Anseth, Tissue Eng. 14 (2008) 1959. 10.1155/2016/1314709.
[40] P. Shrestha, S. Regmi, J.H. Jeong, J. Pharm. Investig. 50 (2020) 29. [78] S.M. Watt, F. Gullo, M. Van Der Garde, D. Markeson, R. Camicia, C.P. Khoo, J.
[41] F. Pati, J. Jang, D.H. Ha, S. Won Kim, J.W. Rhie, J.H. Shim, D.H. Kim, D.W. Cho, J. Zwaginga, Br. Med. Bull. 108 (2013) 25.
Nat. Commun. 5 (2014), https://doi.org/10.1038/ncomms4935. [79] J. Idaszek, M. Costantini, T.A. Karlsen, J. Jaroszewicz, C. Colosi, S. Testa,
[42] M.K. Kim, W. Jeong, S.M. Lee, J.B. Kim, S. Jin, H.W. Kang, Biofabrication 12 E. Fornetti, S. Bernardini, M. Seta, K. Kasarełło, et al., Biofabrication 11 (2019),
(2020), https://doi.org/10.1088/1758-5090/ab5d80. https://doi.org/10.1088/1758-5090/ab2622.
[43] V. Beachley, G. Ma, C. Papadimitriou, M. Gibson, M. Corvelli, J. Elisseeff, [80] T.A. Karlsen, R.B. Jakobsen, T.S. Mikkelsen, J.E. Brinchmann, Stem Cell. Dev. 23
J. Biomed. Mater. Res. 106 (2018) 147. (2014) 290.
[44] R.A. Pouliot, P.A. Link, N.S. Mikhaiel, M.B. Schneck, M.S. Valentine, F.J. Kamga [81] S.R. Herlofsen, A.M. Küchler, J.E. Melvik, J.E. Brinchmann, Tissue Eng. 17 (2011)
Gninzeko, J.A. Herbert, M. Sakagami, R.L. Heise, J. Biomed. Mater. Res. 104 1003.
(2016) 1922. [82] A. Shahdadfar, K. Frønsdal, T. Haug, F.P. Reinholt, J.E. Brinchmann, Stem Cell. 23
[45] R. Liang, G. Yang, K.E. Kim, A. D’Amore, A.N. Pickering, C. Zhang, S.L.Y. Woo, (2005) 1357.
J. Orthop. Transl. 3 (2015) 114. [83] X. Chen, A.S. Aledia, S.A. Popson, L. Him, C.C.W. Hughes, S.C. George, Tissue Eng.
[46] U. Blache, M. Ehrbar, Adv. Wound Care 7 (2018) 232. 16 (2010) 585.
[47] R. Gorodetsky, R.A.F. Clark, J. An, J. Gailit, L. Levdansky, A. Vexler, E. Berman, [84] K. Pill, J. Melke, S. Mühleder, M. Pultar, S. Rohringer, E. Priglinger, H.R. Redl,
G. Marx, J. Invest. Dermatol. 112 (1999) 866. S. Hofmann, W. Holnthoner, Front. Bioeng. Biotechnol. 6 (2018), https://doi.org/
[48] A. Sahni, C.W. Francis, Blood 96 (2000) 3772. 10.3389/fbioe.2018.00156.
[49] E. Chung, J.A. Rytlewski, A.G. Merchant, K.S. Dhada, E.W. Lewis, L.J. Suggs, Acta [85] G. Vargas, E.K. Chan, J.K. Barton, H.G. Rylander, A.J. Welch, Laser Surg. Med. 24
Biomater. 17 (2015) 78. (1999) 133.

12

You might also like