Vous êtes sur la page 1sur 29

Biofabrication

ACCEPTED MANUSCRIPT

Scaffold-free, label-free and nozzle-free biofabrication technology using


magnetic levitational assembly
To cite this article before publication: Vladislav A Parfenov et al 2018 Biofabrication in press https://doi.org/10.1088/1758-5090/aac900

Manuscript version: Accepted Manuscript


Accepted Manuscript is “the version of the article accepted for publication including all changes made as a result of the peer review process,
and which may also include the addition to the article by IOP Publishing of a header, an article ID, a cover sheet and/or an ‘Accepted
Manuscript’ watermark, but excluding any other editing, typesetting or other changes made by IOP Publishing and/or its licensors”

This Accepted Manuscript is © 2018 IOP Publishing Ltd.

During the embargo period (the 12 month period from the publication of the Version of Record of this article), the Accepted Manuscript is fully
protected by copyright and cannot be reused or reposted elsewhere.
As the Version of Record of this article is going to be / has been published on a subscription basis, this Accepted Manuscript is available for reuse
under a CC BY-NC-ND 3.0 licence after the 12 month embargo period.

After the embargo period, everyone is permitted to use copy and redistribute this article for non-commercial purposes only, provided that they
adhere to all the terms of the licence https://creativecommons.org/licences/by-nc-nd/3.0

Although reasonable endeavours have been taken to obtain all necessary permissions from third parties to include their copyrighted content
within this article, their full citation and copyright line may not be present in this Accepted Manuscript version. Before using any content from this
article, please refer to the Version of Record on IOPscience once published for full citation and copyright details, as permissions will likely be
required. All third party content is fully copyright protected, unless specifically stated otherwise in the figure caption in the Version of Record.

View the article online for updates and enhancements.

This content was downloaded from IP address 131.211.208.19 on 01/06/2018 at 22:57


Page 1 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2
3
1 Scaffold-free, label-free and nozzle-free biofabrication
4
5 2 technology using magnetic levitational assembly
6
7
8
3

pt
9
10 4 Vladislav A. Parfenov1*, Elizaveta V. Koudan1*, Elena A. Bulanova1, Pavel A. Karalkin1,
11
12 5 Frederico DAS Pereira1, Nikita E. Norkin1, Alisa D. Knyazeva1, Anna A. Gryadunova1, Oleg F.
13
6 Petrov2, Mikhail M. Vasiliev2, Maxim I. Myasnikov2, Valery P. Chernikov3, Vladimir A.

cri
14
15 7 Kasyanov4, Artem Yu. Marchenkov5, Kenn Brakke6, Yusef D. Khesuani1, Utkan Demirci7,
16
17 8 Vladimir A. Mironov1
18
19 9
20
10

us
21
22 11 1
Laboratory for Biotechnological Research “3D Bioprinting Solutions”, Moscow, Russia;
23 2
24 12 Joint Institute for High Temperatures, Russian Academy of Sciences, Moscow, Russia;
25 13 3
Institute of Human Morphology, Russian Academy of Science, Moscow, Russia;
26
27
28
29
14
15
4

5
Riga Stradins University, Riga, Latvia;
an
National Research University “Moscow Power Engineering Institute”, Moscow, Russia;
30 16 6
Susquehanna University, Selinsgrove, Pennsylvania 17870, USA
31 7
32 17 Stanford University, Palo Alto, USA
dM
33 18
34
35 19
36
37 20 Сorresponding authors: Vladislav A. Parfenov, Russian Federation, Moscow 115409, Kashirskoye shosse,
38 21 68-2, tel. +7 (499) 769-50-18, vapar@mail.ru; Vladimir A Mironov, Russian Federation, Moscow 115409,
39
40 22 Kashirskoye shosse, 68-2, tel. +7 (499) 769-50-18, vladimir.mironov54@gmail.com; Utkan Demirci,
41 23 Stanford University, 1-650-9069227, utkan@stanford.edu
42
43 24
pte

44
45
25 * Both authors contributed equally to this paper
46 26
47
48 27 Author contributions: V.A.P., U.D. and V.A.M. designed research; V.A.P., E.V.K., F.D.P., V.P.C., A.D.K.
49 28 and A.A.G. performed research; O.F.P., M.M.V. and M.I.M. performed MD simulation of dynamics of the
50
51 29
ce

polystyrene particles in the cusp magnetic trap; KB made surface evolver simulation, V.A.P. performed
52
30 computer simulation of magnetic field; V.A.P., E.V.K., P.A.K., A.Y.M., N.E.N. and V.A.K. analyzed data;
53
54 31 V.A.P., E.V.K., E.A.B., Y.D.K., U.D. and V.A.M. wrote the paper.
55
56 32
Ac

57
33
58
59 34
60
35
1
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 2 of 28

1
2 1
3
4 2 Abstract
5
6
3 Tissue spheroids have been proposed as building blocks in 3D biofabrication.
7 4 Conventional magnetic force-driven 2D patterning of tissue spheroids requires prior cell labeling
8

pt
9 5 by magnetic nanoparticles, meanwhile a label-free approach for 3D magnetic levitational
10
11 6 assembly has been introduced. Here we present first-time report on rapid assembly of 3D tissue
12
7 construct using scaffold-free, nozzle-free and label-free magnetic levitation of tissue spheroids.
13

cri
14 8 Chondrospheres of standard size, shape and capable to fusion have been biofabricated
15
16 9 from primary sheep chondrocytes using non-adhesive technology. Label-free magnetic levitation
17
18 10 was performed using a prototype device equipped with permanent magnets in presence of
19 11 gadolinium (Gd3+) in culture media, which enables magnetic levitation.
20

us
21 12 Mathematical modeling and computer simulations were used for prediction of magnetic
22
23 13 field and kinetics of tissue spheroids assembly into 3D tissue constructs. First, we used
24
25
14 polystyrene beads to simulate the assembly of tissue spheroids and to determine the optimal
26 15 settings for magnetic levitation in presence of Gd3+. Second, we proved the ability of
27
28
29
16
an
chondrospheres to assemble rapidly into 3D tissue construct in the permanent magnetic field in

30 17 the presence of Gd3+.


31 18 Thus, scaffold- and label-free magnetic levitation of tissue spheroids is a promising
32
dM
33 19 approach for rapid 3D biofabrication and attractive alternative to label-based magnetic force-
34
35 20 driven tissue engineering.
36
37
21
38 22 Keywords: tissue spheroids, magnetic levitation (MagLev), scaffold-free, label-free,
39
40 23 chondrospheres, gadolinium (Gd3+), biofabrication.
41
42 24
43
pte

25
44
45 26
46
47 27
48
49 28
50 29
51
ce

52 30
53
54 31
55
56
32
Ac

57 33
58
59 34
60
35
2
Page 3 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 Introduction
3
4 2 Tissue spheroids represent densely packed aggregates of living cells [1]. At first, they
5
6
3 were used to design the human diseases models in vitro and test lead candidates at preclinical
7 4 stages of drug development [2-5]. More recently, tissue spheroids were proposed as building
8

pt
9 5 blocks for bioprinting of functional human tissues and organs [6-8].
10
11 6 The research on tissue spheroids was focused on three main directions: 1) the
12
7 development of robotic scalable standardized methods for tissue spheroids biofabrication [9]; 2)
13

cri
14 8 systematic phenotyping of tissue spheroids [10] and 3) the elaboration of methods for assembling
15
16 9 tissue spheroids into 3D tissues and organ structures [11-13].
17
18 10 Several beneficial advantages for using tissue spheroids as building blocks exist. First,
19 11 tissue spheroids have a highest theoretically possible cell density comparable with natural tissue
20

us
21 12 [2]. Second, tissue spheroids have a compact rounded shape, which is ideally suitable for their
22
23 13 handling, manipulation, transfer, processing and bioprinting. Third, they have a complex internal
24
25
14 structure and multicellular composition. Moreover, tissue spheroids can have one or several
26 15 lumens and even can be pre-vascularized [14]. Finally, they have intrinsic properties for fusion.
27
28
29
16
an
Tissue fusion is an ubiquitous phenomenon during embryonic development [15] and it is a

30 17 fundamental principle of rapidly emerging bioprinting and biofabrication technologies based on


31 18 the use of tissue spheroids as building blocks. Closely placed and directly touching each other,
32
dM
33 19 tissue spheroids begin to fuse thus producing complex 3D tissue constructs [6].
34
35 20 Several approaches exist for tissue and organ biofabrication using tissue spheroids. Using
36
37
21 one method, cell aggregates were placed robotically or manually into 3D scaffolds made from
38 22 various biodegradable biomaterials [16-18]. Another approach involves the attachment and
39
40 23 spreading of tissue spheroids on electrospun matrices [19-21]. Furthermore, magnetic forces
41
42 24 have been used for 2D patterning of tissue spheroids, biofabricated from cells, labelled with
43
pte

25 magnetic nanoparticles [22-26]. All described biofabrication approaches are scaffold-based,


44
45 26 nozzle-based or label-based. These methods have certain advantages and inherent limitations.
46
47 27 For example, robotic 3D bioprinting allows to biofabricate complex 3D tissues and organs [13,
48
49 28 27, 28]. Further, the majority of current approaches involves the use of biomaterials or
50 29 nanomaterials as scaffolds. The main goal of our work was to demonstrate a path-breaking rapid
51
ce

52 30 approach for biofabrication of 3D engineered constructs from tissue spheroids employing


53
54 31 scaffold-free, nozzle-free and label-free magnetic levitational assembly.
55
56
32 Application of magnetic forces in tissue engineering started from series of pioneer studies
Ac

57 33 of Honda and Ito [29]. They coined this approach as a magnetic force-driven tissue engineering.
58
59 34 Initially, magnets and associated magnetic fields have been used for the rapid seeding of cells,
60
35 labelled by magnetic nanoparticles on the conventional scaffolds. For example, this approach has
3
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 4 of 28

1
2 1 been used for rapid seeding of labelled endothelial cells on vascular scaffold [30] and for
3
4 2 improvement of cell sheets detachment and transfer in cell sheet technology originally developed
5
6
3 by Teruo Okano’s group [31]. The next step involved magnetic forces for 2D patterning of tissue
7 4 spheroids, labelled by magnetic nanoparticles [22-26]. Recently, it was reported that
8

pt
9 5 superparamagnetic iron oxide nanoparticles in moderate concentrations are not toxic [32-34] and
10
11 6 can even be biodegraded and metabolized into physiological proteins such as ferritins [35].
12
7 First, the levitation of large scale non-biological objects was demonstrated [36, 37].
13

cri
14 8 MagLev labeled-free approach enabled living material hovering, sorting and fabrication [38]. In
15
16 9 these pioneering studies authors approved the use of magnetic levitation to guide the assembly of
17
18 10 millimeter to centimeter scale diamagnetic objects in a paramagnetic fluid medium, each
19 11 programmed by shape and density distribution, positioned in the magnetic field gradient,
20

us
21 12 generated by NdFeB magnets. In absence of direct contacts between components, their
22
23 13 equilibrium configuration was dependent on the counterbalance of acting magnetic and
24
25
14 gravitational forces. This technique enables positioning of components in 3D.
26 15 The label-free levitational diamagnetic assembly strategy could be used as a powerful
27
28
29
16
an
tool to fabricate soft small living blocks [38]. This strategy allowed for the first time the

30 17 alignment of living blocks in a paramagnetic suspending media for remote 3D assembly. During
31 18 reported magnetic levitation experiments Gd3+ has been used to paramagnetize suspending
32
dM
33 19 media. The paramagnetic Gd3+ formulations such as Omniscan have been already approved by
34
35 20 FDA for clinical use as a contrast agent for magnetic resonance imaging investigations in
36
37
21 humans. However, the high concentrations of Gd3+ could be potentially toxic for tissue spheroids
38 22 and a certain risk exists for osmotic pressure imbalance due to excessive use of ions in
39
40 23 paramagnetic medium. Theoretically, these limitations could be overcome by combination of
41
42 24 stronger magnetic fields and smaller density differences between building blocks and medium.
43
pte

25 All this enable self-assembly approaches based on magnetic forces that are particularly
44
45 26 significant since i) magnetic forces enable contactless manipulation in 3D; ii) the spatial
46
47 27 distribution of the magnetic forces can be designed to allow the variation of used arrays of
48
49 28 magnets and electromagnets; and iii) a globally applied magnetic field is capable for addressing
50 29 a large number of components in parallel, iv) self-assembly approaches enable scaffold-free
51
ce

52 30 biofabrication of 3D tissue constructs [35, 38].


53
54 31 Our preliminary experiments confirmed that relatively weak magnetic fields allow
55
56
32 levitational assembly of tissue spheroids only in the presence of toxic Gd3+ concentrations.
Ac

57 33 However, the employment of high gradient magnetic field enables MagLev assembly of tissue
58
59 34 spheroids at non-toxic Gd3+ concentrations in paramagnetic medium. Here, we report for the first
60
35 time a successful rapid assembly of 3D engineered construct using scaffold-free, label-free and
4
Page 5 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 nozzle-free magnetic levitation of tissue spheroids in non-toxic paramagnetic medium. These
3
4 2 data provide a proof of concept for new strategies in tissue engineering.
5
6
3
7 4 Materials and methods
8

pt
9 5 Reagents
10
11 6 Dulbecco's modified Eagle's medium (DMEM, cat.# 12491-015), fetal bovine serum
12
7 (FBS, cat.# 16000-044), antibiotic-antimycotic (cat.# 15240-062), trypsin/EDTA (cat.# 25200-
13

cri
14 8 114), phosphate-buffered saline (PBS, cat.# 18912-014) were obtained from Gibco (USA). L-
15
16 9 glutamine (cat.# F032) were obtained from Paneco (RF). Glutaraldehyde (cat.# G5882) was
17
18 10 obtained from Sigma-Aldrich (USA). CellTiter-Glo 3D kit (cat.# G9682) was purchased from
19 11 Promega (USA). Omniscan (gadodiamide) was purchased from GE Healthcare (Ireland).
20

us
21 12
22
23 13 Cell culture
24
25
14 The primary culture of articular sheep chondrocytes was kindly provided by Dr. N.P.
26 15 Omelianenko (N.N. Priorov’s Central Research Institute of Traumatology and Orthopedics,
27
28
29
16
an
Moscow, Russia). Cells were grown in DMEM medium containing 10% FBS, supplemented

30 17 with antibiotic/antimycotic and 2mM L-glutamine. The cells were incubated at 37ºC in a
31 18 humidified atmosphere with 5% CO2 and routinely split at 85-95% confluence. Cell transfer and
32
dM
33 19 preparation of single-cell suspensions were performed using mild enzymatic dissociation with a
34
35 20 0.25% trypsin/0.53mM EDTA solution. Cells were confirmed free of mycoplasma contamination
36
37
21 according DAPI (Invitrogen, cat. # D1306) staining protocol.
38 22
39
40 23 Formation of tissue spheroids using Corning spheroid microplates
41
42 24 Tissue spheroids were formed using ultra-low adhesion Corning spheroid microplates
43
pte

25 (Corning, cat.# 4520) according to the manufacturer protocol. Briefly, monolayer cells with 95%
44
45 26 confluence were rinsed by EDTA solution, harvested from the culture flasks by 0.25%
46
47 27 trypsin/0.53mM EDTA and then suspended in cell culture medium. The concentration of the
48
49 28 cells was 8x104 per milliliter. 100μl of cell suspensions were dispensed to the wells of Corning
50 29 spheroid microplates. Corning spheroid microplates were incubated at 37ºC in a humidified
51
ce

52 30 atmosphere with 5% CO2 for 3 days.


53
54 31
55
56
32 Determination of spheroid diameter and roundness distribution
Ac

57 33 Tissue spheroids were biofabricated and captured at 3d day in culture using bright-field
58
59 34 imaging at inverted microscope Nikon Eclipse Ti-S, Japan. Spheroid diameters and roundness
60
35 were measured using Image J 1.48v software (NIH, Bethesda, MD, USA). Briefly, all original
5
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 6 of 28

1
2 1 grayscale images were converted to simplified threshold images under the same converting
3
4 2 condition and the edges of the spheroids were automatically detected. MinFeret’s diameters of
5
6
3 the detected spheroid edges were measured initially as pixels, and converted to micrometers by
7 4 comparing to a reference length. Roundness was measured using Image J 1.48v shape descriptor
8

pt
9 5 and calculated as 4*area/(π*major axis2) [39].
10
11 6
12
7 Estimation of tissue spheroids viability at different concentrations of gadolinium
13

cri
14 8 The viability of tissue spheroids was assessed using the CellTiter-Glo 3D kit according to
15
16 9 the manufacturer protocol. Briefly, 3-day-old tissue spheroids with a starting cell number of
17
18 10 8000 cells were placed in 0, 50 and 250mM Gd3+ concentrations for 24 hours. Then CellTiter-
19 11 Glo 3D kit was added and luminescence was recorded after 30 min incubation using VICTOR
20

us
21 12 X3 Multilabel Plate Reader (Perkin Elmer, USA).
22
23 13
24
25
14 Mechanical testing
26 15 The mechanical properties of tissue spheroids were measured by a micro-scale parallel-
27
28
29
16
an
plate compression testing system Microsquisher (CellScale, Canada) with associated

30 17 SquisherJoy software. Tissue spheroids with a starting cell number of 8000 cells were formed
31 18 using Corning spheroid microplates. They were cultured 3 days and then placed in 0, 50 and
32
dM
33 19 250mM Gd3+ concentrations for 24 hours prior to mechanical characterization. For mechanical
34
35 20 testing spheroids were placed in a PBS-filled bath at 37 ºC and compressed to 50% deformation
36
37
21 in 20 sec. The microbeams with diameters 152.4μm (recommended max force 57 mN) and 304.8
38 22 μm (recommended max force 917 mN) were employed depending on the stiffness and sensitivity
39
40 23 required to measure tissue spheroids from different cell types. The force-displacement data
41
42 24 obtained from the compression test were converted to stress-strain curves and the lower portion
43
pte

25 of the curve (0-20% strain) was used to obtain a linear regression line and estimate the Young’s
44
45 26 modulus. In each group eight samples of spheroids were measured.
46
47 27
48
49 28 Spheroid fusion assay
50 29 Spheroid fusion assay was performed using Corning spheroid microplates (Corning, cat.#
51
ce

52 30 4520). Pairs of 3-day-old tissue spheroids with a starting cell number of 8000 cells were placed
53
54 31 in contact in 0, 50 and 250mM Gd3+ concentrations and incubated for 7 days. Bright-field images
55
56
32 of spheroid doublets were obtained at points 0 h, 4 h, 6 h, 1 day, 2 days, 3 days, 4 days and 7
Ac

57 33 days using Nikon Eclipse Ti-S microscope. Contact length, intersphere angle and doublet length
58
59 34 were measured using Image J 1.48v software (NIH, Bethesda, MD, USA) and plotted as a
60
35 function of time using GraphPad Prism software (GraphPad Software, Inc., La Jolla, CA).
6
Page 7 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 Doublet length (Fig. 5) was normalized to initial doublet length. Measurements for each
3
4 2 parameter were reported as mean ± S.E.M.
5
6
3
7 4 Electrospinning of polyurethane
8

pt
9 5 Polyurethane was kindly provided by Dr. Xuejun Wen (EG-85A, Lubrizol, USA).
10
11 6 Electrospinning of microfibrous polyurethane matrix was performed using commercial
12
7 Professional Electrospinning Lab Device (Yflow, Spain). Polyurethane was dissolved to 17%
13

cri
14 8 concentration in solvents containing 40% N,N-dimethylformamide and 60% tetrahydrofuran.
15
16 9
17
18 10 Scanning Electron Microscopy (SEM)
19 11 Tissue spheroids and constructs made from chondrospheres were placed on the surface of
20

us
21 12 electrospun polyurethane matrix and allowed to attach during 6 hours. Subsequently samples
22
23 13 were fixed with 2.5% glutaraldehyde/PBS, dehydrated through ethanol series and then were
24
25
14 dried in a critical point dryer (HCP-2, Hitachi Koki Co. Ltd., Japan). The samples were
26 15 transferred on a stub of metal with adhesive surface, coated with gold using ion coater (IB-3,
27
28
29
16
an
EIKO, Japan) and then observed using the microscope JSM -6510 LV (JEOL, Japan).

30 17
31 18 Tissue spheroids morphology
32
dM
33 19 Tissue spheroids were fixed with 2.5% glutaraldehyde/PBS and then treated with 1%
34
35 20 osmium tetraoxide/PBS. The samples were dehydrated by soaking in a series of solutions of
36
37
21 increasing concentrations of ethanol, stained with 1% uranyl acetate/70% ethanol and finally
38 22 embedded to araldite-epon mixture. Semi-thin sections were prepared on a LKB-III ultratome
39
40 23 (Sweden), stained with 1% toluidine blue and then examined using a light microscope (Leica
41
42 24 DM 2500, Germany) equipped with a digital camera (Leica DFC 290, Germany).
43
pte

25
44
45 26 Tissue construct histology
46
47 27 The constructs, assembled from chondrospheres in magnetic field, were fixed in 10%
48
49 28 buffered formalin (pH 7.4) for 24 hours and then embedded in paraffin with a melting point of
50 29 +54°C (Biovitrum, Russia). Serial sections with a thickness of 5μm were made with microtome
51
ce

52 30 Microm HMS 740 (ThermoFisher Scientific, USA), routinely stained with hematoxylin and
53
54 31 eosin (Biovitrum, Russia) and then covered with Bio-Mount medium (Bio Optica Milano S.P.A.,
55
56
32 Italy) before histological examination.
Ac

57 33
58
59 34 Estimation of Spheroid Cell Density
60

7
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 8 of 28

1
2 1 Spheroid cell density was estimated by analysis of semi-thin sections of tissue spheroids
3
4 2 using Image J 1.48v software (NIH, Bethesda, MD, USA).
5
6
3
7 4 Determination of polystyrene bead density
8

pt
9 5 The density of polystyrene beads (Polyscience, Inc. USA, cat.# 64235) was determine by
10
11 6 means of equating the force of gravity with buoyancy force. We put polystyrene beads with
12
7 diameter 170μm in the container with deionized water, and then the dextrose powder was added
13

cri
14 8 to the container followed by the thorough mixing. Gradual adjusting of the dextrose
15
16 9 concentration in the solution provided floating of the beads in the water volume without sinking
17
18 10 or emerging at the surface. Thus, we obtained the polystyrene beads density equal to 1.0405
19 11 g/cm3 .
20

us
21 12
22
23 13 Surface evolver simulation
24
25
14 The fusion behaviour of tissue spheroids was illustrated using the open source software
26 15 Surface Evolver, developed and described in details earlier [40]. Tissue spheroids were
27
28
29
16
an
approximated and modelled as ball-like liquid droplets of standardized size and volume. The

30 17 initial positions of closely contacting tissue spheres were random, then evolved by gradient
31
18 descent to minimize distance between centers subject to centers being at least a diameter apart.
32
dM
33 19 The progressive process of fusion of tissue spheres was modeled by iterating gradient descent to
34
35 20 minimize the surface energy subject to the constraint of constant spheroid volumes, until
36
37 21 movement stopped. The evolution script created interfaces between spheroids where spheroids
38 22 touch. The configuration reached is a local minimum of energy, but not necessarily a global
39
40 23 minimum. The progressive changes in the shape of single spheroids contacted with each other
41
42 24 inside forming compacted tissue constructs have been visualized. The simulations have been
43
pte

44
25 performed for 40 tissue spheroids.
45 26
46
47 27 Magnetic experimental setup
48
49 28 To perform the experiment we designed custom laboratory installation with construction
50
29 shown on Fig.1 (a, d). The installation consists of 2 CMOS digital cameras (DMK41AU02, The
51
ce

52 30 Imaging Source Europe GmbH, Germany), light sources, lens system (Optem ZOOM 70XL,
53
54 31 Qioptiq, Germany), which are mounted on 3-axis positioning system assembled from 3 linear
55
56 32 stages (TSX-1D, Newport Corporation, USA), magnet holding system and 2 ring-shape NdFeB
Ac

57 33 magnets with cutouts for video capture (Fig. S1). The external diameter of magnets is 85 mm;
58
59 34 the internal diameter is 20 mm; thickness (height) is 24 mm. Magnets are assembled in such a
60
35 way that they oriented to each other with the same poles. This experimental setup was placed in
8
Page 9 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 the incubator to ensure the appropriate temperature regime (37°C) during the process of the
3
4 2 constructs assembly.
5
6
3 Non-homogeneous magnetic field with orifice in the center of the magnets was created in
7 4 the axial hole of the magnetic installation (working zone). The distribution of the magnetic
8

pt
9 5 induction values in the working area vertical section is shown in the 3D model graph (Fig. 1c). A
10
11 6 transparent glass cuvette with dimensions of 12 x 12 x 50 mm we inserted into the hole of the
12
7 magnet system (Fig. S1). The cuvettes were filled with paramagnetic liquid contained
13

cri
14 8 diamagnetic particles: polystyrene beads or tissue spheroids. The paramagnetic liquid consisted
15
16 9 of the DMEM medium and range of Gd3+ concentration: 0, 50, and 250mM. The process of
17
18 10 levitational assembly for polystyrene beads and tissue spheroids was recorded by two video
19 11 cameras with 1x and 3x optical magnification. After tissue spheroids assembling, resulted
20

us
21 12 constructs continued to levitate in a magnetic field for 24 hours to full complete the fusion
22
23 13 process.
24
25
14 The principle of operation of experimental installation implicates the creation of a local
26 15 microgravity zone where the effects of all acting on diamagnetic objects forces are compensated.
27
28
29
16
an
In our setup, the directed upward Archimedes force, the directed downward force of gravity, and

30 17 the directed toward the local minimum of the magnetic field magnetic force simultaneously act
31 18 on the object under static conditions. The magnetic forces moment appears only if the magnetic
32
dM
33 19 field is non-homogeneous. Then the effective magnetic force [41], acting on the object in non-
34
35 20 homogeneous magnetic field will be equal to the volume integral:
36 𝜒𝜒𝜒𝜒
37 21 F= ∇(𝑩𝑩2 ),
2
38
39 22 where for paramagnets 𝜒𝜒 > 0, while for diamagnets 𝜒𝜒 < 0. The sign determines the
40
41
23 direction of the magnetic force action.
42 24 As a result, diamagnetic objects will be pushed out into an area with lower field strength
43
pte

44 25 (magnetic trap) under the action of a magnetic force. In Earth’s gravity condition the
45
46 26 equilibration of objects occurs at a certain distance from the local minimum of the magnetic
47 27 field.
48
49 28
50
51 29 Computer simulation of magnetic field
ce

52
53 30 The simulation of three-dimensional non-homogeneous static magnetic field in a
54 31 paramagnetic medium from two permanent magnets we performed using "ANSYS
55
56 32 Electromagnetics Suite" software for Maxwell3D. The characteristics of magnetic field used in
Ac

57
58 33 the simulation were as follows: relative permeability paramagnetic medium 1.00004135; and
59
34 material grade of NdFeB magnet N38 (VG=1.21 TL).
60

9
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 10 of 28

1
2 1
3
4 2 Molecular dynamics (MD) simulation of dynamics of the polystyrene particles in the
5
6
3 cusp magnetic trap
7 4 The MD modeling was carried out as described previously [42-44]. We assumed that all
8

pt
9 5 particles in MD calculations are spherical, having the same size and mass mp. For modeling
10
11 6 dynamics of the diamagnetic polystyrene particles in the cusp magnetic trap, we have
12
7 numerically solved the system of the Newton equations for all the particles (1 < k < N)
13

cri
14 d 2rk r
15 8 mp 2
= ∑ F ( rkl ) kl + FkB + f k + f g + fb , (1)
16 dt l rkl
17
18 9 where rk is the position of the center of a particle k, rkl= rk − rl . The first term in the right-hand
19
20 10 part of Eq. (1) is the Lennard-Jones interaction with other particles, the second term is the

us
21
22 11 interaction with the magnetic field of the trap (Fig. 1c ), the third term is the force of viscous
23
12 friction against the continuum liquid which is determined by the Stokes formula f k = −3πdηu k , η
24
25
26
13 is the viscosity of Gd3+ suspension, d is the particle diameter and uk = drk / dt is the particle
an
27
28
14 velocity relative to the stationary liquid, the fourth and the fifth terms is a force of gravity f g
29
30 15 and buoyancy force f b acting on the particle. Numerical simulation was performed for N = 100.
31
32 16 The simulation was performed for polystyrene beads and tissue spheroids with densities
dM
33 17 ρ = 1.0405 g/cm3 and ρ = 1.05 g/cm3, respectively. At the first step, particles were distributed
34
35 18 randomly within the computational domain with initial zero velocity. The computational domain
36
37 19 corresponds to the internal volume of the experimental cell - 12×12×47 mm.
38
39 20
40
21 Data analysis
41
42 22 Statistical data was analyzed using GraphPad Prism software (GraphPad Software, Inc.,
43
pte

44 23 La Jolla, CA) and represented as mean ± S.E.M. The Analysis of Variance (ANOVA) test was
45
46 24 used to find the significant differences between the means of the three groups with P< 0.0001.
47 25
48
49 26 Results
50
51 27 Magnetic experimental setup
ce

52
53
28 We created a magnetic installation (Fig. 1d), which generates a magnetic field in the
54 29 working volume with a very high (calculated value is up to 2.2 T/cm) gradient along the vertical
55
56 30 axis. The distribution of magnetic induction, created by the system of magnets, is shown in Fig.
Ac

57
58 31 1 (b, c, e). We used molecular dynamics modeling to estimate the time, shape and height of
59
32 constructs assembly in a magnetic field.
60

10
Page 11 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 The fusion dynamics modeling resulted in the graph (Fig. 1f), which shows the relation
3
4 2 between the projection on the vertical axis of the resultant force, acting on particles, tissue
5
6
3 spheroids or polystyrene beads, during the experiment, and the vertical coordinate in the
7 4 laboratory installation system of axes.
8

pt
9 5 We used polystyrene beads to test the operations and debugs for all modes. They proved to
10
11 6 be sufficiently good physical analogues of tissue spheroids for such parameters as magnetic
12
7 susceptibility and density. The use of tissue spheroids for routine testing of assembly modes was
13

cri
14 8 time and cost consuming, so we used polystyrene beads as their substitutes. Indeed, beads and
15
16 9 tissue spheroids have the similar parameters of density and magnetic susceptibility. After initial
17
18 10 determination of the optimal magnetic field configuration, we continued our experiments with
19 11 tissue spheroids.
20

us
21 12 The construct’s center of mass corresponds to the point, where the resultant force turns into
22
23 13 zero, thus forming levitation point. According to the graph (Fig. 1f) this point has the following
24
25
14 coordinates in the vertical axis: 3.2 mm for polystyrene beads and 5.6 mm for tissue spheroids.
26 15 The vertical coordinate of construct’s center of mass was calculated by means of video analysis.
27
28
29
16
an
The coordinates, defined in such way were as follows: 3.9 mm for polystyrene beads, 5.1 mm for

30 17 tissue spheroids (Fig. 6). Results of our simulation are in a good agreement with experiments.
31 18
32
dM
33 19 Biofabrication of tissue spheroids
34
35 20 Tissue spheroids were formed from primary sheep chondrocytes using ultra-low adhesion
36
37
21 Corning spheroid microplates (Fig. 2a). Their morphology was estimated using bright-field
38 22 microscopy and SEM. We’ve used 3-day-old spheroids for construct biofabrication. Tissue
39
40 23 spheroids had well-defined borders and spherical shape at 3d day in culture (Fig. 2b). In order to
41
42 24 investigate morphology and surface characteristics of tissue spheroids we analyzed them by
43
pte

25 SEM (Fig. 2c). The surface of spheroids was formed by closely packed cells. Standard size and
44
45 26 shape of tissue spheroids is an essential prerequisite for their use in bioprinting as building
46
47 27 blocks. Spheroid diameters (Fig. 2d) and roundness (Fig. 2e) were measured after 3 days of
48
49 28 culture. The average spheroid diameter was 346±16.58 µm. The average spheroid roundness was
50 29 0.907±0.047. The standard deviations were <10% of the mean value for both diameter and
51
ce

52 30 roundness. These results revealed that tissue spheroids had uniform standardized geometry
53
54 31 and could be used for biofabrication of three-dimensional tissues.
55
56
32
Ac

57 33 Histological analysis and viability of tissue spheroid


58
59 34 A histological analysis on semi-thin sections of 3-day-old tissue spheroids was performed
60
35 in order to estimate possible toxic effects and to determine an optimal nontoxic Gd3+
11
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 12 of 28

1
2 1 concentration (Fig. 3a). In non-treated conditions, chondrospheres demonstrated high cell
3
4 2 density, swirl-like cell arrangement and intensive staining of cytoplasm. In 50mM Gd3+ solution
5
6
3 cell density did not change, but cytoplasm staining was slightly reduced. Finally, at toxic 250mM
7 4 Gd3+ concentration, cell density dramatically reduced, that led to manifestations of cell death and
8

pt
9 5 apoptosis in form of pycnotic nuclei and accumulation of extracellular debris.
10
11 6 The viability of cells within tissue spheroids was analyzed by measuring the luminescent
12
7 signal generated by luciferin-luciferase interconnection as a function of cytoplasmic ATP
13

cri
14 8 concentration. Fig. 3b shows that tissue spheroids in 50mM Gd3+ solution remained viable after
15
16 9 24 hours of incubation, while 250mM Gd3+ concentration revealed significant toxic effect. High
17
18 10 statistically significant difference, p<0.0001, in cell density corresponded to 85% in 50mM Gd3+
19 11 solution compared to 100% in 0mM Gd3+ solution, while the cell density at 250mM Gd3+
20

us
21 12 concentration was 40% (Fig. 3c). Thus, cell counting showed that the increase of Gd3+
22
23 13 concentration leads to the reduction in the cell density.
24
25
14
26 15 The Mechanical Properties of Tissue Spheroids
27
28
29
16
an
The influence of gadolinium Gd3+ on mechanical properties of tissue spheroids was

30 17 estimated by tensiometry using parallel plates modification. As shown in Figure 4, the


31 18 mechanical properties of spheroids strongly depended on concentration of gadolinium Gd3+. The
32
dM
33 19 values of spheroids elastic modulus in 0 and 50mM Gd3+ solutions were 2.91±0.15 kPa and
34
35 20 2.95±0.1 kPa, correspondingly. The increase of Gd3+ concentration to 250mM resulted in
36
37
21 decrease of elastic modulus value to 0.14±0.05 kPa. This observation can be explained by low
38 22 viability of tissue spheroids in 250mM Gd3+ solution.
39
40 23 Fig. 4a shows the stress-strain diagram obtained from the compression load-displacement
41
42 24 curve. The sharp rise of the curve in the area of high strain values is associated with an increase
43
pte

25 of the tissue spheroid cross section. Fig. 4c shows the stages of chondrosphere compression
44
45 26 process between two parallel plates. When compressing up to 20% of the original diameter, an
46
47 27 increase of the cross section does not occur. The stress-strain curve changes virtually linearly
48
49 28 before this strain; in this section, we calculated the Young's modulus. According to our results,
50 29 the nontoxic concentration 50mM Gd3+ in the growth medium does not affect the tissue
51
ce

52 30 spheroids Young's modulus at all, which apparently means no significant changes in the internal
53
54 31 structure of the tissue spheroid (Fig. 4b). Finally, at a toxic concentration 250mM Gd3+, the
55
56
32 Young's modulus sharply decreases by ~ 93%, indicating that at a given concentration of
Ac

57 33 gadolinium salts irreversible changes occurred within the cells, that clearly affects the
58
59 34 mechanical properties of the tissue spheroid.
60
35
12
Page 13 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 The Fusion of Tissue Spheroids
3
4 2 To investigate tissue spheroids fusion dynamics at different Gd3+concentrations,
5
6
3 spheroids in pairs were placed in contact with each other in the same well of low adhesion
7 4 microplate and allowed to fuse for 7 days. The kinetics of tissue spheroid fusion was monitored
8

pt
9 5 and analyzed daily in bright-field under inverted microscope. Contact length, intersphere angle
10
11 6 and doublet length were measured according to the method described by Susienka et al. [45]
12
7 (Fig. 5a). As shown in Figure 5b, for spheroid pairs in 0 and 50mM Gd3+ solutions contact
13

cri
14 8 lengths and intersphere angles increased as a function of time and doublet lengths shortened
15
16 9 gradually from first day in culture. Spheroid doublet in 0mM Gd3+ solution shortened faster than
17
18 10 spheroid doublet in 50mM Gd3+ solution. At the seventh day of incubation the intersphere angle
19 11 increased up to 179º, indicating complete spheroid fusion. Spheroid pair in 250mM Gd3+
20

us
21 12 solution continued to fuse during first 24 hours and then no further progressive aggregation was
22
23 13 observed. This observation confirmed also the high cytotoxicity of 250mM Gd3+ concentration.
24
25
14 To check the possibility of microtissue construct biofabrication at different
26 15 concentrations of Gd3+, we placed 30 chondrospheres in the same well of non-adhesive
27
28
29
16
an
microplate and allowed fusion for 48 hours (Fig. 5c). At 0 and 50mM Gd3+ concentrations the

30 17 formation of compact constructs was observed after 2 days of incubation, while no construct was
31 18 formed in 250mM Gd3+ solution.
32
dM
33 19
34
35 20 Assembly of constructs in a magnetic field
36
37
21 We performed construct assembly in magnetic field using two types of building blocks.
38 22 The time length for tissue spheroids assembly was shorter by 20% ± 1.5% compare to one for
39
40 23 polystyrene beads assembly due to the difference in the magnetic susceptibility of diamagnetic
41
42 24 materials. Meanwhile, the height of the assembled constructs was lower by 1.2mm ± 0.2 mm
43
pte

25 because the spheroids density is higher than the polystyrene beads density, and, as a result, the
44
45 26 gravity force acts more strongly on them. It means that the force compensation will occur in the
46
47 27 area where the magnetic force is higher, that is closer to the magnet surface. The density of the
48
49 28 chamber medium was assumed ρ = 1.015 g/cm3. Considering the near-spherical configuration of
50
51 29 the magnetic field in the trap we can conclude that the final form of the cluster obtained in MD
ce

52 30 simulation should be an oblate body with relation between the semi axes a y / ar = 1 .
53
54
31 In MD simulations for the formation of cluster from polystyrene particles with diameter d
55
56 32 = 170µm and tissue spheroids of diameter d = 350µm, we set χ = −0.65⋅10−6 cm3/g and
Ac

57
58 33 −0.72⋅10−6 cm3/g, respectively. For sufficiently large N the shape of the cluster became almost
59
60 34 independent of N, and the ratio of the thickness and diameter of the cluster H c / Dc was close to

13
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 14 of 28

1
2 1 the theoretical value of the relation between the semi axes a y / ar = 1 . Experimental values of the
3
4 2 ratio H c / Dc for polystyrene and tissue spheroids were almost equal and corresponded to the
5
6 3 theoretical values.
7
8 4 Configurations of the polystyrene beads and tissue spheroids clusters obtained

pt
9
10 5 experimentally and in MD simulation are shown in Fig. 6a and presented in video S1 and S2.
11 6 The shape and size of the clusters were similar. Inside the formation area, the force of gravity,
12
13 7 acted on the particles, was counterbalanced by the magnetic force of the trap and the buoyancy

cri
14
15 8 force. The center of the mass for the polystyrene particles structure formed by MD simulation
16
17
9 located by a distance 3.2mm downward from the center of the trap. For tissue spheroids, this
18 10 displacement was up to 5.6mm.
19
20 11 SEM analysis proved the fusion of tissue spheroids into the compact 3D tissue construct

us
21
22 12 (Fig. 6e). Despite the presence of some grooves and pits, corresponding to separate
23
13 chondrospheres, the ongoing tissue fusion process was obvious, revealed by gradual smoothing
24
25 14 of the construct surface Mathematical modeling and computer simulation using Surface Evolver
26
27
28
29
15
16
an
software demonstrated that the formation of grooves is unavoidable in case of random packing
(Fig. 7a,b,c). Thus, the formation of holes and empty space could be explained by either the
30 17 incomplete fusion process or by a chondrospheres rigidity. Our previous experiments involved
31
32 18 softer tissue spheroids, namely, embryonic mouse thyroid gland explants, these showed complete
dM
33
34 19 fusion in rounded and compact tissue construct without any holes and empty space [13].
35
36
20 We performed histological analysis to visualize chondrospheres status and fusion in
37 21 biofabricated tissue construct (Fig. 7d). It was demonstrated, that viable chondrocytes were
38
39 22 tightly packed inside tissue spheroids. The spheroids fusion kinetics within biofabricated 3D
40
41 23 tissue construct was different. As results, there were some areas of complete fusion and there
42 24 were some gaps or incomplete fusion in tissue which probably reflects random initial packing of
43
pte

44 25 chondrospheres. Similar void areas have been shown previously during cell aggregates fusion on
45
46 26 pre-stretched electrospun synthetic matrices [20] and also during fusion of chondrospheres
47
48
27 biofabricated from cells labeled with iron oxide magnetic nanoparticles [46, 47].
49 28
50
51
ce

29 Discussion
52
53 30 The main outcome of our study is a proof of concept for magnetic levitational assembly
54
31 of microtissue constructs applying viable tissue spheroids.
55
56 32 The horizons of magnetic levitational assembly have been demonstrated in several
Ac

57
58 33 pioneering publications [36-38]. In early studies, authors used predominantly non-biological
59
60 34 microobjects, such as plastic beads, cells on the beads and cells within hydrogel blocks. We

14
Page 15 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 reported for the first time the successful rapid biofabrication of millimeter scale microtissue
3
4 2 constructs from chondrospheres applying magnetic levitational assembly.
5
6
3 The Earth’s gravity is one of the most serious hindrances for MagLev assembly of
7 4 biological constructs. One way to overcome it is to use strong magnets with addition of
8

pt
9 5 paramagnetic substances, i.e. Gd3+ into working solution. However, the toxicity of paramagnet
10
11 6 concentration enabling the levitation of objects comprises the feasible limitation of approach. In
12
7 our study, we designed an optimal form of permanent magnets and estimated their mutual
13

cri
14 8 emplacement using the MD simulation that allowed us to perform levitational assembly with
15
16 9 nontoxic concentration of the paramagnet in the culture medium. The rate of construct assembly
17
18 10 and its geometrical shape were pre-calculated. Thus, a rather simple, but original magnetic
19 11 system was created, which provided generation of a magnetic field with a gradient of up to 2.2
20

us
21 12 T/cm with a local minimum at the center of the working area. In the course of an experiment, we
22
23 13 obtained the data on the construct assembly kinetics and its spatial characteristics, which
24
25
14 confirmed the validity of the calculated magnetic field distribution in the magnetic system
26 15 working area.
27
28
29
16
an
Our data demonstrated that the use of permanent magnets with defined shape and relative

30 17 position significantly accelerates the assembly of both polystyrene beads and tissue spheroids.
31 18 Quantitative estimation of cytoplasmic ATP production to prove cell’s viability, histological
32
dM
33 19 analysis and confirmed tissue spheroids capacity for fusion have justified the vitality of
34
35 20 chondrospheres, exposed to 50mM of Gd3+ in course of magnetic levitation.
36
37
21 For biofabrication of tissue-engineered constructs of much larger sizes it is necessary
38 22 either to use superconducting magnets, providing the required magnetic field gradient throughout
39
40 23 the working volume, or apply a higher concentration of the paramagnetic solution. However, per
41
42 24 our data, the high concentration of the paramagnetic induces the undesirable cytotoxic effects.
43
pte

25 Achievement of sufficient magnetic field gradient in a larger working volume is still unsolved
44
45 26 and challenging bioengineering task.
46
47 27 Alternative approach involves the use of space microgravity which will allow
48
49 28 significantly reduce the concentration of Gd3+. Our preliminary calculations, mathematical
50 29 modeling and computer simulations strongly suggest that application of strong magnetic field in
51
ce

52 30 space microgravity conditions will allow to reduce significantly the concentration of Gd3+ and to
53
54 31 increase the size of biofabricated tissue constructs. After rapid levitational assembly of spheroids
55
56
32 in space, Gd3+ could be simply washed out and replaced by conventional cell culture media for
Ac

57 33 post-processing, thus providing biofabricated organoids suitable for prolonged space missions.
58
59 34 In our study, we have chosen to use chondrospheres, fabricated from articular
60
35 chondrocytes. Previously it has been shown that closely placed chondrospheres both loaded and
15
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 16 of 28

1
2 1 unloaded with magnetic nanoparticles can fuse into large size cartilage constructs using scaffold-
3
4 2 free approach [26, 48]. Moreover, the treatment of cartilage defects by chondrospheres, capable
5
6
3 to attachment and spreading on injured surface of cartilage had already been demonstrated in
7 4 several independent preclinical and clinical studies [49-51]. However, despite the many positive
8

pt
9 5 examples for medical application of magnetic, especially, iron oxide based, nanoparticles, their
10
11 6 possible cytotoxicity in vivo is still a challenge [52, 53]. Our data demonstrates that magnetic
12
7 levitation provides simple and rapid way for scaffold-free and label-free tissue construct
13

cri
14 8 bioassembly. Recently, the systematical investigation of the solid scaffold-free and label-free
15
16 9 acoustic levitation has been initiated by several groups as an additional potential alternative
17
18 10 technology for conventional solid scaffold-based or label-based 3D patterning [54, 55]. In this
19 11 context, one could assume that combination of magnetic and acoustic levitation will potentially
20

us
21 12 enable the rapid assembly of tissue and organ constructs with increased complexity. Specially
22
23 13 designed system of repulsive acoustic holes with multiple spatially arranged acoustic field
24
25
14 generators can ensure successful implementation of this intriguing approach. Similarly, the
26 15 employment of tissue spheroids with more advanced intrinsic properties and compositions such
27
28
29
16
an
as heterocellular aggregates, lumenized vascular tissue spheroids, pre-vascularised organo-

30 17 specific aggregates and sacrificial vascular tissue spheroids will ensure highly desirable
31 18 vascularization state and dramatically enhance the level of complexity of biofabricated tissue and
32
dM
33 19 organ constructs [56-59].
34
35 20 Theoretically, scaffold-free approach implies the absence of any temporal support such as
36
37
21 biodegradable biomaterials or scaffolds. Taking this, it looks like that we indeed employed
38 22 scaffold-free or at least biomaterials-free approach. However, magnetic field as well as acoustic
39
40 23 or electric fields provides temporal and removable support and could be considered as a new
41
42 24 form of scaffold. In order to escape possible confusion, we propose to call this new type of
43
pte

25 temporal support-based employment of physical fields as a “scaffield”.


44
45 26 Thus, presented scaffold-free, label-free and nozzle-free magnetic levitational assembly
46
47 27 in possible combination with acoustic hoist of tissue spheroids and other forms of physical force-
48
49 28 based 3D patterning of cell aggregates can potentially be a novel and more efficient approach for
50 29 rapid manufacturing of complex tissue and organ constructs. Design, development and
51
ce

52 30 implementation of new types of tissue and organ bioassemblers, based on the proposed concept
53
54 31 of “scaffield” could dramatically advance the emerging field of organ and tissue biofabrication.
55
56
32
Ac

57 33 Conclusion
58
59 34 Scaffold-free, label-free and nozzle-free biofabrication technology applying magnetic
60
35 levitational assembly has been demonstrated. 3D living tissue constructs have been biofabricated
16
Page 17 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 from chondrospheres in paramagnetic medium using non-toxic Gd3+concentration. The
3
4 2 introduced technology can became a powerful biofabrication tool that enables rapid assembly of
5
6
3 various tissues and organs.
7 4
8

pt
9 5 Acknowledgements
10
11 6 Authors thank Prof. Xuejun Wen (Virginia Commonwealth University, USA) for
12
7 providing polyurethane for electrospinning.
13

cri
14 8
15
16 9 References:
17
18 10 1. Lin RZ, Chang HY. Recent advances in three-dimensional multicellular spheroid culture
19 11 for biomedical research. Biotechnol J. 2008;3(9–10):1172–84.
20

us
21 12 2. Achilli T-M, Meyer J, Morgan JR. Advances in the formation, use and understanding of
22
23 13 multi-cellular spheroids. Expert Opin Biol Ther. 2012;12(10):1347–60.
24
25
14 3. Fennema E, Rivron N, Rouwkema J, van Blitterswijk C, De Boer J. Spheroid culture as a
26 15 tool for creating 3D complex tissues. Trends Biotechnol. 2013;31(2):108–15.
27
28
29
16 4.
an
Chatzinikolaidou M. Cell spheroids: the new frontiers in in vitro models for cancer drug

30 17 validation. Drug Discov Today. 2016;21(9):1553–60.


31 18 5. Cui X, Hartanto Y, Zhang H. Advances in multicellular spheroids formation. J R Soc
32
dM
33 19 Interface. 2017;14(127):20160877.
34
35 20 6. Mironov V, Visconti RP, Kasyanov V, Forgacs G, Drake CJ, Markwald RR. Organ
36
37
21 printing: Tissue spheroids as building blocks. Biomaterials. 2009;30(12):2164–74.
38 22 7. Kelm JM, Lorber V, Snedeker JG, Schmidt D, Broggini-Tenzer A, Weisstanner M, et al.
39
40 23 A novel concept for scaffold-free vessel tissue engineering: Self-assembly of microtissue
41
42 24 building blocks. J Biotechnol. 2010;148(1):46–55.
43
pte

25 8. Mattix B, Olsen TR, Gu Y, Casco M, Herbst A, Simionescu DT, et al. Biological


44
45 26 magnetic cellular spheroids as building blocks for tissue engineering. Acta Biomater.
46
47 27 2014;10(2):623–9.
48
49 28 9. Mehesz AN, Brown J, Hajdu Z, Beaver W, Silva JVL da, Visconti RP, et al. Scalable
50 29 robotic biofabrication of tissue spheroids. Biofabrication. 2011;3(2):25002.
51
ce

52 30 10. Koudan E V., Korneva J V., Karalkin PA, Gladkaya IS, Gryadunova AA, Mironov VA, et
53
54 31 al. The Scalable Standardized Biofabrication of Tissue Spheroids from Different Cell
55
56
32 Types Using Nonadhesive Technology. 3D Print Addit Manuf. 2017;4(1):53–60.
Ac

57 33 11. Hesuani YD, Pereira FDAS, Parfenov V, Koudan E, Mitryashkin A, Replyanski N, et al.
58
59 34 Design and Implementation of Novel Multifunctional 3D Bioprinter. 3d Print Addit
60
35 Manuf. 2016;3(1):65–8.
17
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 18 of 28

1
2 1 12. Gutzweiler L, Kartmann S, Troendle K, Benning L, Finkenzeller G, Zengerle R, Koltay P,
3
4 2 Stark GB, Zimmermann S. Large scale production and controlled deposition of single
5
6
3 HUVEC spheroids for bioprinting applications. Biofabrication. 2017; 9(2):025027.
7 4 13. Bulanova EA, Koudan E V, Degosserie J, Heymans C, Parfenov VA, Akhmedova SA, et
8

pt
9 5 al. Bioprinting of functional vascularized mouse thyroid gland. Biofabrication. 2017;9(3):
10
11 6 034105.
12
7 14. Kelm JM, Djonov V, Ittner LM, Fluri D, Born W, Hoerstrup SP, et al. Design of custom-
13

cri
14 8 shaped vascularized tissues using microtissue spheroids as minimal building units. Tissue
15
16 9 Eng. 2006;12(8):2151–60.
17
18 10 15. Pérez-Pomares JM, Foty RA. Tissue fusion and cell sorting in embryonic development
19 11 and disease: Biomedical implications. BioEssays. 2006;28(8):809–21.
20

us
21 12 16. Kim TG, Park SH, Chung HJ, Yang DY, Park TG. Hierarchically assembled
22
23 13 mesenchymal stem cell spheroids using biomimicking nanofilaments and microstructured
24
25
14 scaffolds for vascularized adipose tissue engineering. Adv Funct Mater.
26 15 2010;20(14):2303–9.
27
28
29
16 17.
an
Schon BS, Schrobback K, Van Der Ven M, Stroebel S, Hooper GJ, Woodfield TBF.

30 17 Validation of a high-throughput microtissue fabrication process for 3D assembly of tissue


31 18 engineered cartilage constructs. Cell Tissue Res. 2012;347(3):629–42.
32
dM
33 19 18. Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, et al. A bioprosthetic
34
35 20 ovary created using 3D printed microporous scaffolds restores ovarian function in
36
37
21 sterilized mice. Nat Commun. 2017;8(May):15261.
38 22 19. Chua KN, Lim WS, Zhang P, Lu H, Wen J, Ramakrishna S, et al. Stable immobilization
39
40 23 of rat hepatocyte spheroids on galactosylated nanofiber scaffold. Biomaterials.
41
42 24 2005;26(15):2537–47.
43
pte

25 20. Beachley V, Kasyanov V, Nagy-Mehesz A, Norris R, Ozolanta I, Kalejs M, et al. The


44
45 26 fusion of tissue spheroids attached to pre-stretched electrospun polyurethane scaffolds. J
46
47 27 Tissue Eng. 2014;5(X):2041731414556561.
48
49 28 21. Mironov V, Khesuani YD, Bulanova EA, Koudan E V, Parfenov VA, Knyazeva AD, et al.
50 29 Patterning of tissue spheroids biofabricated from human fibroblasts on the surface of
51
ce

52 30 electrospun polyurethane matrix using 3D bioprinter. Int J Bioprinting. 2016;2(1):45–52.


53
54 31 22. Lin R-Z, Chu W-C, Chiang C-C, Lai C-H, Chang H-Y. Magnetic reconstruction of three-
55
56
32 dimensional tissues from multicellular spheroids. Tissue Eng Part C Methods.
Ac

57 33 2008;14(3):197–205.
58
59 34 23. Ho VHB, Müller KH, Barcza A, Chen R, Slater NKH. Generation and manipulation of
60
35 magnetic multicellular spheroids. Biomaterials. 2010;31(11):3095–102.
18
Page 19 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 24. Bratt-Leal AM, Kepple KL, Carpenedo RL, Cooke MT, McDevitt TC. Magnetic
3
4 2 manipulation and spatial patterning of multi-cellular stem cell aggregates. Integr Biol.
5
6
3 2011;3(12):1224.
7 4 25. Whatley BR, Li X, Zhang N, Wen X. Magnetic-directed patterning of cell spheroids. J
8

pt
9 5 Biomed Mater Res - Part A. 2014;102(5):1537–47.
10
11 6 26. Luciani N, Du V, Gazeau F, Richert A, Letourneur D, Le Visage C, et al. Successful
12
7 chondrogenesis within scaffolds, using magnetic stem cell confinement and bioreactor
13

cri
14 8 maturation. Acta Biomater. 2016;37:101–10.
15
16 9 27. Derby B. Printing and prototyping of tissues and scaffolds. Science. 2012;338(6109):921–
17
18 10 6.
19 11 28. Murphy SV, Atala A. 3D Bioprinting of Tissues and Organs. Nat Biotechnol.
20

us
21 12 2014;32(8):773–85.
22
23 13 29. Ito A, Ino K, Hayashida M, Kobayashi T, Matsunuma H, Kagami H, et al. Novel
24
25
14 methodology for fabrication of tissue-engineered tubular constructs using magnetite
26 15 nanoparticles and magnetic force. Tissue Eng. 2005;11(9–10):1553–61.
27
28
29
16 30.
an
Mironov V, Kasyanov V, Markwald RR. Nanotechnology in vascular tissue engineering:

30 17 from nanoscaffolding towards rapid vessel biofabrication. Trends Biotechnol.


31 18 2008;26(6):338–44.
32
dM
33 19 31. Ito A, Hayashida M, Honda H, Hata K-I, Kagami H, Ueda M, et al. Construction and
34
35 20 harvest of multilayered keratinocyte sheets using magnetite nanoparticles and magnetic
36
37
21 force. Tissue Eng. 2004;10(5–6):873–80.
38 22 32. Chaudhary S, Smith CA, Del Pino P, de la Fuente JM, Mullin M, Hursthouse A, Stirling
39
40 23 D, Berry CC. Elucidating the function of penetratin and a static magnetic field in cellular
41
42 24 uptake of magnetic nanoparticles. Pharmaceuticals (Basel). 2013; 6(2):204-22.
43
pte

25 33. Lewis EE, Child HW, Hursthouse A, Stirling D, McCully M, Paterson D, Mullin M, Berry
44
45 26 CC. The influence of particle size and static magnetic fields on the uptake of magnetic
46
47 27 nanoparticles into three dimensional cell-seeded collagen gel cultures. J Biomed Mater
48
49 28 Res B Appl Biomater. 2015; 103(6):1294-301.
50 29 34. Jeong YG, Lee JS, Shim JK, Hur W. A scaffold-free surface culture of B16F10 murine
51
ce

52 30 melanoma cells based on magnetic levitation. Cytotechnology. 2016; 68(6):2323-2334.


53
54 31 35. Mazuel F, Espinosa A, Luciani N, Reffay M, Le Borgne R, Motte L, et al. Massive
55
56
32 Intracellular Biodegradation of Iron Oxide Nanoparticles Evidenced Magnetically at
Ac

57 33 Single-Endosome and Tissue Levels. ACS Nano. 2016;10(8):7627–38.


58
59 34 36. Mirica KA, Ilievski F, Ellerbee AK, Shevkoplyas SS, Whitesides GM. Using magnetic
60
35 levitation for three dimensional self-assembly. Adv Mater. 2011;23(36):4134–40.
19
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 20 of 28

1
2 1 37. Subramaniam AB, Yang D, Yu H-D, Nemiroski A, Tricard S, Ellerbee AK, et al.
3
4 2 Noncontact orientation of objects in three-dimensional space using magnetic levitation.
5
6
3 Proc Natl Acad Sci U S A. 2014;111(36):12980–5.
7 4 38. Tasoglu S, Yu CH, Liaudanskaya V, Guven S, Migliaresi C, Demirci U. Magnetic
8

pt
9 5 Levitational Assembly for Living Material Fabrication. Adv Healthc Mater.
10
11 6 2015;4(10):1469–76.
12
7 39. Picard M, Gentil BJ, McManus MJ, White K, St. Louis K, Gartside SE, et al. Acute
13

cri
14 8 exercise remodels mitochondrial membrane interactions in mouse skeletal muscle. J Appl
15
16 9 Physiol. 2013;115(10):1562–71.
17
18 10 40. Li F, Zhang C, Brakke KA, Lei Z. Computation of Equilibrium Bilayer Monodisperse
19 11 Foam Structures Using the Surface Evolver. Sci Rep. 2017;7(1):1–11.
20

us
21 12 41. Savin SF, D’yachkov LG, Myasnikov MI, Petrov OF, Fortov VE. The formation of
22
23 13 Coulomb clusters in a magnetic trap. Phys Scr. 2012;85(3):35403.
24
25
14 42. Petrov OF, Myasnikov MI, D’Yachkov LG, Vasiliev MM, Fortov VE, Savin SF, et al.
26 15 Coulomb clusters of dust particles in a cusp magnetic trap under microgravity conditions.
27
28
29
16
an
Phys Rev E - Stat Nonlinear, Soft Matter Phys. 2012;86(3):1–10.

30 17 43. D’Yachkov LG, Myasnikov MI, Petrov OF, Hyde TW, Kong J, Matthews L. Two-
31 18 dimensional and three-dimensional Coulomb clusters in parabolic traps. Phys Plasmas.
32
dM
33 19 2014;21(9): 093702.
34
35 20 44. Myasnikov MI, Petrov OF, Vasiliev MM, Fortov VE, Savin SF, Serova EO. Coulomb
36
37
21 Scatter of Diamagnetic Dust Particles in a Cusp Magnetic Trap under Microgravity
38 22 Conditions. 2017;124(2):318–9.
39
40 23 45. Susienka MJ, Wilks BT, Morgan JR, A P-PJM and FR, Mironov V VRPKVFGDCJ and
41
42 24 MRR, Fleming P A AWSGCNAFG and DCJ, et al. Quantifying the kinetics and
43
pte

25 morphological changes of the fusion of spheroid building blocks. Biofabrication.


44
45 26 2016;8(4):45003.
46
47 27 46. Fayol D, Frasca G, Le Visage C, Gazeau F, Luciani N, Wilhelm C. Use of magnetic
48
49 28 forces to promote stem cell aggregation during differentiation, and cartilage tissue
50 29 modeling. Adv Mater. 2013;25(18):2611–6.
51
ce

52 30 47. Van de Walle A, Wilhelm C, Luciani N. 3D Magnetic Stem Cell Aggregation and
53
54 31 Bioreactor Maturation for Cartilage Regeneration. J Vis Exp [Internet].
55
56
32 2017;(122):e55221–e55221.
Ac

57 33 48. Schubert T, Anders S, Neumann E, Schölmerich J, Hofstädter F, Grifka J, et al. Long-term


58
59 34 effects of chondrospheres on cartilage lesions in an autologous chondrocyte implantation
60
35 model as investigated in the SCID mouse model. Int J Mol Med. 2009;23(4):455-60.
20
Page 21 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2 1 49. Siebold R, Suezer F, Schmitt B, Trattnig S, Essig M. Good clinical and MRI outcome
3
4 2 after arthroscopic autologous chondrocyte implantation for cartilage repair in the knee.
5
6
3 Knee Surgery, Sport Traumatol Arthrosc. 2017;0(0):1–9.
7 4 50. Bartz C, Meixner M, Giesemann P, Roël G, Bulwin G-C, Smink JJ. An ex vivo human
8

pt
9 5 cartilage repair model to evaluate the potency of a cartilage cell transplant. J Transl Med.
10
11 6 2016;14(1):317.
12
7 51. Meyer U, Wiesmann HP, Libera J, Depprich R, Naujoks C, Handschel J. Cartilage defect
13

cri
14 8 regeneration by ex vivo engineered autologous microtissue - Preliminary results. In Vivo
15
16 9 (Brooklyn). 2012;26(2):251–7.
17
18 10 52. Patil US, Adireddy S, Jaiswal A, Mandava S, Lee BR, Chrisey DB. In vitro/in vivo
19 11 toxicity evaluation and quantification of iron oxide nanoparticles. Int J Mol Sci. 2015;
20

us
21 12 16(10):24417-50.
22
23 13 53. Singh D, McMillan JMM, Kabanov A V, Sokolsky-Papkov M, Gendelman HE. Bench-to-
24
25
14 bedside translation of magnetic nanoparticles. Nanomedicine. 2014;9(4):501–16.
26 15 54. Bouyer C, Chen P, Güven S, Demirtaş TT, Nieland TJ, Padilla F, Demirci U. A Bio-
27
28
29
16
an
Acoustic Levitational (BAL) Assembly Method for Engineering of Multilayered, 3D

30 17 Brain-Like Constructs, Using Human Embryonic Stem Cell Derived Neuro-Progenitors.


31 18 Adv Mater. 2016;28(1):161–7.
32
dM
33 19 55. Serpooshan V, Chen P, Wu H, Lee S, Sharma A, Hu DA, et al. Bioacoustic-enabled
34
35 20 patterning of human iPSC-derived cardiomyocytes into 3D cardiac tissue. Biomaterials.
36
37
21 2017;131:47–57.
38 22 56. Gentile C, Fleming PA, Mironov V, Argraves KM, Argraves WS, Drake CJ. VEGF-
39
40 23 mediated fusion in the generation of uniluminal vascular spheroids. Dev Dyn.
41
42 24 2008;237(10):2918–25.
43
pte

25 57. Fleming PA, Argraves WS, Gentile C, Neagu A, Forgacs G, Drake CJ. Fusion of
44
45 26 uniluminal vascular spheroids: a model for assembly of blood vessels. Dev Dyn.
46
47 27 2010;239(2):398–406.
48
49 28 58. Laschke MW, Menger MD. Life is 3D: Boosting Spheroid Function for Tissue
50 29 Engineering. Trends Biotechnol. 2017;35(2):133–44.
51
ce

52 30 59. Laschke MW, Menger MD. Spheroids as vascularization units: From angiogenesis
53
54 31 research to tissue engineering applications. Biotechnol Adv. 2017;35(6):782-791.
55
56
32
Ac

57 33
58
59 34
60
35
21
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 22 of 28

1
2 1 Figures Legends.
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25 2
26
27
28
29
3
an
Figure 1. Experimental magnetic installation. (a) Schematic diagram of a magnetic installation,
4 containing: two annular neodymium magnets, oriented with the same poles to each other; glass
30
31 5 cuvette placed in the axial hole of the magnets; backlight; two video cameras. (b) Vertical cross
32
dM
33 6 section of the magnetic field. (c) Vertical distribution of the vector of the magnetic induction at
34 7 the working area. (d) 3D model of the magnetic experimental setup. (e) Horizontal cross-section
35
36 8 of the magnetic field at the working area. (f) The resultant force acting on the vertical axis y on
37
38 9 the diamagnetic bodies, with the resultant force equal to zero, conditions of levitation are created
39
40
10 (3.9 mm for polystyrene beads, 5.1 mm for tissue spheroids).
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

22
Page 23 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37
38 1
39
40 2 Figure 2. Biofabrication of tissue spheroids for 3D microtissue construct assembly. (a) The
41
42 3 diagram for tissue spheroids biofabrication using ultra-low adhesion Corning spheroid
43
pte

44
4 microplates; (b) Phase-contrast micrograph of chondrosphere biofabricated from primary sheep
45 5 chondrocytes. Scale bar – 100µm; (c) Scanning electron microscopy micrograph of
46
47 6 chondrosphere on the surface of electrospun polyurethane matrix. Scale bar – 100µm; (d) The
48
49 7 distribution of chondrospheres diameter, n=192; (e) The distribution of chondrospheres
50
8 roundness, n=192.
51
ce

52
53
54
55
56
Ac

57
58
59
60

23
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 24 of 28

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
1
an
30 2 Figure 3. Estimation of toxic effect of Gd3+. (a) The inner structure of chondrospheres at
31
32 3 different Gd3+ concentrations. Toluidine blue staining revealed severe toxic effect of 250mM
dM
33
34 4 Gd3+ on chondrospheres morphology. Scale bar – 100µm. (b) The viability of chondrospheres
35
5 decreased essentially at 250mM Gd3+ in cell culture medium. Meanwhile 50mM Gd3+ was non-
36
37 6 toxic for tissue spheroids (c) The percentage of cells with altered morphology in chondrosheres
38
39 7 cross-sections at different Gd3+ concentrations.
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

24
Page 25 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
1
an
30 2 Figure 4. The mechanical properties of tissue spheroids. (a) Compression stress-strain
31
3 diagram for chondrosphere; (b) Elastic moduli for chondrospheres at different concentrations of
32
dM
33 4 Gd3+ in solution; n=12 (c) Compression stages in course of the testing of chondrospheres
34
35 5 mechanical properties.
36
37
38
39
40
41
42
43
pte

44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

25
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 26 of 28

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31
32
dM
33
34
35
36
37
38
39
40
41
42
43
pte

1
44
45 2 Figure 5. Fusion of tissue spheroids as function of time. (a) Phase-contrast images after 0 h, 6
46
47 3 h and 24 h of fusion for two chondrospheres. Morphological parameters measured to
48
49 4 characterize fusion stage were shown; (b) Gd3+ effect on the kinetics of contact length,
50
51
5 intersphere angle and doublet length change; (c) Gd3+ effect on the fusion of 30 tissue
ce

52 6 chondrospheres placed in ultra-low adhesive microplate. Phase-contrast micrograph. Scale bar –


53
54 7 500µm.
55
56
Ac

57
58
59
60

26
Page 27 of 28 AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30
31 1
32
dM
33 2 Figure 6. Levitation assembly of constructs in a magnetic field. (a) MD simulation of
34
35 3 chondrospheres dynamics in the cusp magnetic trap. The step by step assembly of construct from
36
37 4 polystyrene beads (b) and chondrospheres (c). (d) The kinetics of simulated and experimental
38 5 construct assembly from polystyrene beads and chondroshperes. (e) Scanning electron
39
40 6 microscopy micrograph of chondrospheres construct assembled in a magnetic field during 24
41
42 7 hours. Inserts demonstrate typical chondrocyte surface structures including multiple microvilli at
43
pte

8 high magnification.
44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

27
AUTHOR SUBMITTED MANUSCRIPT - BF-101339.R1 Page 28 of 28

1
2
3
4
5
6
7
8

pt
9
10
11
12
13

cri
14
15
16
17
18
19
20

us
21
22
23
24
25
26
27
28
29
an
30 1
31
32 2 Figure 7. Computer simulation and histology analysis demonstrated formation of gaps and
dM
33
34 3 empty spaces between tissue spheroids as result of their incomplete fusion within
35
36 4 constructs. (a) Initial touching of tissue spheroids with each other. (b) Intermediate stage of
37
5 tissue spheroids fusion. (с) The theoretical resulting construct due to tissue spheroids fusion. (d)
38
39 6 The construct histology demonstrated ongoing tissue fusion and formation of random gaps as
40
41 7 result of incomplete chondrospheres fusion.
42
43
pte

8
44
45
46
47
48
49
50
51
ce

52
53
54
55
56
Ac

57
58
59
60

28

Vous aimerez peut-être aussi