ArticlePDF Available

Three-dimensional bioprinting of thick vascularized tissues

Authors:

Abstract and Figures

Significance Current tissue manufacturing methods fail to recapitulate the geometry, complexity, and longevity of human tissues. We report a multimaterial 3D bioprinting method that enables the creation of thick human tissues (>1 cm) replete with an engineered extracellular matrix, embedded vasculature, and multiple cell types. These 3D vascularized tissues can be actively perfused with growth factors for long durations (>6 wk) to promote differentiation of human mesenchymal stem cells toward an osteogenic lineage in situ. The ability to construct and perfuse 3D tissues that integrate parenchyma, stroma, and endothelium is a foundational step toward creating human tissues for ex vivo and in vivo applications.
Three-dimensional vascularized tissue fabrication. (A) Schematic illustration of the tissue manufacturing process. (i) Fugitive (vascular) ink, which contains pluronic and thrombin, and cell-laden inks, which contain gelatin, fibrinogen, and cells, are printed within a 3D perfusion chip. (ii) ECM material, which contains gelatin, fibrinogen, cells, thrombin, and TG, is then cast over the printed inks. After casting, thrombin induces fibrinogen cleavage and rapid polymerization into fibrin in both the cast matrix, and through diffusion, in the printed cell ink. Similarly, TG diffuses from the molten casting matrix and slowly cross-links the gelatin and fibrin. (iii) Upon cooling, the fugitive ink liquefies and is evacuated, leaving behind a pervasive vascular network, which is (iv) endothelialized and perfused via an external pump. (B) HUVECs growing on top of the matrix in 2D, (C) HNDFs growing inside the matrix in 3D, and (D) hMSCs growing on top of the matrix in 2D. (Scale bar: 50 μm.) (E and F) Images of printed hMSC-laden ink prepared using gelatin preprocessed at 95 °C before ink formation (E) as printed and (F) after 3 d in the 3D printed filament where actin (green) and nuclei (blue) are stained. (G) Gelatin preprocessing temperature affects the plateau modulus and cell viability after printing. Higher temperatures lead to lower modulus and higher HNDF viability postprinting. (H) Photographs of interpenetrated sacrificial (red) and cell inks (green) as printed on chip. (Scale bar: 2 mm.) (I) Top-down bright-field image of sacrificial and cell inks. (Scale bar: 50 μm.). (J–L) Photograph of a printed tissue construct housed within a perfusion chamber (J) and corresponding cross-sections (K and L). (Scale bars: 5 mm.)
… 
Content may be subject to copyright.
Three-dimensional bioprinting of thick
vascularized tissues
David B. Kolesky
a,1
, Kimberly A. Homan
a,1
, Mark A. Skylar-Scott
a,1
, and Jennifer A. Lewis
a,2
a
School of Engineering and Applied Sciences, Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
Edited by Kristi S. Anseth, Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO, and approved February 2, 2016 (received for review
October 28, 2015)
The advancement of tissue and, ultimately, organ engineering
requires the ability to pattern human tissues composed of cells,
extracellular matrix, and vasculature with controlled microenviron-
ments that can be sustained over prolonged time periods. To date,
bioprinting methods have yielded thin tissues that only survive for
short durations. To improve their physiological relevance, we report a
method for bioprinting 3D cell-laden, vascularized tissues that exceed
1 cm in thickness and can be perfused on chip for long time periods
(>6 wk). Specifically, we integrate parenchyma, stroma, and endothe-
lium into a single thick tissue by coprinting multiple inks composed of
human mesenchymal stem cells (hMSCs) and human neonatal dermal
fibroblasts (hNDFs) within a customized extracellular matrix alongside
embedded vasculature, which is subsequently lined with human um-
bilical vein endothelial cells (HUVECs). These thick vascularized tissues
are actively perfused with growth factors to differentiate hMSCs to-
ward an osteogenic lineage in situ. This longitudinal study of emer-
gent biological phenomena in complex microenvironments represents
a foundational step in human tissue generation.
bioprinting
|
stem cells
|
vasculature
|
tissues
|
biomaterials
The ability to manufacture human tissues that replicate the
essential spatial (1), mechanochemical (2, 3), and temporal
aspects of biological tissues (4) would enable myriad applica-
tions, including 3D cell culture (5), drug screening (6, 7), disease
modeling (8), and tissue repair and regeneration (9, 10). Three-
dimensional bioprinting is an emerging approach for creating
complex tissue architectures (10, 11), including those with em-
bedded vasculature (1215), that may address the unmet needs
of tissue manufacturing. Recently, Miller et al. (15) reported an
elegant method for creating vascularized tissues, in which a
sacrificial carbohydrate glass is printed at elevated temperature
(>100 °C), protectively coated, and then removed, before in-
troducing a homogeneous cell-laden matrix. Kolesky et al. (14)
developed an alternate approach, in which multiple cell-laden, fu-
gitive (vasculature), and extracellular matrix (ECM) inks are
coprinted under ambient conditions. However, in both cases, the
inability to directly perfuse these vascularized tissues limited
their thickness (12 mm) and culture times (<14 d). Here, we
report a route for creating thick vascularized tissues (1cm)
within 3D perfusion chips that provides unprecedented control
over tissue composition, architecture, and microenvironment
over several weeks (>6 wk). This longitudinal study of emergent
biological phenomena in complex microenvironments repre-
sents a foundational step in human tissue generation.
Central to the fabrication of thick vascularized tissues is the design
of biological, fugitive, and elastomeric inks for multimaterial 3D
bioprinting. To satisfy the concomitant requirements of process-
ability, heterogeneous integration, biocompatibility, and long-term
stability, we first developed printable cell-laden inks and castable
ECM based on a gelatin and fibrinogen blend (16). Specifically,
these materials form a gelatinfibrin matrix cross-linked by a dual-
enzymatic, thrombin and transglutaminase (TG), strategy (Fig. 1
and SI Appendix,Fig.S1). The cell-laden inks must facilitate printing
of self-supporting filamentary features under ambient conditions
as well as subsequent infilling of the printed tissue architectures by
casting without dissolving or distorting the patterned construct (Fig.
1A). The thermally reversible gelation of the gelatinfibrinogen
network enables its use in both printing and casting, where gel and
fluid states are required, respectively (SI Appendix,Fig.S2).
Thrombin is used to rapidly polymerize fibrinogen (17), whereas TG
is a slow-acting Ca
2+
-dependent enzymatic cross-linker that imparts
the mechanical and thermal stability (18) needed for long-term
perfusion. Notably, the cell-laden ink does not contain either enzyme
to prevent polymerization during printing. However, the castable
matrix contains both thrombin and TG, which diffuse into adjacent
printed filaments, forming a continuous, interpenetrating polymer
network, in which the native fibrillar structure of fibrin is preserved
(SI Appendix,Fig.S3). Importantly, our approach allows arbitrarily
thick tissues to be fabricated, because the matrix does not require
UV curing (19), which has a low penetration depth in tissue (20) and
can be readily expanded to other biomaterials, including fibrin and
hyaluronic acid (SI Appendix,Fig.S4).
The gelatinfibrin matrix supports multiple cell types of in-
terest to both 2D and 3D culture conditions, including human
umbilical vein endothelial cells (HUVECs), human neonatal
dermal fibroblasts (HNDFs), and human bone marrow-derived
mesenchymal stem cells (hMSCs) (Fig. 1 BDand SI Appendix,
Fig. S5). We find that endothelial cells express vascular endo-
thelial-cadherin (VE-Cad) (Fig. 1B), and HNDFs (Fig. 1C) and
hMSCs (Fig. 1D) proliferate and spread on this matrix surface
and in bulk. Moreover, the printed cell viability can be as high as
95%, depending on how gelatin is processed before ink formu-
lation. At higher processing temperatures, the average molecular
weight of gelatin is reduced from 69 kDa at 70 °C to 32 kDa at
95 °C processing, resulting in softer gels with lower viscosity,
Significance
Current tissue manufacturing methods fail to recapitulate the
geometry, complexity, and longevity of human tissues. We
report a multimaterial 3D bioprinting method that enables the
creation of thick human tissues (>1 cm) replete with an engi-
neered extracellular matrix, embedded vasculature, and mul-
tiple cell types. These 3D vascularized tissues can be actively
perfused with growth factors for long durations (>6 wk) to
promote differentiation of human mesenchymal stem cells to-
ward an osteogenic lineage in situ. The ability to construct and
perfuse 3D tissues that integrate parenchyma, stroma, and
endothelium is a foundational step toward creating human
tissues for ex vivo and in vivo applications.
Author contributions: D.B.K., K.A.H., M.A.S.-S., and J.A.L. designed research; D.B.K., K.A.H., and
M.A.S.-S. performed research; D.B.K., K.A.H., M.A.S.-S., and J.A.L. analyzed data; and D.B.K. and
J.A.L. wrote the paper.
The authors declare no conflict of interest.
This article is a PNAS Direct Submission.
Freely available online through the PNAS open access option.
1
D.B.K., K.A.H., and M.A.S.-S. contributed equally to this work.
2
To whom correspondence should be addressed. Email: jalewis@seas.harvard.edu.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
1073/pnas.1521342113/-/DCSupplemental.
www.pnas.org/cgi/doi/10.1073/pnas.1521342113 PNAS Early Edition
|
1of6
ENGINEERING
shear yield stress, and shear elastic modulus. These cell-laden
inks can be printed with ease and accommodate cell densities
ranging from 0.1 million per mL to 10 million cells per mL (Fig.
1Eand SI Appendix, Fig. S6). Upon printing, hMSCs within this
soft gelatinfibrinogen matrix continue to spread, proliferate,
and contract into dense, cellular architectures that align along
the printing direction (Fig. 1F), likely arising due to cellular
confinement (21) and contraction via the Poisson effect (22).
To construct thick, vascularized tissues within 3D perfusion
chips, we coprinted cell-laden, fugitive, and silicone inks (Fig. 1
Hand I). First, the silicone ink is printed on a glass substrate and
cured to create customized perfusion chips (Movie S1 and SI
Appendix, Fig. S1). Next, the cell-laden and fugitive inks are
printed on chip, and then encapsulated with the castable ECM
(Fig. 1 JLand Movie S2). The fugitive ink, which defines the
embedded vascular network, is composed of a triblock copolymer
[i.e., polyethylene oxide (PEO)polypropylene oxide (PPO)PEO].
This ink can be removed from the fabricated tissue upon cooling
to roughly 4 °C, where it undergoes a gel-to-fluid transition
(14, 23). This process yields a pervasive network of inter-
connected channels, which are then lined with HUVECs. The
resulting vascularized tissues are perfused via their embedded
vasculature on chip over long time periods using an external pump
(Movie S3) that generates smooth flow over a wide range of flow
rates (24).
To demonstrate the formation of stable vasculature, we prin-
ted a simple tissue construct composed of two parallel channels
embedded within a fibroblast cell-laden matrix (Fig. 2). The
channels are lined with HUVECs, perfused with 1:1 ratio of
endothelial growth media (EGM-2 Bullet kit) and HNDF growth
media [DMEM plus 10% (vol/vol) FBS], and subsequently form
a confluent monolayer that lines each blood vessel (Fig. 2A). The
medium is preincubated for 5 h in the incubator at 37 °C and 5%
CO
2
and replaced every other day. Importantly, after 6 wk of
active perfusion, these endothelial cells maintain endothelial
phenotype and remain confluent, characterized by expression of
CD31, von Willebrand factor (vWF), and VE-Cad (Fig. 2 Band
C). The cross-sectional view of a representative vessel reveals
lumen formation (Fig. 2Dand Movie S4). Confirming the barrier
function of the endothelium, we measured a fivefold reduction
in the diffusional permeability compared with unlined (bare)
channels (Fig. 2Eand SI Appendix, Fig. S7). Stromal HNDFs
residing within the surrounding matrix exhibit cell spreading and
proliferative phenotypes localized to regions within 1mmof
Vascular ink Cell ink
Fibrinogen / Fibrin
Gelatin
Printed Cells
Thrombin
Pluronic F-127
Transglutaminase
Cell media
Endothelial cells
AA’
Section A-A’
1 cm
10
2
10
3
10
4
0
20
40
60
80
100
70 75 80 85 90 95
Plateau
Modulus
Viability
(iv)
LK
J
IH
G
E
D
hBM-MSCs
Actin
DAPI
CB
(iii)
(ii)
(i)
A
b
print
cast
evacuate
perfuse
b
HUVECs
VE-Cadherin
DAPI
hBM-MSCs
Actin
HNDFs
Smooth Muscle Actin
DAPI
BM-M
Alkaline
phosphotase
F
Fig. 1. Three-dimensional vascularized tissue fabrication. (A) Schematic illustration of the tissue manufacturing process. (i) Fugitive (vascular) ink, which contains
pluronic and thrombin, and cell-laden inks, which contain gelatin, fibrinogen, and cells, are printed within a 3D perfusion chip. (ii) ECM material, which contains
gelatin, fibrinogen, cells, thrombin, and TG, is then cast over the printed inks. After casting, thrombin induces fibrinogen cleavage and rapid polymerization into
fibrin in both the cast matrix, and through diffusion, in the printed cell ink. Similarly, TG diffuses from the molten casting matrix and slowly cross-links the gelatin
and fibrin. (iii) Upon cooling, the fugitive ink liquefies and is evacuated, leaving behind a pervasive vascular network, which is (iv) endothelialized and perfused
viaanexternalpump.(B) HUVECs growing on top of the matrix in 2D, (C) HNDFs growing inside the matrix in 3D, and (D) hMSCs growing on top of the matrix in
2D. (Scale bar: 50 μm.) (Eand F) Images of printed hMSC-laden ink prepared using gelatin preprocessed at 95 °C before ink formation (E) as printed and (F)after
3 d in the 3D printed filament where actin (green) and nuclei (blue) are stained. (G) Gelatin preprocessing temperature affects the plateau modulus and cell viability
after printing. Higher temperatures lead to lower modulus and higher HNDF viability postprinting. (H) Photographs of interpenetrated sacrificial (red) and cell
inks (green) as printed on chip. (Scale bar: 2 mm.) (I) Top-down bright-field image of sacrificial and cell inks. (Scale bar: 50 μm.). (JL) Photograph of a printed tissue
construct housed within a perfusion chamber (J) and corresponding cross-sections (Kand L). (Scale bars: 5 mm.)
2of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1521342113 Kolesky et al.
the vasculature (Fig. 2Fand SI Appendix, Fig. S8); cells further
away from these regions become quiescent likely due to an in-
sufficient nutrient supply. As cell density increases, their viability
rapidly decreases at distances beyond 1 mm from the embedded
blood vessels (e.g., only 5% of the cells remain viable at 7 mm).
Clearly, the perfusable vasculature is critical to support living
tissues thicker than 1 mm over long time periods.
To explore emergent phenomena in complex microenvironments,
we created a heterogeneous tissue architecture (>1cmthickand
10 cm
3
in volume) by printing a hMSC-laden ink into a 3D lattice
geometry along with intervening in- and out-of-plane (vertical)
features composed of fugitive ink, which ultimately transform into a
branched vascular network lined with HUVECs. After printing, the
remaining interstitial space is infilled with an HNDF-laden ECM
(Fig. 3A) to form a connective tissue that both supports and binds to
the printed stem cell-laden and vascular features. In this example,
fibroblasts serve as model cells that surround the heterogeneously
patterned stem cells and vascular network. These model cells could
be replaced with either support cells (e.g., immune cells or peri-
cytes) or tissue-specific cells (e.g., hepatocytes, neurons, or islets) in
future embodiments. The embedded vascular network is designed
with a single inlet and outlet that provides an interface between the
printed tissue and the perfusion chip. This network is symmetrically
branched to ensure uniform perfusion throughout the tissue, in-
cluding deep within its core. In addition to providing transport of
nutrients, oxygen, and waste materials, the perfused vasculature is
used to deliver specific differentiation factors to the tissue in a more
uniform manner than bulk delivery methods, in which cells at the
core of the tissue are starved of factors (25). This versatile
platform (Fig. 3A) is used to precisely control growth and dif-
ferentiation of the printed hMSCs. Moreover, both the printed
cellular architecture and embedded vascular network are visible
macroscopically with this thick tissue (Fig. 3B).
To develop a dense osteogenic tissue, we transvascularly de-
livered growth media to the tissue during an initial proliferation
phase (6 d) followed by an osteogenic differentiation mixture that is
perfused for several weeks. Our optimized mixture is composed of
BMP-2, ascorbic acid, and glycerophosphate, to promote mineral
deposition and alkaline phosphatase (AP) expression (SI Appendix,
Fig. S9). To assess tissue maturation, changes in cell function and
matrix composition are observed over time. In good agreement with
prior studies (21), we find that AP expression in hMSCs occurs
within 3 d, whereas mineral deposition does not become noticeable
until 14 d, which coincides with visible collagen-1 deposition by
hMSCs (SI Appendix,Fig.S9)(21).Fig.3Cshows an avascular
tissue produced with comparable hMSC density, in which positive
alizarin stains are only observed within a few hundred microns of
the tissue surface. By contrast, the thick vascularized tissue stains
positive in hMSC regions deep within its core after 30 d of osteo-
genic differentiation by perfusion. We characterized the mineral
deposits, which consist of particulates 20200 nm in size, using
SEM/energy-dispersive X-ray spectroscopy (EDS) analysis. Calcium
A
EF
B
D
C
Fig. 2. Three-dimensional vascularized tissues remain stable during long-term perfusion. (A) Schematic depicting a single HUVEC-lined vascular channel
supporting a fibroblast cell-laden matrix and housed within a 3D perfusion chip. (Band C) Confocal microscopy image of the vascular network after 42 d,
CD-31 (red), vWF (blue), and VE-Cadherin (magenta). (Scale bars: 100 μm.) (D) Long-term perfusion of HUVEC-lined (red) vascular network supporting HNDF-
laden (green) matrix shown by top-down (Left) and cross-sectional confocal microscopy at 45 d (Right). (Scale bar: 100 μm.) (E) Quantification of barrier
properties imparted by endothelial lining of channels, demonstrated by reduced diffusional permeability of FITC-dextran. (F) GFP-HNDF distribution within
the 3D matrix shown by fluorescent intensity as a function of distance from vasculature.
Kolesky et al. PNAS Early Edition
|
3of6
ENGINEERING
and phosphorous peaks are only observed for vascularized tissues, not
the avascular control (SI Appendix,Fig.S9Eand F). The phenotype
of hMSCs varies across the printed filamentary features: cells are
close-packed, compacted, and exhibit a high degree of mineraliza-
tion within the filament core, whereas those in the periphery are
more elongated and exhibit less mineralization. We observe that
subpopulations of HNDFs and hMSCs migrate from their initial
patterned geometry toward the vascular channels and wrap cir-
cumferentially around each channel (Fig. 3D). After 30 d, the
printed hMSCs express osteocalcin within the tissue, and osteocalcin
expression is proportional to distance from the nearest vessel (Fig.
3E). Furthermore, we find that collagen deposition is localized
A
D
E
B
C
F
G
H
I
Fig. 3. Osteogenic differentiation of thick vascularized tissue. (A) Schematic depicting the geometry of the printed heterogeneous tissue within the customized
perfusion chip, whereinthe branched vascular architecture pervades hMSCs that are printed into a 3D lattice architecture, and HNDFs are cast within an ECM that
fills the interstitial space. (B) Photographs of a printed tissue construct within and removed from the customized perfusion chip. (C) Comparative cross-sections of
avascular tissue (Left) and vascularized tissue (Right) after 30 d of osteogenicmedia perfusion with alizarin red stain showing location of calcium phosphate. (Scale
bar: 5 mm.) (D) Confocal microscopy image through a cross-section of 1-cm-thick vascularized osteogenic tissue construct after 30 d of active perfusion and in situ
differentiation. (Scale bar: 1.5 mm.) (E) Osteocalcin intensity across the thick tissue sample inside the red lines shown in C.(F) High-resolution image showing
osteocalcin (purple) localized within hMSCs, and they appear to take on symmetric osteoblast-like morphologies. (Scale bar: 100 μm.) After 30 d (Gand H), thick
tissue constructs are stained for collagen-I (yellow), which appears to be localized near hMSCs. (Scale bars: 200 μm.) (I) Alizarin red is used to stain calcium
phosphate deposition, and fast blue is used to stain AP, indicating tissue maturation and differentiation over time. (Scale bar: 200 μm.)
4of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1521342113 Kolesky et al.
within printed filaments and around the circumference of the
vasculature (Fig. 3 FHand SI Appendix, Fig. S9).
In summary, thick, vascularized human tissues with programmable
cellular heterogeneity that are capable of long-term (>6-wk) perfu-
sion on chip have been fabricated by multimaterial 3D bioprinting.
The ability to recapitulate physiologically relevant, 3D tissue mi-
croenvironments enables the exploration of emergent biological
phenomena, as demonstrated by observations of in situ development
of hMSCs within tissues containing a pervasive, perfusable, endo-
thelialized vascular network. Our 3D tissue manufacturing platform
opens new avenues for fabricating and investigating human tissues
for both ex vivo and in vivo applications.
Methods
Solution Preparation. Ink and matrix precursor solutions are prepared before
printing the tissue engineered constructs. A 15 wt/vol% gelatin solution (type A;
300 bloom from porcine skin; Sigma) is produced by warming in DPBS (1×
Dulbeccos PBS without calcium and magnesium) to 70 °C (unless otherwise noted)
and adding gelatin powder to the solution while vigorously stirring for 12 h at
70 °C (unless otherwise noted), and then the pH is adjusted to 7.5 using 1 M
NaOH. The warm gelatin solution is sterile filtered and stored at 4 °C in aliquots
for later use (<3 mo). Fibrinogen solution (50 mg·mL
1
) is produced by dis-
solving lyophilized bovine blood plasma protein (Millipore) at 37 °C in sterile
DPBS without calcium and magnesium. The solution is held at 37 °C for 45 min
to allow complete dissolution. The TG solution (60 mg·mL
1
) is prepared by
dissolving lyophilized powder (Moo Glue) in DPBS without calcium and mag-
nesium and gently mixing for 20 s. The solution is then placed at 37 °C for 20 min
and sterile filtered before use. A 250 mM CaCl
2
stock solution is prepared by
dissolving CaCl
2
powder in DPBS without calcium and magnesium (Corning). To
prepare stock solution of thrombin, lyophilized thrombin (Sigma-Aldrich) is
reconstituted at 500 U·mL
1
using sterile DPBS and stored at 20 °C. The
thrombin aliquots are thawed immediately before use.
Matrix Formulations. The solutions are mixed together at 37 °C to achieve a final
concentration of 10 mg·mL
1
fibrinogen, 7.5 wt% gelatin, 2.5 mM CaCl
2
,and
0.2 wt% TG. For printing, we use 1 wt% TG to account for diffusion and dilution
into printed cell filaments. The equilibration time before mixing with thrombin
(at a ratio of 500:1) determines optical clarity (SI Appendix,Fig.S3). After mixing,
the matrix must be quickly cast, as rapid polymerization ensues. Native fibrin
matrix is created by the same procedure without gelatin and TG (SI Appendix,Fig.
S4). Alternatively, hyaluronic acid methacrylate can be synthesized and used (26).
Ink Formulations. A silicone ink, composed of a two-part silicone elastomer (SE
1700; Dow Chemical) with a 10:1 base to catalyst (by weight), is used to create
customized perfusion chips. It is homogenized using a mixer (2,000 speed; AE-310;
Thinky Corporation) and printed within 2 h of mixing. A fugitive ink, composed of
38 wt% Pluronic F127 (Sigma) and 100 U·mL
1
thrombin in deionized, ultra-
filtrated water, is used to print the vasculature. A stock solution (40% Pluronic
F127) is homogenized using a Thinky mixer and subsequently stored at 4 °C.
Before use, 2,000 U·mL
1
thrombin solution is added to ink at a ratio of 1:20,
homogenized, loaded into a syringe (EFD, Inc.) at 4 °C, and centrifuged to
remove any air bubbles. All inks are printed at room temperature.
A cell-laden ink, composed of 7.5 wt/vol% gelatin and 10 mg·mL
1
fibrinogen, is
prepared for printing. Ink stiffness is tuned by varying the gelatin-processing
temperature (7095 °C) (SI Appendix,Fig.S6). This ink is prepared similarly to the
matrix, but without TG and thrombin. Upon printing, cross-linking is achieved by
diffusion of these enzymes from the surrounding matrix. To disperse cells in the
ink, the fibrinogengelatin blend is held at 37 °C, and then cell suspensions are
introduced via gentle pipetting. After mixing, the ink is held at 4 ° C for 15 min to
drive thermal gelation of the gelatin phase. Next, the ink is warmed to room
temperature for at least 15 min, where it can be immediately printed for up to 2 h.
FibrinogenFluorophore Conjugation. To visualize the fibrin network in printed
filaments and the cast matrix (SI Appendix,Fig.S3), fibrinogen is conjugated to
two fluorophores. Specifically, 1 g of bovine fibrinogen is dissolved in 100 mL of
50 mM borate buffer, pH 8.5 (Thermo Scientific), to form a 10 mg·mL
1
solution.
N-Hydroxysuccinimide, conjugated with either fluorescein or rhodamine, is
added at a 10:1 molar ratio of dye/fibrinogen. After reacting for 2 h at room
temperature, the labeled fibrinogen is separated from unconjugated dye by
dialysis using 10-kDa MWCO dialysis tubing in a 2-L bath against PBS for 3 d,
changing the PBS in the bath twice daily. After dialysis is complete, the fluo-
rescently conjugated fibrinogen is frozen at 80 °C, lyophilized, and stored at
20 °C before use.
Rheological Characterization. Ink rheology is measured using a controlled stress
rheometer (DHR-3; TA Instruments) with a 40-mm diameter, 2° cone and plate
geometry. The shear storage (G)andloss(G’’) moduli are measured at a fre-
quency of 1 Hz and an oscillatory strain (γ) of 0.01. Temperature sweeps are
performed using a Peltier plate over the range from 5to4C.Samplesare
equilibrated for 5 min before testing and for 1 min at each subsequent tem-
perature to minimize thermal gradients throughout the sample. Time sweeps
are conducted by rapidly placing a premixed solution onto the temperature-
controlled Peltier plate held at 37 or 22 °C, unless otherwise noted.
Cell Culture and Maintenance. hMSCs (Rooster Bio) are cultured in Booster
Media (Rooster Bio) and are not used beyond two passages. Green fluo-
rescent protein-expressing HNDFs (GFP-HNDFs) (Angio-Proteomie) are cul-
tured in Dulbeccos modified Eagle medium containing high glucose and
sodium pyruvate (DMEM) (GlutaMAX; Gibco) and supplemented with 10%
FBS (Gemini Bio-Products). Primary red fluorescent protein-expressing HUVECs
(RFP-HUVECs) (Angio-Proteomie) are cultured in EGM-2 media (complete
EGM-2 BulletKit; Lonza). GFP-HNDFs and RFP HUVECs are not used beyond
the 15th and 9th passages, respectively.
Three-Dimensional Tissue Fabrication on Perfusable Chips. All vascularized tis-
sues are created on a custom-designed multimaterial 3D bioprinter equipped with
four independently addressable print heads mounted onto a three-axis, motion-
controlled gantry with build volume of 725 ×650 ×125 mm (AGB 10000; Aer-
otech). Each ink is housed in a syringe equipped with a leur-locked nozzle of
varying size (i.e., 100-μmto410-μm diameter) (EFD, Inc.). Inks are deposited by
applying air pressure (800 Ultra dispensing system; EFD, Inc.), ranging from 10 to
140 psi, corresponding to print speeds from 1 mm·s
1
to 5 cm·s
1
.
To manufacture the customized perfusion chips, the silicone ink is loaded into a
10-mL syringe, centrifuged to remove air bubbles, and deposited through a ta-
pered 410-μm nozzle. The gasket design is created using custom MATLAB soft-
ware and the structures are printed onto 50 ×75-mm glass slides. After printing,
the chips are cured at 80 °C in an oven for >1 h and stored at room temperature.
To produce thick vascularized tissues, multiple inks are sequentially coprinted
within the customized perfusion chips. To form a base layer, a thin film of gelatin
fibrin matrix, containing 0.1 wt% TG, is cast onto the base of the perfusion chip
and allowed to dry. Next, the fugitive Pluronic F127 and cell-laden inks are
printed onto the surface using 200-μm straight and tapered nozzles, respectively.
After printing, stainless metal tubes are fed through the guide channels of the
perfusion chip and pushed into physical contact with printed vertical pillars of
the fugitive ink positioned at the inlet and outlet of each device (SI Appendix,
Fig. S1,andMovie S2). Before encapsulation, TG is added to the molten 37 ° C
gelatinfibrin matrix solution and preincubated for 220 min depending on the
desired matrix transparency (SI Appendix,Fig.S3). To form a cell-laden matrix,
the molten 37 °C gelatinfibrin matrix is first mixed with HNDF-GFP cells and
then mixed with thrombin. Next, this matrix is cast around the printed tissue,
where it undergoes rapid gelation due to thrombin activity. The 3D tissue chips
are stored at 37 °C for 1 h before cooling to 4 °C to liquefy and remove the
printed fugitive ink, which is flushed through the device using cold cell media,
leaving behind open conduits.
The 3D perfusion chips are loaded onto a machined stainless-steel base, and
a thick acrylic lid is placedon top. The lid and baseare clamped together by four
screws, forming a seal around the silicone 3D printed gasket top. Next, sterile
two-stopperistaltic tubing(PharMed BPT) is filled with mediaand connected to
the outlet of a sterile filter that is attached to a 10-mL syringe (EFD Nordson),
which servesas a media reservoir. Media that has been equilibratingfor >6hin
an incubator at 37 °C, 5% CO
2
is added to the media reservoir, and by means
of gravity, is allowed to flow through the filter and peristaltic tubing, until all
of the air is displaced, before connecting the peristaltic tubing to the inlet of
each perfusion chip. Hose pinch-off clamps are added at the inlet and outlet of
the perfusion chip to prevent uncontrolled flow when disconnected from the
peristaltic pump, which can damage the endothelium or introduce air bubbles
to the vasculature. The media reservoir is allowed to equilibrate with atmo-
spheric pressure at all times by means of a sterile filter connecting the in-
cubator environment with the reservoir.
Endothelialization of Vascular Networks. With the peristaltic tubing removed
from the chip outlet, 50500 μL of HUVEC suspensions (1 ×10
7
cells per mL)
are injected via pipette to fill the vascular network. The silicone tubing is
then replaced, and both the outlet and inlet pinch-clamp are sealed. The
perfusion chip is incubated at 37 °C to facilitate cell adhesion to the channels
under zero-flow conditions. After 30 min, the chip is flipped 180° to facili-
tate cell adhesion to the other side of the channel, and achieve circumfer-
ential seeding of cells in the channel. Finally, the cells are further incubated
for between 5 h and overnight at 37 °C before commencing active perfusion.
Kolesky et al. PNAS Early Edition
|
5of6
ENGINEERING
Active Perfusion. After endothelial cell seeding, the peristaltic tubing is
affixed to a 24-channel peristaltic pump (Ismatec), after which the hose
clamps are removed. For single vascular channels, the perfusion rate is set at
13 μL·min
1
, whereas for thick vascularized tissues, it is set at 27 μL·min
1
.
Cell Viability Assay. Cell viability is determined postprinting by printing inks
with 2 ×10
6
cells per mL for each condition. Printed cell-laden filaments (2 ×
10
6
cells per mL for each condition) are deposited onto a glass substrate and
then stained using calcein-AM (live;1μL·mL
1
; Invitrogen) and ethidium
homodimer (dead;4μL·mL
1
; Invitrogen) for 20 min before confocal im-
aging (n=3 unique samples, imaged n=10 times). To assess cell viability,
live tissue is removed from the perfusion chip, cross-sectioned, and stained
using the same staining protocol. Live and dead cell counts are obtained
using the 3D objects counter plugin in ImageJ software. The results are av-
eraged and SDs determined for each sample.
Imaging and Analysis. Photographs and videos of tissue fabrication are ac-
quired using a DSLR camera (Canon EOS, 5D Mark II; Canon). Fluorescent dyes
are used to improve visualization of Pluronic F127 (Red, Risk Reactor) and
gelatinfibrin ink (Fluorescein; Sigma-Aldrich). Printed tissue structures are
imaged using a Keyence Zoom (VHX-2000; Keyence), an inverted fluores-
cence (Axiovert 40 CFL; Zeiss), and an upright confocal microscope (LSM710;
Zeiss). ImageJ is used to generate composite microscopy images by com-
bining fluorescent channels. Three-dimensional rendering and visualization
of confocal stacks are performed in Imaris 7.6.4, Bitplane Scientific Software,
and ImageJ software. Cell counting is performed using semiautomated na-
tive algorithms in Imaris and ImageJ counting and tracking algorithms.
Immunostaining. Immunostaining and confocal microscopy are used to assess
the 3D vascularized tissues. Printed tissues are first washed with PBS via
perfusion for several minutes. Next, 10% buffered formalin is perfused
through the 3D tissue for 1015 min. The tissue is removed from the per-
fusion chip and bathed in 10% buffered formalin. A 2-h fixation time is
required for a 1-cm-thick tissue. The 3D tissues are then washed in PBS for
several hours and blocked overnight using 1 wt% BSA in PBS. Primary an-
tibodies to the cell protein or biomarker of interest are incubated with the
constructs for 2 d in a solution of 0.5 wt% BSA and 0.125 wt% Triton X-100
(SI Appendix, Table S1). Removal of unbound primary antibodies is accom-
plished using a wash step against a solution of PBS or 0.5 wt% BSA and
0.125 wt% Triton X-100 in PBS for 1 d. Secondary antibodies are incubated
with the constructs for 1 d at the dilutions listed in SI Appendix, Table S1,in
a solution of 0.5 wt% BSA and 0.125 wt% Triton X-100 in PBS. Samples are
counterstained with NucBlue or ActinGreen for 2 h and then washed for
1 d in PBS before imaging. Confocal microscopy is performed using an up-
right Zeiss LSM 710 with water-immersion objectives ranging from 10×to
40×using spectral lasers at 405-, 488-, 514-, 561-, and 633-nm wavelengths.
Image recons tructions of zstacks are performed in ImageJ using the z-project
function with the maximum pixel intensity setting. Three-dimensional image
reconstructions are performed using Imaris software.
hMSC Staining. Fast Blue (Sigma-Aldrich) and alizarin red (SigmaFast; Sigma-
Aldrich) are used to visualize AP activity and calcium deposition. One tablet of Fast
Blue is dissolved in 10 mL of deionized (DI) water. This solution is stored in the
dark and used within 2 h. Cells are washed using 0.05% Tween 20 in DPBS
without calcium and magnesium and fixed as described above. The samples are
then covered with Fast Blue solution and incubated in the dark for 510 min and
washed using PBS-Tween buffer. To assess mineralization, 2% alizarin red so-
lution is dissolved in DI water, mixed vigorously, filtered, and used within 24 h.
Samples are equilibrated in DI water and incubated with alizarin red solution for
a few minutes, then the staining solution is removed, and samples are washed
three times in DI water or until background dye is unobservable. Representative
slices of both avascular and vascularized, thick tissues are digested using 2 wt%
Collagenase I in PBS without Ca
2+
,Mg
2+
at 37 °C for >24 h. The resulting solu-
tions are filtered using a 0.2-μm sterile filter and rinsed with DI water. SEM/EDS is
used to carry out elemental analysis on harvested mineral particulates.
FITC-Dextran Permeability Testing. To assess barrier function of the printed
vasculature, diffusional permeability was quantified by perfusing culture media
in the vascular channel, while alive, containing 25 μg/mL FITC-conjugated
70-kDa dextran (FITC-Dex; Sigma product 46945) at a rate of 20 μL·min
1
for
3minand1μL·min
1
thereafter for 33 min. The diffusion pattern of FITC-Dex
was detected using a wide-field fluorescent microscope (Zeiss Axiovert 40 CFL).
Fluorescence images were captured before perfusion and every 35 min after
for 33 min. Diffusional permeability of FITC-Dex is calculated by quantifying
changes of fluorescence intensity over time using the following equation:
Pd=1
I1IbI2I1
td
4.
P
d
is the diffusional permeability coefficient, I
1
is the average intensity at an
initial time point, I
2
is an average intensity after some time (t,30 min), I
b
is
background intensity (before introducing FITC-Dex), and dis the channel
diameter (27). The measurements are performed on embedded channels
with and without endothelium (n=3).
ACKNOWLEDGMENTS. We thank Donald Ingber, David Mooney, and Christopher
Hinojosa for useful discussions; Jessica Herrmann, Humphrey Obuobi, Hayley
Price, Nicole Black, Tom Ferrante, and Oktay Uzun for their experimental
assistance; and Lori K. Sanders for help with photography and videography.
This work was supported by NSF Early-concept Grants for Exploratory Research
(EAGER) Award Division of Civil, Mechanical and Manufacturing Innovation
(CMMI)-1548261 and by the Wyss Institute for Biologically Inspired Engineering.
1. Nelson CM, Vanduijn MM, Inman JL, Fletcher DA, Bissell MJ (2006) Tissue geometry
determines sites of mammary branching morphogenesis in organotypic cultures.
Science 314(5797):298300.
2. Lee K, Silva EA, Mooney DJ (2011) Growth factor delivery-based tissue engineering: General
approaches and a review of recent developments. J R Soc Interface 8(55):153170.
3. Ingber DE (2003) Mechanobiology and diseases of mechanotransduction. Ann Med
35(8):564577.
4. Abbott RD, Kaplan DL (2015) Strategies for improving the physiological relevance of
human engineered tissues. Trends Biotechnol 33(7):401407.
5. Pampaloni F, Reynaud EG, Stelzer EHK (2007) The third dimension bridges the gap
between cell culture and live tissue. Nat Rev Mol Cell Biol 8(10):839845.
6. Huh D, Hamilton GA, Ingber DE (2011) From 3D cell culture to organs-on-chips. Trends
Cell Biol 21(12):745754.
7. Bhatia SN, Ingber DE (2014) Microfluidic organs-on-chips. Nat Biotechnol 32(8):760772.
8. Lee GY, Kenny PA, Lee EH, Bissell MJ (2007) Three-dimensional culture models of
normal and malignant breast epithelial cells. Nat Methods 4(4):359365.
9. Langer R, Vacanti JP (1993) Tissue engineering. Science 260(5110):920926.
10. Murphy SV, Atala A (2014) 3D bioprinting of tissues and organs. Nat Biotechnol 32(8):
773785.
11. Atala A, Kasper FK, Mikos AG (2012) Engineering complex tissues. Sci Transl Med
4(160):160rv12.
12. Cui X, Boland T (2009) Human microvasculature fabrication using thermal inkjet
printing technology. Biomaterials 30(31):62216227.
13. Norotte C, Marga FS, Niklason LE, Forgacs G (2009) Scaffold-free vascular tissue en-
gineering using bioprinting. Biomaterials 30(30):59105917.
14. Kolesky DB, et al. (2014) 3D bioprinting of vascularized, heterogeneous cell-laden
tissue constructs. Adv Mater 26(19):31243130.
15. Miller JS, et al. (2012) Rapid casting of patterned vascular networks for perfusable
engineered three-dimensional tissues. Nat Mater 11(9):768774.
16. Lee KY, Mooney DJ (2001) Hydrogels for tissue engineering. Chem Rev 101(7):
18691879.
17. Mosesson MW (1998) Fibrinogen structure and fibrin clot assembly. Semin Thromb
Hemost 24(2):169174.
18. Chen R-N, Ho H-O, Sheu M-T (2005) Characterization of collagen matrices crosslinked
using microbial transglutaminase. Biomaterials 26(20):42294235.
19. Stoien JD, Wang RJ (1974) Effect of near-ultraviolet and visible light on mammalian
cells in culture II. Formation of toxic photoproducts in tissue culture medium by
blacklight. Proc Natl Acad Sci USA 71(10):39613965.
20. Jayakumar MKG, Idris NM, Zhang Y (2012) Remote activation of biomolecules in deep
tissues using near-infrared-to-UV upconversion nanotransducers. Proc Natl Acad Sci
USA 109(22):84838488.
21. Klumpers DD, Zhao X, Mooney DJ, Smit TH (2013) Cell mediated contraction in 3D
cell-matrix constructs leads to spatially regulated osteogenic differentiation. Integr
Biol (Camb) 5(9):11741183.
22. Oster GF, Murray JD, Harris AK (1983) Mechanical aspects of mesenchymal morpho-
genesis. J Embryol Exp Morphol 78:83125.
23. Wu W, DeConinck A, Lewis JA (2011) Omnidirectional printing of 3D microvascular
networks. Adv Mater 23(24):H178H183.
24. Giulitti S, Magrofuoco E, Prevedello L, Elvassore N (2013) Optimal periodic perfusion
strategy for robust long-term microfluidic cell culture. Lab Chip 13(22):44304441.
25. Griffith LG, Swartz MA (2006) Capturing complex 3D tissue physiology in vitro. Nat
Rev Mol Cell Biol 7(3):211224.
26. Burdick JA, Chung C, Jia X, Randolph MA, Langer R (2005) Controlled degradation and
mechanical behavior of photopolymerized hyaluronic acid networks. Biomacromolecules
6(1):386391.
27. Price G, Tien J (2011) Methods in Molecular Biology, ed Khademhosseini A (Humana,
Totowa, NJ).
6of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1521342113 Kolesky et al.
... 45 Together, the results presented here support the contention that freeze-dried scaffolds are suitable materials for complex in vitro tissue models and offer benefits compared to collagen hydrogels: Although it has been shown that endothelial cells can self-assemble into network structures within hydrogels, they cannot be perfused without using technically difficult procedures such as templating 46,47 or sacrificial bioprinting. 48 In this investigation, we demonstrated the selfassembly of endothelial cells into hollow vessel-like structures without using any pre-patterning methods or a bioprinter, but simply by using a freeze-dried collagen type I scaffold. Beyond a high biocompatibility, the use of such scaffolds potentially allows the perfusion of the in vitro tissue model with suitable growth factors as previously investigated by Kolesky et al. ...
... 48 In this investigation, we demonstrated the selfassembly of endothelial cells into hollow vessel-like structures without using any pre-patterning methods or a bioprinter, but simply by using a freeze-dried collagen type I scaffold. Beyond a high biocompatibility, the use of such scaffolds potentially allows the perfusion of the in vitro tissue model with suitable growth factors as previously investigated by Kolesky et al. 48 and enables further systematic studies on the effect of shear stress on self-assembly of endothelial cells into a vascular network. We suggest the use of other supporting cells such as primary fibroblasts, mesenchymal stem cells (MSCs) and pericytes to further investigate the interaction of these cells with HUVECs F I G U R E 8 Immunofluorescent images show the cellular organization within hydrogel-permeated collagen type I scaffolds after 21 days of cultivation under high flow rate dynamic conditions. ...
Article
Full-text available
Despite recent advances in the field of tissue engineering, the development of complex tissue‐like structures in vitro is compromised by the lack of integration of a functioning vasculature. In this study, we propose a mesoscale three‐dimensional (3D) in vitro vascularized connective tissue model and demonstrate its feasibility to prompt the self‐assembly of endothelial cells into vessel‐like structures. Moreover, we investigate the effect of perfusion on the organization of the cells. For this purpose, primary endothelial cells (HUVECs) and a cell line of human foreskin fibroblasts are cultivated in ECM‐like matrices made up of freeze‐dried collagen scaffolds permeated with collagen type I hydrogel. A tailored bioreactor is designed to investigate the effect of perfusion on self‐organization of HUVECs. Immunofluorescent staining, two‐photon microscopy, second‐harmonic generation imaging, and scanning electron microscopy are applied to visualize the spatial arrangement of the cells. The analyses reveal the formation of hollow, vessel‐like structures of HUVECs in hydrogel‐permeated collagen scaffolds under both static and dynamic conditions. In conclusion, we demonstrate the feasibility of a 3D porous collagen scaffolding system that enables and maintains the self‐organization of HUVECs into vessel‐like structures independent of a dynamic flow.
... Most efforts have been focused on using MBB to create artificial mm-sized vascular networks. [10][11][12][13][14][15][16] DOD bioprinting methods (such as inkjet 17,18 and LIFT [19][20][21][22] ) exhibit superior spatial resolution compared to MBB 2 and have primarily been used for printing including human umbilical vein endothelial cells (HUVECs) to replicate networks known as capillary beds, a subset of the physiological microvasculature. Despite these advancements, there is a consensus in the bioprinting field that no single technology can achieve the required multiscale capability for printing multiple cell types at the scales encountered in physiological microvascular systems. ...
Article
Full-text available
We present a drop-on-demand (DOD) bioprinting method based on a novel implementation of laser-induced side transfer (LIST). Our approach involves continuous bioink perfusion through a glass capillary featuring a laser-machined hole in the capillary wall, serving as a nozzle. Focused low-energy nanosecond laser pulses are employed for precise droplet ejection. This innovative design separates the control of the bioink flow rate inside the capillary from the printing rate (drop ejection), leading to an enhanced printing workflow. We assessed the impact of key printing parameters, such as laser energy and flow conditions, on printing quality. Furthermore, we utilized the redesigned LIST to bioprint human umbilical vein endothelial cells (HUVECs). Our findings indicate that the printed HUVECs exhibit no viability loss and demonstrate the ability to recruit perivascular cells, including pericytes and fibroblasts. The redesigned LIST can be utilized in tissue engineering applications requiring DOD cell printing.
... They enhance understanding of cellular functions like transendothelial migration, particularly for less explored cell types than leukocytes such as MABs, offering valuable insights for therapy optimization [13]. 3D in vitro vessel models typically consist of a vessel model embedded in a hydrogel, which functions as an extracellular matrix substitute [17][18][19]. Fibrin, renowned for its exceptional cytocompatibility due to abundant cell adhesion molecules, serves as a precursor material for hydrogel formation and enables the release of incorporated growth factors [20][21][22]. Fibrin can be adjusted in its mechanical properties, transparency and microstructure by fibrinogen-tothrombin-ratio, calcium concentration and pH [23,24]. ...
Article
Full-text available
Systemic stem cell therapies hold promise for treating severe diseases, but their efficiency is hampered by limited migration of injected stem cells across vascular endothelium towards diseased tissues. Understanding transendothelial migration is crucial for improving therapy outcomes. We propose a novel 3D in vitro vessel model that aids to unravel these mechanisms and thereby facilitates stem cell therapy development. Our model simulates inflammation through cytokine diffusion from the tissue site into the vessel. It consists of a biofabricated vessel embedded in a fibrin hydrogel, mimicking arterial wall composition with smooth muscle cells and fibroblasts. The perfusable channel is lined with a functional endothelium which expresses vascular endothelial cadherin, provides an active barrier function, aligns with flow direction and is reconstructed by in situ two-photon-microscopy. Inflammatory cytokine release (tumor necrosis factor α, stromal-derived factor (1) is demonstrated in both a transwell assay and the 3D model. In proof-of-principle experiments, mesoangioblasts, known as a promising candidate for a stem cell therapy against muscular dystrophies, are injected into the vessel model, showing shear-resistant endothelial adhesion under capillary-like flow conditions. Our 3D in vitro model offers significant potential to study transendothelial migration mechanisms of stem cells, facilitating the development of improved stem cell therapies.
Article
Full-text available
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
Article
Full-text available
Current organ‐on‐a‐chip technologies confront limitations in effectively recapitulating the intricate in vivo microenvironments and accommodating diverse experimental conditions on a single device. Here, a novel approach for constructing a multi‐composition tumor array on a single microfluidic device, mimicking complex transport phenomena within tumor microenvironments (TMEs) and allowing for simultaneous evaluation of drug efficacy across 12 distinct conditions is presented. The TME array formed by bioprinting on a microfluidic substrate consists of 36 individual TME models, each characterized by one of three different compositions and tested under four varying drug concentrations. Notably, the TME model exhibits precise compartmentalization, fostering the development of self‐organized vascular endothelial barriers surrounding breast cancer spheroids affecting substance transport. Multivariable screening and analysis of diverse conditions, including model complexity, replicates, and drug concentrations, within a single microfluidic platform, highlight the synergistic potential of integrating bioprinting with microfluidics to evaluate drug responses across diverse TME conditions comprehensively.
Article
Full-text available
The ability to promote three‐dimensional (3D) self‐organization of induced pluripotent stem cells into complex tissue structures called organoids presents new opportunities for the field of developmental biology. Brain organoids have been used to investigate principles of neurodevelopment and neuropsychiatric disorders and serve as a drug screening and discovery platform. However, brain organoid cultures are currently limited by a lacking ability to precisely control their extracellular environment. Here, this work employs 3D bioprinting to generate a high‐throughput, tunable, and reproducible scaffold for controlling organoid development and patterning. Additionally, this approach supports the coculture of organoids and vascular cells in a custom architecture containing interconnected endothelialized channels. Printing fidelity and mechanical assessments confirm that fabricated scaffolds closely match intended design features and exhibit stiffness values reflective of the developing human brain. Using organoid growth, viability, cytoarchitecture, proliferation, and transcriptomic benchmarks, this work finds that organoids cultured within the bioprinted scaffold long‐term are healthy and have expected neuroectodermal differentiation. Lastly, this work confirms that the endothelial cells (ECs) in printed channel structures can migrate toward and infiltrate into the embedded organoids. This work demonstrates a tunable 3D culturing platform that can be used to create more complex and accurate models of human brain development and underlying diseases.
Article
Full-text available
Additive manufacturing, otherwise known as three-dimensional (3D) printing, is driving major innovations in many areas, such as engineering, manufacturing, art, education and medicine. Recent advances have enabled 3D printing of biocompatible materials, cells and supporting components into complex 3D functional living tissues. 3D bioprinting is being applied to regenerative medicine to address the need for tissues and organs suitable for transplantation. Compared with non-biological printing, 3D bioprinting involves additional complexities, such as the choice of materials, cell types, growth and differentiation factors, and technical challenges related to the sensitivities of living cells and the construction of tissues. Addressing these complexities requires the integration of technologies from the fields of engineering, biomaterials science, cell biology, physics and medicine. 3D bioprinting has already been used for the generation and transplantation of several tissues, including multilayered skin, bone, vascular grafts, tracheal splints, heart tissue and cartilaginous structures. Other applications include developing high-throughput 3D-bioprinted tissue models for research, drug discovery and toxicology.
Article
Full-text available
During embryonic development, morphogenetic processes give rise to a variety of shapes and patterns that lead to functional tissues and organs. While the impact of chemical signals on these processes is widely studied, the role of physical cues is less understood. The aim of this study was to test the hypothesis that the interplay of cell mediated contraction and mechanical boundary conditions alone can result in spatially regulated differentiation in simple 3D constructs. An experimental model consisting of a 3D cell-gel construct and a finite element (FE) model were used to study the effect of cellular traction exerted by mesenchymal stem cells (MSCs) on an initially homogeneous matrix under inhomogeneous boundary conditions. A robust shape change is observed due to contraction under time-varying mechanical boundary conditions, which is explained by the finite element model. Furthermore, distinct local differences in osteogenic differentiation are observed, with a spatial pattern independent of osteogenic factors in the culture medium. Regions that are predicted to have experienced relatively high shear stress at any time during contraction correlate with the regions of distinct osteogenesis. Taken together, these results support the underlying hypothesis that cellular contractility and mechanical boundary conditions alone can result in spatially regulated differentiation. These results will have important implications for tissue engineering and regeneration.
Article
Full-text available
In the absence of perfusable vascular networks, three-dimensional (3D) engineered tissues densely populated with cells quickly develop a necrotic core. Yet the lack of a general approach to rapidly construct such networks remains a major challenge for 3D tissue culture. Here, we printed rigid 3D filament networks of carbohydrate glass, and used them as a cytocompatible sacrificial template in engineered tissues containing living cells to generate cylindrical networks that could be lined with endothelial cells and perfused with blood under high-pressure pulsatile flow. Because this simple vascular casting approach allows independent control of network geometry, endothelialization and extravascular tissue, it is compatible with a wide variety of cell types, synthetic and natural extracellular matrices, and crosslinking strategies. We also demonstrated that the perfused vascular channels sustained the metabolic function of primary rat hepatocytes in engineered tissue constructs that otherwise exhibited suppressed function in their core.
Article
This review examines important robust methods for sustained, steady-state, in vitro culture. To achieve 'physiologically relevant' tissues in vitro additional complexity must be introduced to provide suitable transport, cell signaling, and matrix support for cells in 3D environments to achieve stable readouts of tissue function. Most tissue engineering systems draw conclusions on tissue functions such as responses to toxins, nutrition, or drugs based on short-term outcomes with in vitro cultures (2-14 days). However, short-term cultures limit insight with physiological relevance because the cells and tissues have not reached a steady-state. Copyright © 2015 Elsevier Ltd. All rights reserved.
Article
An organ-on-a-chip is a microfluidic cell culture device created with microchip manufacturing methods that contains continuously perfused chambers inhabited by living cells arranged to simulate tissue- and organ-level physiology. By recapitulating the multicellular architectures, tissue-tissue interfaces, physicochemical microenvironments and vascular perfusion of the body, these devices produce levels of tissue and organ functionality not possible with conventional 2D or 3D culture systems. They also enable high-resolution, real-time imaging and in vitro analysis of biochemical, genetic and metabolic activities of living cells in a functional tissue and organ context. This technology has great potential to advance the study of tissue development, organ physiology and disease etiology. In the context of drug discovery and development, it should be especially valuable for the study of molecular mechanisms of action, prioritization of lead candidates, toxicity testing and biomarker identification.
Article
J. A. Lewis and co-workers report on page 3124 a new bio-printing method for fabricating 3D tissue constructs replete with vasculature, multiple types of cells, and extracellular matrix. These intricate, heterogeneous structures are created by precisely co-printing multiple materials, known as bio-inks, in three dimensions. These 3D microengineered environments open new avenues for drug screening and tissue engineering. Cover image by Helena Sue Martin, David Kolesky, and Jennifer Lewis (Harvard University).
Article
Long-term cell culture in microfluidic devices is an essential prerequisite for "on a chip" biological and physiological based studies. We investigated how medium delivery, from continuous to periodic perfusion, affects long-term cell cultures in a microfluidic platform. Computational simulations suggested that different delivery strategies result in different temporal profiles of accumulation and washing out of endogenous (EnF) and exogenous (ExF) factors, respectively. Thus, cultures exposed to the same overall amount of medium with different temporal profiles were analysed in terms of homogeneity, cell morphology and phenotype. Murine and human cell lines (C2C12 and HFF) and mouse embryonic stem cells (mESC) were cultured in microfluidic channels. An ad hoc experimental setup was developed to perform continuous and periodic medium delivery into the chip, tuning the flow rate, the perfusion time, and the interval of perfusion while using the same amount of medium volume. Periodic medium delivery with a short perfusion pulse ensured cell homogeneity compared to standard cell culture. Conversely, a continuous flow resulted in cell heterogeneity, with abnormal morphology and vesiculation. Only dramatic and unfeasible increasing of perfused medium volume in the continuous configuration could rescue normal cell behaviour. Consistent results were obtained for C2C12 and HFF. In order to extend these results to highly sensitive cells, mESC were cultured for 6 days in the microfluidic channels. Our analysis demonstrates that a periodic medium delivery with fast pulses (with a frequency of 4 times per day) resulted in a homogeneous cell culture in terms of cell viability, colony morphology and maintenance of pluripotency markers. According to experimental observations, the computational model provided a rational description of the perfusion strategies and of how they deeply shape the cell microenvironment in microfluidic cell cultures. These results provide new insight to define optimal strategies for homogeneous and robust long-term cell culture in microfluidic systems, an essential prerequisite for lab on chip cell-based applications.
Article
Tissue engineering has emerged at the intersection of numerous disciplines to meet a global clinical need for technologies to promote the regeneration of functional living tissues and organs. The complexity of many tissues and organs, coupled with confounding factors that may be associated with the injury or disease underlying the need for repair, is a challenge to traditional engineering approaches. Biomaterials, cells, and other factors are needed to design these constructs, but not all tissues are created equal. Flat tissues (skin); tubular structures (urethra); hollow, nontubular, viscus organs (vagina); and complex solid organs (liver) all present unique challenges in tissue engineering. This review highlights advances in tissue engineering technologies to enable regeneration of complex tissues and organs and to discuss how such innovative, engineered tissues can affect the clinic.
Article
Controlled activation or release of biomolecules is very crucial in various biological applications. Controlling the activity of biomolecules have been attempted by various means and controlling the activity by light has gained popularity in the past decade. The major hurdle in this process is that photoactivable compounds mostly respond to UV radiation and not to visible or near-infrared (NIR) light. The use of UV irradiation is limited by its toxicity and very low tissue penetration power. In this study, we report the exploitation of the potential of NIR-to-UV upconversion nanoparticles (UCNs), which act as nanotransducers to absorb NIR light having high tissue penetration power and negligible phototoxicity and emit UV light locally, for photoactivation of caged compounds and, in particular, used for photo-controlled gene expression. Both activation and knockdown of GFP was performed in both solution and cells, and patterned activation of GFP was achieved successfully by using upconverted UV light produced by NIR-to-UV UCNs. In-depth photoactivation through tissue phantoms and in vivo activation of caged nucleic acids were also accomplished. The success of this methodology has defined a unique level in the field of photo-controlled activation and delivery of molecules.