SlideShare a Scribd company logo
1 of 127
Title
ABC/123 Version X
1
Leadership Newsletter Article
LDR/300 Version 7
1
University of Phoenix Material
Headline
[The title or overall description of the newsletter article]
Newsletter Tips
1. Interview your individual and research any information
necessary.
2. Carefully consider your individual and audience so you offer
an article that is engaging and worthwhile.
3. Create a compelling headline and opening to get your
audience’s attention.
4. Use clear and concise sentences to generate your individual’s
leadership profile.
5. Add any pictures, tables, or graphs that offer insight or
clarification for the reader.
6. Review newsletter template websites for assistance. For
example, open your web browser and search for either “smore
newsletter” or “templates cakemail”.
Opening
[A statement that describes the individual, their position, and
organization]
Content
[An interpretation of the individual’s leadership style based on
the Five-Factor personality model, and an explanation of an
incident in which the individual had to solve a difficult problem
or situation – including examples]
Wrap-Up
Conclusion
[A statement that summarizes your overall message]
Citations (APA)
[References for any citations or additional resources]
Leadership Newsletter Article Template
Copyright © XXXX by University of Phoenix. All rights
reserved.
Copyright © 2017 by University of Phoenix. All rights reserved.
REPORT
◥
BIOMEDICINE
Multivascular networks and
functional intravascular topologies
within biocompatible hydrogels
Bagrat Grigoryan1*, Samantha J. Paulsen1*, Daniel C.
Corbett2,3*, Daniel W. Sazer1,
Chelsea L. Fortin3,4, Alexander J. Zaita1, Paul T. Greenfield1,
Nicholas J. Calafat1,
John P. Gounley5†, Anderson H. Ta1, Fredrik Johansson2,3,
Amanda Randles5,
Jessica E. Rosenkrantz6, Jesse D. Louis-Rosenberg6, Peter A.
Galie7,
Kelly R. Stevens2,3,4‡, Jordan S. Miller1‡
Solid organs transport fluids through distinct vascular networks
that are biophysically and
biochemically entangled, creating complex three-dimensional
(3D) transport regimes
that have remained difficult to produce and study.We establish
intravascular andmultivascular
design freedoms with photopolymerizable hydrogels by using
food dye additives as
biocompatible yet potent photoabsorbers for projection
stereolithography.We demonstrate
monolithic transparent hydrogels, produced in minutes,
comprising efficient intravascular
3D fluid mixers and functional bicuspid valves.We further
elaborate entangled vascular
networks from space-filling mathematical topologies and
explore the oxygenation and
flow of human red blood cells during tidal ventilation and
distension of a proximate airway.
In addition, we deploy structured biodegradable hydrogel
carriers in a rodent model of chronic
liver injury to highlight the potential translational utility of this
materials innovation.
T
he morphologies of the circulatory and
pulmonary systems are physically and evo-
lutionarily entangled (1). In air-breathing
vertebrates, these bounded and conserved
vessel topologies interact to enable the
oxygen-dependent respiration of the entire or-
ganism (2–4). To build and interrogate soft hy-
drogels containing such prescribed biomimetic
and multivascular architectures, we sought to
use stereolithography (fig. S1) (5), commonly em-
ployed to efficiently convert photoactive liquid
resins into structured plastic parts through lo-
calized photopolymerization (6, 7). Comparedwith
extrusion 3D printing, which deposits voxels in
a serial fashion (8–12), photocrosslinking can
be highly parallelized via image projection to
simultaneously and independently address mil-
lions of voxels per time step. In stereolithography,
xy resolution isdeterminedby the lightpath,whereas
z resolution is dictated by light-attenuating ad-
ditives that absorb excess light and confine the
polymerization to the desired layer thickness,
thereby improving pattern fidelity. In the ab-
sence of suitable photoabsorber additives, 3D
photopatterning of soft hydrogels has been lim-
ited in the types of patterns that can be generated
(13–16) or has required complex, expensive, and
low-throughput microscopy to enhance z reso-
lution via the multiphoton effect (17–19). How-
ever, common light-blocking chemicals used for
photoresist patterning or plastic part fabrication,
such as Sudan I, are not suitable for biomanu-
facturing owing to their known genotoxic and
carcinogenic characteristics (20). Therefore, we
hypothesized that the identification of nontoxic
light blockers for projection stereolithography
could provide a major advance to the architec-
tural richness available for the design and gen-
eration of widely used biocompatible hydrogels.
Here, we establish that synthetic and natural
food dyes, widely used in the food industry, can
be applied as potent biocompatible photoabsorbers
to enable the stereolithographic production of
hydrogels containing intricate and functional
vascular architectures. We identified candidate
photoabsorbers among food additives whose ab-
sorbance spectra encompass visible light wave-
lengths that can be used for biocompatible
photopolymerization. We initially sought to gen-
eratemonolithic hydrogels, composed primarily
of water and poly(ethylene glycol) diacrylate
[PEGDA, 6 kDa, 20weight% (wt%)], with a 1-mm
cylindrical channel oriented perpendicular to the
light-projection axis. The fabrication of even this
trivial design cannot be easily realized because of
the dilute nature of such aqueous formulations,
in which the low mass fraction of crosslinkable
groups and the requisite longer polymerization
times result in inadvertent polymerization and
solidification within the narrow void spaces that
were designed to be hollowperfusable vasculature
(figs. S2 to S4).
We determined that aqueous pre-hydrogel
solutions containing tartrazine (yellow food
coloring FD&C Yellow 5, E102), curcumin (from
turmeric), or anthocyanin (from blueberries) can
each yield hydrogels with a patent vessel (figs. S2
to S5). In addition to these organic molecules,
inorganic gold nanoparticles (50 nm), widely
regarded for their biocompatibility and light-
attenuating properties (21), also function as an
effective photoabsorbing additive to generate
perfusable hydrogels (fig. S4).
To understand how these photoabsorbers
affect the gelation kinetics of photopolymeriz-
able hydrogels, we performed photorheological
characterization with short-duration light ex-
posures, which indicate that these additives cause
a dose-dependent delay in the induction of photo-
crosslinking (figs. S2D and S4E). Saturating light
exposures that extend beyond the reaction termi-
nation point demonstrate that suitable additives
did not ultimately interfere with the reaction be-
cause hydrogels eventually reached an equivalent
storage modulus independent of the additive
concentration (figs. S2D and S4E). We selected
tartrazine as a photoabsorber for further studies.
In addition to its low toxicity in humans and
broadutility in the food industry (22), we observed
that this hydrophilic dye is easily washed out of
generated hydrogels (70% elutes within 3 hours
for small gels), resulting in nearly transparent
constructs suitable for imaging (fig. S2E). Some
tartrazine may also be degraded during poly-
merization, as tartrazine is known to be sensitive
to free radicals (23). Submerging gels in water
or saline solution to remove soluble tartrazine
also flushes the vascular topology and removes
unreacted pre-hydrogel solution. In contrast to
tartrazine, curcumin is lipophilic and does not
wash out in aqueous solutions; anthocyanin has
a peak absorbance far fromour intended 405-nm
light source, requiring high concentrations for
suitable potency; and gold nanoparticles are
physically entrapped and make transmission or
fluorescence microscopy impractical (fig. S4E).
We assessed whether this materials insight
could similarly impart new architectural freedoms
tomore-advanced photoactivematerials. Photo-
absorber additives are necessary and sufficient
to enable vessel construction in thiol-ene step-
growth photopolymerization (24) of hydrogels
and in a continuous liquid interface production
(6) workflow for the generation of hydrogels (fig.
S5). We observed strong lamination between ad-
jacent fabricated layers and a rapid response
of the patterned hydrogel tomechanical deforma-
tions (fig. S6). This facile generation of soft
hydrogels with patent cylindrical vessels oriented
orthogonal to the light-projection axis sug-
gests an extensive design flexibility toward the
generation of complex vascular topologies, and
RESEARCH
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 1 of
7
1Department of Bioengineering, Rice University, Houston, TX
77005, USA. 2Department of Bioengineering, University of
Washington, Seattle, WA 98195, USA. 3Institute for Stem Cell
and Regenerative Medicine, University of Washington,
Seattle, WA 98195, USA. 4Department of Pathology,
University of Washington, Seattle, WA 98195, USA.
5Department of Biomedical Engineering, Duke University,
Durham, NC 27708, USA. 6Nervous System, Somerville, MA
02143, USA. 7Department of Biomedical Engineering, Rowan
University, Glassboro, NJ 08028, USA.
*These authors contributed equally to this work. †Present
address:
Computational Science and Engineering Division, Oak Ridge
National Laboratory, Oak Ridge, TN 37830, USA.
‡Corresponding author. Email: [email protected] (K.R.S.);
[email protected] (J.S.M.)
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
the optical clarity of resultant hydrogels implies
imaging methodologies suitable for characteri-
zation and validation of fluid flows.
Next, we investigated the ability to form
hydrogels containing functional intravascular
topologies. We first explored chaotic mixers:
intravascular topologies that homogenize fluids
as a result of interactions between fluid flow
streams and the vessel geometry (25, 26). Where-
as macroscale static mixers have found broad
utility in industrial processes (27) because of their
unparalleled efficiency, translation of intravascular
static mixers into microfluidic systems has been
difficult to implement, owing to their complex
3D topology. To this end, we generated mono-
lithic hydrogels with an integrated static mixer
composed of 3D twisted-fin elements (150 mm
thick) of alternating chirality inside a 1-mm
cylindrical channel. We applied laminar fluid
streams to the static mixer at a low Reynolds
number (0.002) and observed rapid mixing per
unit length (Fig. 1A) and as a function of fin
number (fig. S7). The elasticity and compliance of
PEG-based hydrogels (fig. S6) enabled the facile
generation of a 3D functional bicuspid venous
valve (Fig. 1B).We observed that the valve leaflets
are dynamic, respond rapidly to pulsatile antero-
grade and retrograde flows, and promote the for-
mation of stable mirror image vortices in the
valve sinuses (Fig. 1B and movie S1) according to
established mappings of native tissue (28, 29).
Solid organs contain distinct fluid networks
that are physically and chemically entangled,
providing the rich extracellular milieu that is
a hallmark of multicellular life. The ability to
fabricate such multivascular topologies within
biocompatible and aqueous environments could
enable a step change in the fields of biomaterials
and tissue engineering. A first objective is the
development of an efficient framework to design
entangled networks that can provide suitable
blueprints for their fabrication within hydrogels.
Separate vascular networks must not make a
direct fluid connection or they would topolog-
ically reduce to a single connected network. We
find that mathematical space-filling and fractal
topology algorithms provide an efficient para-
metric language to design complex vascular blue-
prints and a mathematical means to design a
second vascular architecture that does not inter-
sect the first (Fig. 2). We demonstrate a selection
of hydrogels (20 wt %, 6-kDa PEGDA) contain-
ing entangled vascular networks based on 3D
mathematical algorithms (Fig. 2, A to D): a helix
surrounding an axial vessel, 1° and 2° Hilbert
curves, a bicontinuous cubic lattice (based on a
Schwarz P surface), and a torus entangled with
a torus knot. Perfusion with colored dyes and
micro-computed tomography (mCT) analysis de-
monstrate pattern fidelity, vascular patency, and
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 2 of
7
Fig. 1. Monolithic hydrogels with functional intravascular
topologies.
(A) Monolithic hydrogels with a perfusable channel containing
integrated fin elements of alternating chirality. These static
elements rapidly promote fluid dividing and mixing (as shown
by
fluorescence imaging), consistent with a computational model
of flow
(scale bars, 1 mm). (B) Hydrogels with a functional 3D bicuspid
valve integrated into the vessel wall under anterograde and
retrograde flows (scale bars, 500 mm). Particle image
velocimetry
demonstrates stable mirror image vortices in the sinus region
behind open valve leaflets.
RESEARCH | REPORT
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
fluidic independence between the two networks
(Fig. 2, A to D, and movie S2).
We sought to evaluate the efficiency of inter-
vascular interstitial transport by measuring the
delivery of oxygen from a source vessel to per-
fused human red blood cells (RBCs) flowing in
an adjacent 3D topology. We tessellated the en-
tangled helical topology shown in Fig. 2A along
a serpentine path while maintaining the inter-
vessel distance at 300 mm (Fig. 2E). Perfusion of
deoxygenated RBCs [oxygen partial pressure
(PO2) ≤ 40 mmHg; oxygen saturation (SO2) ≤
45%] into the helical channel during ventilation of
the serpentine channel with humidified gaseous
oxygen (7 kPa) caused a noticeable color change
of RBCs fromdark red at the inlet to bright red at
the outlet (Fig. 2, E and F). Collection of perfused
RBCs showed significantly higher SO2 and PO2
relative to deoxygenated RBCs loaded at the
inlet and negative control gels ventilated with
humidified nitrogen gas (Fig. 2G and fig. S8).
Although this serpentine-helix design demon-
strates the feasibility of intervascular oxygen trans-
port between 3D entangled networks, we sought
to introduce additional structural features of
native distal lung into a bioinspired model of
alveolar morphology and oxygen transport. In
particular, the realization of 3D hydrogels that
contain branching networks and that can sup-
port mechanical distension during cyclic ventila-
tion of a pooled airway could enable investigations
of the performance of lung morphologies derived
from native structure (30) and could provide a
complete workflow for the development and ex-
amination of new functional topologies. Over the
past several decades, alveolar morphology has
been approximated mathematically as 3D space-
filling tessellations of polyhedra (31–34). However,
the translation of these ideas into useful blue-
prints has remained nontrivial because of the
need for efficient space-filling tessellations and
an ensheathing vasculature that closely tracks
the curvature of the 3D airway topography. Our
solution is to calculate a 3D topological offset of
the airway (moving each face in its local normal
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 3 of
7
Fig. 2. Entangled vascular networks. (A to D) Adaptations of
mathematical
space-filling curves to entangled vessel topologies within
hydrogels
(20 wt % PEGDA, 6 kDa): (A) axial vessel and helix, (B)
interpenetrating
Hilbert curves, (C) bicontinuous cubic lattice, and (D) torus and
(3,10) torus
knot (scale bars, 3 mm). (E) Tessellation of the axial vessel and
its
encompassing helix along a serpentine pathway. The photograph
is a
top-down view of a fabricated hydrogel with oxygen and RBC
delivery to
respective vessels. During perfusion, RBCs change color from
dark red
(at the RBC inlet) to bright red (at the RBC outlet) (scale bar, 3
mm). Boxed
regions are magnified in (F) (scale bar, 1 mm). (G) Perfused
RBCs were
collected at the outlet and quantified for SO2 and PO2. Oxygen
flow increased
SO2 and PO2 of perfused RBCs compared with deoxygenated
RBCs perfused
at the inlet (dashed line) and a nitrogen flow negative control
(N ≥ 3
replicates, data are mean ± SD, *P < 2 × 10−7 by Student’s t
test).
RESEARCH | REPORT
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 4 of
7
Fig. 3. Tidal ventilation and oxygenation in hydrogels with
vascularized
alveolar model topologies. (A) (Top) Architectural design of an
alveolar
model topology based on a Weaire-Phelan 3D tessellation and
topologic offset
to derive an ensheathing vasculature. (Bottom) Cutaway view
illustrates the
model alveoli (alv.) with a shared airway atrium. Convex (blue)
and concave
(green) regions of the airway are highlighted. (B) Photograph of
a printed
hydrogel during RBC perfusion while the air sac was ventilated
with O2 (scale
bar, 1 mm). (C) Upon airway inflation with oxygen, concave
regions of the
airway (dashed black circles) squeeze adjacent blood vessels
and cause RBC
clearance (scale bar, 500 mm). (D) A computational model of
airway inflation
demonstrates increased displacement at concave regions (dashed
yellow
circles). (E) Oxygen saturation of RBCs increased with
decreasing RBC flow
rate (N = 3, data aremean ± SD, *P < 9 × 10−4 by Student’s t
test).The dashed
line indicates SO2 of deoxygenated RBCs perfused at the inlet.
(F) Elaboration
of a lung-mimetic design through generative growth of the
airway, offset
growth of opposing inlet and outlet vascular networks, and
population of branch
tips with a distal lung subunit. (G) The distal lung subunit is
composed of a
concave and convex airway ensheathed in vasculature by 3D
offset and
anisotropicVoronoi tessellation. (H) Photograph of a printed
hydrogel containing
the distal lung subunit during RBC perfusion while the air sac
was ventilated
with O2 (scale bar, 1 mm). (I) Threshold view of the area
enclosed by the dashed
box in (H) demonstrates bidirectional RBC flow during
ventilation. (J) Distal lung
subunit can stably withstand ventilation for more than 10,000
cycles (24 kPa,
0.5 Hz) and demonstrates RBC sensitivity to ventilation gas (N2
or O2).
RESEARCH | REPORT
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 5 of
7
Fig. 4. Engraftment of functional hepatic hydrogel carriers. (A
to C)
Albumin promoter activity was enhanced in hydrogel carriers
containing
hepatic aggregates after implantation in nude mice. Data from
all time points
for each condition are shown in (B) [N = 4, *P < 0.05 by two-
way analysis of
variance (ANOVA) followed by Tukey’s post-hoc test].
Cumulative bio-
luminescence for each condition is shown in (C) (N = 4, *P <
0.05 by one-way
ANOVA followed by Tukey’s post-hoc test). Error bars indicate
SEM. GelMA,
gelatin methacrylate. (D) Gross images of hydrogels upon
resection (scale
bars, 5 mm). (E) (Left) Prevascularized hepatic hydrogel
carriers are created
by seeding endothelial cells (HUVECs) in the vascular network
after printing.
(Right) Confocal microscopy observations show that hydrogel
anchors
physically entrap fibrin gel containing the hepatocyte
aggregates (Hep)
(scale bar, 1 mm). (F) Hepatocytes in prevascularized hepatic
hydrogel
carriers exhibit albumin promoter activity after implantation in
mice with
chronic liver injury.Graft sections
stainedwithH&Eshowpositioning of hepatic
aggregates (black arrows) relative to printed (case, anchor) and
nonprinted
(fibrin) components of the carrier system (scale bar, 50 mm).
(G) Hydrogel
carriers are infiltrated with host blood (gross, H&E). Carriers
contain
aggregates that express the marker cytokeratin-18 (Ck-18) and
are in close
proximity to Ter-119–positive RBCs (scale bars, 40 mm).
RESEARCH | REPORT
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
direction) and have the new surface serve as the
template on which a vascular skeleton is built.
With this approach, we developed a bioinspired
alveolar model with an ensheathing vasculature
from 3D tessellations of the Weaire-Phelan foam
topology (35) (Fig. 3 and fig. S9). Although the
fundamental units of the Weaire-Phelan foam
are convex polyhedra (fig. S9), 3D tessellations
can produce a surface containing both convex
and concave regions reminiscent of native alveolar
air sacs (30) with a shared airway atrium sup-
porting alveolar buds (Fig. 3A). We extended
the manifold air surface in the normal direc-
tion, removed faces, and ensheathed edges in
a smoothed polygonal mesh to form a highly
branched vascular network (containing 185 vessel
segments and 113 fluidic branch points) that
encloses the airway and tracks its curvature
(fig. S9B).
We printed hydrogels (20wt%, 6-kDa PEGDA)
patterned with the alveolar model topology at a
voxel resolution of 5 pl and a print time of 1 hour
(Fig. 3B). Cyclic ventilation of the pooled airway
with humidified oxygen gas (10 kPa, 0.5Hz) led to
noticeable distension and an apparent change
in the curvature of concave airway regions (fig.
S9C). Perfusion of deoxygenated RBCs at the
blood vessel inlet (10 to 100 mm/min) during
cyclic ventilation led to observable compression
and RBC clearance from vessels adjacent to con-
cave airway regions (Fig. 3, B and C). By observing
dilute RBC streams at the early stages of per-
fusion, we also discerned that the cyclic com-
pression of RBC vessels—actuated by the concave
airway regions upon each inflation cycle—acts as
switching valves to redirect fluid streams to
neighboring vessel segments (movie S3). We im-
plemented a simplified 2D computational model
of airway inflation (fig. S9D), which predicts an-
isotropic distension of the airway and compres-
sion of adjacent blood vessels, corresponding to
local curvature (fig. S9E). In addition, analysis
from a 3D computational model supports an-
isotropic distension of the concave regions of the
airway during inflation (Fig. 3D). Despite the
volume of the alveolar model hydrogel (0.8 ml)
being <25% of that of the serpentine-helix model
(3.5 ml), we measured similar oxygenation ef-
ficiencies for the two designs (Fig. 3E). Our data
suggest that branching topology, hydrogel dis-
tension, and redirection of fluid streams during
ventilation may boost intravascular mixing and
allow faster volumetric uptake of oxygen by the
well-mixed RBCs. Vascular constriction during
breathing has been previously described as an
important fluid control mechanism in the mam-
malian lung (36), and here we provide a means
to actualize these ideas in completely defined
and biocompatiblematerials andwithin aqueous
environments.
To extend this work toward a coherent ap-
proximation of scalable lung-mimetic design, we
must consolidate the location of the vascular
inlet, vascular outlet, and air duct, such that dis-
tal lung subunits can be populated on the tips
of multiscale branching architecture. Therefore,
within a given computational bounding volume,we
first derive a branching airway (Fig. 3F). Next,
the centerlines of inlet and outlet blood vessel
networks are grown 180° opposite each other
across and topologically offset from the airway,
and the blood vessels traverse down to the tips of
all daughter branches. The final step is to pop-
ulate the tips of each distal lung with an alveolar
unit cell (Fig. 3G andmovie S4) whose ensheath-
ing vasculature (containing 354 vessel segments
and 233 fluidic branch points) itself is an an-
isotropic Voronoi surface tessellation along a
topological offset of its local airway (fig. S9, F
and G).We found that hydrogels (20 wt %, 6-kDa
PEGDA) could withstand more than 10,000 ven-
tilation cycles (at 24 kPa and a frequency of
0.5 Hz) over 6 hours during RBC perfusion and
while switching the inflow gas between humidi-
fied oxygen and humidified nitrogen (Fig. 3, H
to J). Color-filtered views of the early stages of
RBC perfusion (Fig. 3I) indicate that ventilation
promotes RBC mixing and bidirectional flows
within selected vessel segments near the mid-
point of the distal lung subunit (movie S4).
We use our custom stereolithography appa-
ratus for tissue engineering (SLATE) to demon-
strate production of tissue constructs containing
mammalian cells (figs. S1, S10, and S11 andmovie
S5). Lung-mimetic architectures can also be pop-
ulatedwith human lung fibroblasts in the bulk of
the interstitial space and human epithelial-like
cells in the airway (fig. S12), which could facilitate
the development of a hydrogel analog of a lab-
on-a-chip lung design (37). Finally, we subjected
primary humanmesenchymal stem cells (hMSCs)
to SLATE fabrication (with mixtures of PEGDA
and gelatinmethacrylate) and show that the cells
within cylindrical fabricated hydrogels remain
viable and can undergo osteogenic differentia-
tion (fig. S13D). In related multiweek perfusion
tissue culture of hMSCs with osteogenic differ-
entiation media, osteogenic marker–positive
hMSCs were visible throughout the gel (fig. S14).
These studies indicate that SLATE fabrication
supports rapid biomanufacturing, can maintain
the viability of mammalian cell lines, supports
the normal function and differentiation of pri-
mary human stem cells, and provides an ex-
perimentally tractablemeans to explore stem cell
differentiation as a function of soluble factor
delivery via vascular perfusion.
We next sought to establish the utility of this
process for fabricating structurally complex and
functional tissues for therapeutic transplanta-
tion. In particular, the liver is the largest solid
organ in the human body, carrying out hundreds
of essential tasks in a manner thought to be
dependent on its structural topology. We created
complex structural features in hydrogel within
the expanded design space imparted by SLATE
to assemblemultimaterial liver tissues. Bioprinted
single-cell tissues andbioprintedhydrogel carriers
containing hepatocyte aggregates were fabricated
(Fig. 4, A to C). The albumin promoter activity
of tissue carriers loaded with aggregates was
enhanced by more than a factor of 60 compared
with that of implanted tissues containing single
cells (Fig. 4, B and C). Furthermore, upon gross
examination of tissues after resection, hydrogel
carrier tissues appeared to havemore integration
with host tissue and blood (Fig. 4D). Despite the
improved utility of hepatic aggregates over single
cells, aggregate size puts substantial architectural
limitations on 3D printing because aggregates
are larger in size than our lowest voxel resolution
(50 mm). To accommodate these design con-
straints, we built a more advanced carrier that
can deliver hepatic aggregates within natural
fibrin gel, has a vascular compartment that can
be seeded with endothelial cells, and incorpo-
rates structural hydrogel anchors to physically,
rather than chemically, retain the fibrin gel and
facilitate remodeling between the graft and host
tissue (Fig. 4E and fig. S15). Microchannel net-
works were seeded with human umbilical vein
endothelial cells (HUVECs) because our previous
studies demonstrated that inclusion of endothelial
cords improved tissue engraftment (38). We then
evaluated whether optimized bioengineered liver
tissueswould survive transplantation in a rodent
model of chronic liver injury. After 14 days of
engraftment in mice with chronic liver injury,
hepatic hydrogel carriers exhibited albumin pro-
moter activity indicative of surviving functional
hepatocytes (Fig. 4F). Immunohistological char-
acterization revealed the presence of hepatic
aggregates adhered to printed hydrogel com-
ponents that stained positively for the marker
cytokeratin-18 (Fig. 4, F and G). Further charac-
terization through gross examination and higher-
magnification images of slides stained with
hematoxylin and eosin (H&E) indicated the pres-
ence of host blood in explanted tissues. Immu-
nostaining using a monoclonal antibody against
Ter-119 confirmed the erythroid identity of cells in
microvessels adjacent to hepaticmicroaggregates
in explanted tissues (Fig. 4G, right). This work
provides an approach to address long-standing
design limitations in tissue engineering that have
hindered progress of preclinical studies.
We have identified readily available food dyes
that can serve as potent photoabsorbers for bio-
compatible and cytocompatible production of
hydrogels containing functional vascular to-
pologies for studies of fluidmixers, valves, inter-
vascular transport, nutrient delivery, and host
engraftment.With our stereolithographic process,
there is potential for simultaneous and orthog-
onal control over tissue architecture and bio-
materials for the design of regenerative tissues.
REFERENCES AND NOTES
1. R. Monahan-Earley, A. M. Dvorak, W. C. Aird,
J. Thromb. Haemost. 11 (suppl. 1), 46–66 (2013).
2. G. R. Scott, J. Exp. Biol. 214, 2455–2462 (2011).
3. E. R. Schachner, J. R. Hutchinson, C. Farmer, PeerJ 1, e60
(2013).
4. C. G. Farmer, Physiology 30, 260–272 (2015).
5. Supplementary figures, as well as materials and methods, are
available as supplementary materials.
6. J. R. Tumbleston et al., Science 347, 1349–1352 (2015).
7. B. E. Kelly et al., Science 363, 1075–1079 (2019).
8. J. S. Miller et al., Nat. Mater. 11, 768–774 (2012).
9. T. J. Hinton et al., Sci. Adv. 1, e1500758 (2015).
10. T. Bhattacharjee et al., Sci. Adv. 1, e1500655 (2015).
11. D. B. Kolesky, K. A. Homan, M. A. Skylar-Scott, J. A.
Lewis,
Proc. Natl. Acad. Sci. U.S.A. 113, 3179–3184 (2016).
12. H.-W. Kang et al., Nat. Biotechnol. 34, 312–319 (2016).
Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 6 of
7
RESEARCH | REPORT
on A
pril 14, 2020
http://science.sciencem
ag.org/
D
ow
nloaded from
http://science.sciencemag.org/
13. V. Liu Tsang et al., FASEB J. 21, 790–801 (2007).
14. H. Lin et al., Biomaterials 34, 331–339 (2013).
15. J. A. S. Neiman et al., Biotechnol. Bioeng. 112, 777–787
(2015).
16. X. Ma et al., Proc. Natl. Acad. Sci. U.S.A. 113, 2206–2211
(2016).
17. M. S. Hahn, J. S. Miller, J. L. West, Adv. Mater. 18, 2679–
2684
(2006).
18. C. A. DeForest, K. S. Anseth, Nat. Chem. 3, 925–931
(2011).
19. K. A. Heintz et al., Adv. Healthc. Mater. 5, 2153–2160
(2016).
20. T. M. Fonovich, Drug Chem. Toxicol. 36, 343–352 (2013).
21. S. Kumar, J. Aaron, K. Sokolov, Nat. Protoc. 3, 314–320
(2008).
22. L. J. Stevens, J. R. Burgess, M. A. Stochelski, T. Kuczek,
Clin. Pediatr. 54, 309–321 (2015).
23. M. Li et al., J. Agric. Food Chem. 62, 12052–12060 (2014).
24. C. A. DeForest, B. D. Polizzotti, K. S. Anseth, Nat. Mater.
8,
659–664 (2009).
25. …
3 1 2 VOLUME 34 NUMBER 3 MARCH 2016 nature
biotechnology
A r t i c l e s
The demand for engineered tissues has risen rapidly owing to
the
limited availability of donor tissues and organs for
transplantation.
Despite some initial successes in engineering relatively simple
tissues,
many challenges remain in developing tissues and organs
suitable for
clinical translation1,2. Three-dimensional (3D) printing
technology
shows promise for creating complex composite tissue
constructs3–8
through precise placement of cell-laden hydrogels in a layer-by-
layer
fashion7,9–17. The most commonly used bioprinting systems
are based
on jetting, extrusion and laser-induced forward transfer
(LIFT)6,18,19.
The jetting method produces picoliter scale drops with a
printing
resolution of 20~100 µm. However, because the hydrogel
concen-
tration is low20–23, the thickness of printed constructs may be
lim-
ited because of inadequate structural support24. Extrusion
methods,
which use a syringe and piston system to dispense material
through
microscale nozzles, can produce more stable 3D cell-laden
structures
using high concentrations of hydrogels such as alginate, fibrin
and
Pluronic F-127 (refs. 18,25–27). However, it is difficult to
construct
large free-form tissue structures owing to inadequate structural
integ-
rity, mechanical stability and printability28–30. The LIFT
method can
precisely print cells in relatively small constructs31 but requires
rapid
gelation of hydrogels to achieve high resolution of the printed
pat-
terns, resulting in low flow rates.
Here we describe a system that deposits cell-laden hydrogels
together
with synthetic biodegradable polymers that impart mechanical
strength, thereby overcoming previous limitations on the size,
shape,
structural integrity and vascularization of bioprinted tissue con-
structs. This was accomplished by designing multidispensing
modules
for delivering various cell types and polymers in a single
construct;
by developing an optimized carrier material for delivering cells
to
discrete locations in the 3D structure in a liquid form; by
designing
sophisticated nozzle systems with a resolution down to 2 µm for
bio-
materials and down to 50 µm for cells; by cross-linking cell-
laden
hydrogels after passage though the nozzle system; by
simultaneously
printing an outer sacrificial acellular hydrogel mold that is
dissolved
after the tissue construct acquires enough rigidity to retain its
shape;
and by creating a lattice of microchannels permissive to nutrient
and
oxygen diffusion into the printed tissue constructs. These
properties,
all designed to work in a coordinated manner, make up the
ITOP. We
demonstrate the printer by fabricating human-scale mandible
bone,
ear-shaped cartilage and organized skeletal muscle. Evaluation
of
the characteristics and function of these tissues in vitro and in
vivo
showed tissue maturation and organization that may be
sufficient for
translation to patients.
RESULTS
Design of the ITOP system
Multiple cartridges (Fig. 1a and Supplementary Fig. 1) are used
to
deliver and pattern multiple cell-laden composite hydrogels,
support-
ing poly(ε-caprolactone) (PCL) polymer and a sacrificial
Pluronic
F-127 hydrogel (Fig. 1b). The end of each cartridge is
connected
to a microscale nozzle, and the top is connected to an air
pressure
controller for precisely controlling the dispensing volume. A
heating
unit ensures that the PCL remains easily dispensable. A three-
axis
motorized stage system enables 3D patterning of multiple cells
and
biomaterials. The system resides in a humidified and
temperature-
controlled (18 °C) enclosure.
The correct shape of a tissue construct is obtained from a human
body by processing computed tomography (CT) or magnetic
resonance
imaging (MRI) data in computer-aided design (CAD) software
(Fig. 1c).
A custom nozzle motion program is generated by incorporating
A 3D bioprinting system to produce human-scale
tissue constructs with structural integrity
Hyun-Wook Kang, Sang Jin Lee, In Kap Ko, Carlos Kengla,
James J Yoo & Anthony Atala
A challenge for tissue engineering is producing three-
dimensional (3D), vascularized cellular constructs of clinically
relevant
size, shape and structural integrity. We present an integrated
tissue–organ printer (ITOP) that can fabricate stable, human-
scale
tissue constructs of any shape. Mechanical stability is achieved
by printing cell-laden hydrogels together with biodegradable
polymers in integrated patterns and anchored on sacrificial
hydrogels. The correct shape of the tissue construct is achieved
by
representing clinical imaging data as a computer model of the
anatomical defect and translating the model into a program that
controls the motions of the printer nozzles, which dispense cells
to discrete locations. The incorporation of microchannels into
the tissue constructs facilitates diffusion of nutrients to printed
cells, thereby overcoming the diffusion limit of 100–200 mm for
cell survival in engineered tissues. We demonstrate capabilities
of the ITOP by fabricating mandible and calvarial bone,
cartilage
and skeletal muscle. Future development of the ITOP is being
directed to the production of tissues for human applications and
to
the building of more complex tissues and solid organs.
Wake Forest Institute for Regenerative Medicine, Wake Forest
School of Medicine, Medical Center Boulevard, Winston-Salem,
North Carolina, USA.
Correspondence should be addressed to A.A.
([email protected]).
Received 27 July 2015; accepted 19 October 2015; published
online 15 February 2016; doi:10.1038/nbt.3413
np
g
©
2
01
6
N
at
ur
e
A
m
er
ic
a,
In
c.
A
ll
ri
gh
ts
r
es
er
ve
d.
http://dx.doi.org/10.1038/nbt.3413
http://www.nature.com/naturebiotechnology/
nature biotechnology VOLUME 34 NUMBER 3 MARCH
2016 3 1 3
A r t i c l e s
the printing pattern in combination with
fabrication conditions (for example, scan
speed, temperature, material information
and air pressure). The ITOP uses a text-based
motion program consisting of a command list
for XYZ stage movements and air pressure
actuation (Supplementary Source Code).
The motion program is then transferred to
the main computer of the 3D printing system
that effects the biofabrication process.
The composite hydrogel for cell delivery
consisted of gelatin, fibrinogen, hyaluronic
acid (HA) and glycerol mixed into DMEM (high glucose). We
tested
various concentrations of each component to achieve proper
print-
ing resolution and dispensing uniformity, mechanical properties
(before and after cross-linking with thrombin) and cell viability
(Supplementary Fig. 2). The optimized concentrations of hydro-
gel ingredients and the numbers of cells needed for fabrication
of
individual tissue constructs are listed in Table 1.
Patterning synthetic polymers confers mechanical strength
Our first series of studies tested the ability of the ITOP to
generate uni-
form two-dimensional (2D) and 3D cell patterns of multiple cell
types.
Using 3T3 fibroblasts labeled with two fluorescent dyes, Dil
(red) and
DiO (green), we demonstrated delivery of the two populations
in a
unique 2D pattern (Fig. 2a,b). To create 3D constructs, we
combined
the fluorescently labeled 3T3 fibroblasts in composite hydrogels
with
supporting PCL and printed them in two patterns—type I (Fig.
2c)
and type II (Fig. 2f). These patterns differ in the placement of
PCL and
thus in the mechanical strength of the printed construct. The
type I
pattern creates multiple PCL frames in each layer throughout
the con-
struct, and places cells and gel materials in between the frames.
The
type II pattern consists of cell-laden hydrogel and porous
structures,
surrounded by a PCL framework on the outer layers and corners
of
each layer, thus protecting the contents from external load.
Type I
constructs (Fig. 2d,e) maintained a more stable structure than
type II
constructs (Fig. 2g,h), owing to the abundance of uniformly
distrib-
uted PCL frames. Therefore, we used the type I pattern to
fabricate
mandible bone and ear-shaped cartilage structures and the type
II
pattern to print organized skeletal muscle constructs.
Next, we produced 3D structures by placing either type I or type
II pat-
terns of cell-laden hydrogel and PCL (~130 µm for type I and
~250 µm
wide for type II). The microchannels (type I: 500 × 300 µm2;
type II:
650 × 450 µm2), formed by the PCL patterns, were designed to
maxi-
mize diffusion of nutrients and oxygen. In addition, we used
Pluronic
F-127 hydrogel as a sacrificial outer layer to support the 3D
architecture
of the dispensed cell-laden structures before crosslinking. After
cross-
linking of fibrinogen using thrombin, the uncross-linked
components
(gelatin, HA, glycerol and Pluronic F-127) were washed out.
To determine cell viability during printing, we examined
survival of
3T3 fibroblasts at 60 min (day 0), 3 d and 6 d after printing.
Live/dead
cell assays showed ≥95% cell viability on day 0, which was
maintained
through days 3 and 6 (Fig. 2i). Cell proliferation, assessed using
the
AlamarBlue assay system, increased over a 15-d period, similar
to the
proliferation of control cells encapsulated in a fibrin construct
(Fig. 2j).
These data indicate that the optimized composite hydrogel
system
maintained cell viability during the printing process and
provided a
favorable microenvironment for cell proliferation.
Mandible bone reconstruction
To demonstrate construction of a human-sized bone structure,
we fab-
ricated a mandible fragment in a size and shape similar to what
would
be needed for facial reconstruction after traumatic injury (Fig.
3).
The cell type used was human amniotic fluid–derived stem cells
Main
computer
3-axis stage
controller
Pressure
controller
Multi-
cartridge
module
3D
printed
construct
PCL (gray)
A: cell A (red)
B: cell B (green)
S: sacrificial material
Closed chamber
Medical imaging
(CT, MRI)
Visualized motion
program
3D printing process
3D bioprinted
tissue product
10 mm
DICOM format STL format Text-based
command list
3D printed
construct
PCL
Cell A
Cell B
XYZ
stage
Heating
unit
PCL
A B S
Po
re
(m
icr
oc
ha
nn
el)
3D CAD model
a b
c
Figure 1 ITOP system. (a) The ITOP system
consists of three major units: (i) 3-axis stage/
controller, (ii) dispensing module including
multi-cartridge and pneumatic pressure
controller and (iii) a closed acrylic chamber
with temperature controller and humidifier.
(b) Illustration of basic patterning of 3D
architecture including multiple cell-laden
hydrogels and supporting PCL polymer.
(c) CAD/CAM process for automated printing of
3D shape imitating target tissue or organ. A 3D
CAD model developed from medical image data
generates a visualized motion program, which
includes instructions for XYZ stage movements
and actuating pneumatic pressure to achieve
3D printing.
Table 1 Preparation of the cell-laden composite hydrogels for
3D bioprinted tissue constructs
Composite hydrogel
Gelatin Fibrinogen HA Glycerol Cell type & density Cell
viability Remark
Bone (type I) 35 mg/ml 20 mg/ml 3 mg/ml 10% v/v Human
AFSCs, 5 × 106 cells/ml 91 ± 2% (day 1) Figs. 3 and 4
Cartilage (Type I) 45 mg/ml 30 mg/ml 3 mg/ml 10% v/v Rabbit
ear chondrocytes, 40 × 106 cells/ml 91 ± 8% (day 1) Fig. 5
Skeletal muscle (type II) 35 mg/ml 20 mg/ml 3 mg/ml 10% v/v
Mouse C2C12 myoblasts, 3 × 106 cells/ml 97 ± 6% (day 1) Fig.
6
np
g
©
2
01
6
N
at
ur
e
A
m
er
ic
a,
In
c.
A
ll
ri
gh
ts
r
es
er
ve
d.
3 1 4 VOLUME 34 NUMBER 3 MARCH 2016 nature
biotechnology
A r t i c l e s
(hAFSCs), which can give rise to osteogenic lineages in
appropriate
media32,33. Mandible bone defects have an arbitrary shape. We
used
data from a CT scan of a human mandible defect in combination
with Mimics software (Materialise, Leuven, Belgium) to
produce a
CAD model of the defect shape, with dimensions of 3.6 cm ×
3.0 cm
× 1.6 cm (Fig. 3a). A text-based command motion program,
gener-
ated from the CAD model with custom CAM software,
determined
the required dispensing paths of cell-laden hydrogel, a mixture
of
PCL and tricalcium phosphate (TCP), and Pluronic F127 (Fig.
3b).
PCL/TCP and hAFSCs mixed with the composite hydrogel
(Table 1)
were printed in a type I pattern with a Pluronic F127 temporary
sup-
port (Fig. 3c). At 1 d of culture, cell viability in the printed
bone
structures was 91 ± 2% (n = 3, Table 1), confirming that the
printing
process did not adversely affect cell viability. After induction
of osteo-
genic differentiation using an established protocol32,33 for 28 d
(n = 5,
Fig. 3d), we stained the structures with Alizarin Red S; staining
at the
surface of the 3D bone structures indicated calcium deposition
in the
hAFSC-laden hydrogel (Fig. 3e). 3D constructs before
differentiation
showed no Alizarin Red S staining (data not shown).
Calvarial bone reconstruction
To study maturation of the bioprinted bone in vivo, we
fabricated rat
calvarial bone constructs in a circular shape (8 mm diameter ×
1.2 mm
thickness) with hAFSCs (Fig. 4a,b and Supplementary Fig. 3),
cultured
them in osteogenic media for 10 d, implanted
them in a calvarial bone defect region of
Sprague Dawley rats (n = 4) and analyzed
them 5 months after implantation (Fig. 4c).
Type l
Pore
1 mm
1 mm
1 mm
1 mm
Type l Top Type l - top
Type ll
PCL
PCL
Cell-laden
hydrogel
Cell-laden
hydrogel
Side
Top
Side
Type ll Type ll – top
500 µm
120
100
80
60
40
20
0
0 3 6
Culture time (d)
C
e
ll
vi
a
b
ili
ty
(
%
)
20
15
10
5
0
Culture time (d)
N
o
rm
a
liz
e
d
in
te
n
si
ty
Control
Printed
500 µm
1 3 7 15
a b
c d e
f g h
i
j
Figure 2 2D/3D patterning using the ITOP system. (a,b) 2D
patterning of ‘WFIRM’ characters written by cell-laden
hydrogels through the integrated organ
printing. Microscopic (a) and fluorescent images (b) of
‘WFIRM’ characters, which were produced using cells labeled
with Dil and DiO. (c–h) Two basic
types of 3D patterning: type I pattern (c–e) and type II pattern
(f–h). Two types of 3D patterning, including cell-A (red), cell-B
(blue) and PCL (green),
were fabricated by the integrated organ printing (c,f);
photographs (d,g) and fluorescent image (e,h) of the 3D printed
patterns. (i) Cell viability was over
95% on day 0 and then maintained on days 3 and 6 (n = 3). (j)
Cell proliferation results showed that the number of cells
continuously increased over a
15-d period, and no significant differences between the control
and the printed constructs were observed (n = 5). Error bars,
mean ± s.d.
Bony
defect
Red: cells
Green: PCL
Blue: Pluronic F-127
Pluronic
F-127 (sacrificial
material)
Printing nozzle
Pore
PCL
Cell-laden
hydrogel
30 mm
3
6
m
m
P
o
re
PCL
Cell-laden
hydrogel
1 mm
a b c
d e
Figure 3 Mandible bone reconstruction.
(a) 3D CAD model recognized a mandible
bony defect from human CT image data.
(b) Visualized motion program was generated
to construct a 3D architecture of the mandible
bone defect using CAM software developed
by our laboratory. Lines of green, blue and
red colors indicate the dispensing paths of
PCL, Pluronic F-127 and cell-laden hydrogel,
respectively. (c) 3D printing process using the
integrated organ printing system. The image
shows patterning of a layer of the construct.
(d) Photograph of the 3D printed mandible
bone defect construct, which was cultured in
osteogenic medium for 28 d. (e) Osteogenic
differentiation of hAFSCs in the printed
construct was confirmed by Alizarin Red
S staining, indicating calcium deposition.
np
g
©
2
01
6
N
at
ur
e
A
m
er
ic
a,
In
c.
A
ll
ri
gh
ts
r
es
er
ve
d.
nature biotechnology VOLUME 34 NUMBER 3 MARCH
2016 3 1 5
A r t i c l e s
The bioprinted constructs showed newly
formed vascularized bone tissue through-
out the implants, including the central por-
tion, with no necrosis (Fig. 4j), whereas
the untreated defect and scaffold-only
treated control groups showed fibrotic tis-
sue ingrowth (Fig. 4d) and minimal bone
tissue formation restricted to the periphery
of the implant (Fig. 4g), respectively. The
modified Tetrachrome staining confirmed
mature bone (red) and osteoid (blue) for-
mation (Fig. 4e,h,k). Von Willebrand factor
(vWF) immunostaining showed large blood
vessel formation within newly formed bone
tissue throughout the bioprinted bone con-
structs, including the central portion (Fig. 4l), whereas the
nontreated
(Fig. 4f) and scaffold-only (Fig. 4i) groups had only limited
vascu-
larization restricted to the periphery of the implant.
Ear cartilage reconstruction
Next, we tested the ability of the ITOP to fabricate tissue
constructs
of complex shape by making human-sized external ears, as the
frame-
work of an auricle consists of a single piece of cartilage with a
com-
plicated geometry of ridges. A CT image of an ear (Fig. 5a) was
used
to develop a motion program (Fig. 5b) to print a chondrocyte-
laden
hydrogel, PCL and Pluronic F-127. Using rabbit ear
chondrocytes
(passages 3 and 4) mixed with the composite hydrogel (Table
1), we
fabricated human ear–shaped cartilage constructs with
dimensions of
3.2 cm × 1.6 cm × 0.9 cm (Fig. 5c–e) in the type I pattern. Cell
viabil-
ity was 91 ± 8% at 1 d after printing (n = 3, Table 1). After 5
weeks
in the culture medium, the constructs were stained with
Safranin-O
and showed production of a new cartilaginous matrix (Fig. 5f).
The constructs with microchannels showed enhanced tissue
forma-
tion as evidenced by the production of new viable cartilaginous
matrix
throughout the entire ear constructs. In contrast, the constructs
without microchannels showed only limited tissue formation
restricted to the peripheral region, likely owing to the diffusion
limits of nutrients and oxygen. The cells in the newly formed
tissues
demonstrated similar morphological characteristics to those in
native
ear cartilage, with cells located within typical chondrocyte
lacunae,
surrounded by a cartilaginous matrix (Fig. 5g). Native human
ear
tissue served as a positive control.
To determine whether the printed ear constructs would mature
in vivo,
we implanted them in the dorsal subcutaneous space of athymic
mice
and retrieved them 1 and 2 months after implantation (n = 4).
The
shape was well maintained, with substantial cartilage formation
upon
gross examination (Fig. 5h). Histological analysis showed the
for-
mation of cartilage tissue (Fig. 5i). The glycosaminoglycan
(GAG)
content (2.7 ± 0.2 µg/mg at 1 month and 4.2 ± 0.3 µg/mg at 2
months)
increased over time, reaching 20% of that of native ear GAG
content
(Fig. 5j). Vascularization of the printed constructs in the outer
region
was suggested by endothelial cell marker expression at 1 and 2
months
after implantation (Supplementary Fig. 4). The inner regions
were
avascular (Supplementary Fig. 4), as in native cartilage, but the
car-
tilage cells were viable, suggesting adequate nutrient diffusion
during
development. Biomechanical analyses (n = 4, Fig. 5k) showed
that
maturation in vivo strengthened the tissue constructs, resulting
in a
higher normalized load during bending compared with pre-
implant
constructs. In addition, resilience, measured by the ∆Load%,
was tested
by repeated bending and relaxation cycles. Resilience between
the
repeated bending cycles was much higher in the constructs
implanted
for 1 month (Fig. 5m and Supplementary Table 1) than in the
con-
structs before implantation (Fig. 5l) . These results demonstrate
the
generation of ear-shaped cartilage with resilience properties
similar to
those of native cartilage (rabbit ear) (Supplementary Table 1).
Skeletal muscle reconstruction
Finally, we applied the ITOP to fabricate an organized soft
tissue—a
3D muscle construct 15 mm × 5 mm × 1 mm in dimension
containing
Figure 4 Calvarial bone reconstruction.
(a) Visualized motion program (top) used to print
a 3D architecture of calvarial bone construct.
Green and red color lines indicate the dispensing
paths of the PCL/TCP mixture and cell-laden
hydrogel, respectively. Photograph of the printed
calvarial bone construct (bottom). (b) Scanning
electron microscope images of the printed bone
constructs. (c) Photographs of the printed bone
constructs at day 0 (top) and 5 months (bottom)
after implantation. (d–l) Histological and
immunohistological images of nontreated (d–f),
scaffold only without cells (g–i) and hAFSCs-printed
construct at 5 months after implantation (j–l).
H&E staining (d,g,j), modified tetrachrome
staining (e,h,k) and vWF immunostaining (f,i,l).
Tetrachrome staining: red, mature bone;
blue, osteoid and lining of lacunae. vWF
immunofluorescent image: red, blood vessel.
NB: new bone; PCL/TCP: remaining scaffold.
Red: cells
Green: PCL/TCP
200 µm2 mm
Top view
530 µm
Side view
Day 0
5 months
5 mm
NB
NB
PCL/
TCP
PCL/
TCP
PCL/
TCP
PCL/
TCP
NB
NB
NB
NB
NB
NB
NB
NB
5 mm
100 µm
100 µm
100 µm
a b c
d e f
g h i
j k l
np
g
©
2
01
6
N
at
ur
e
A
m
er
ic
a,
In
c.
A
ll
ri
gh
ts
r
es
er
ve
d.
3 1 6 VOLUME 34 NUMBER 3 MARCH 2016 nature
biotechnology
A r t i c l e s
mouse myoblasts (Table 1) printed in the type II pattern (Fig.
6a,b).
Immediately after printing, the printed structures contained
mus-
cle fiber–like bundles (~400 µm width), supporting PCL pillars
and Pluronic F-127 hydrogel as a temporary structure (Fig. 6c
and
Supplementary Fig. 5a). Notably, the printed cells began
stretch-
ing along the longitudinal axis of the constructs at day 3 in
growth
media (Fig. 6e and Supplementary Fig. 5b) with high cell
viability
(Fig. 6f), and the constructs underwent compaction34, keeping
the
fibers taut during cell growth and differentiation, whereas the
printed
cells without PCL support did not show cellular alignment (Fig.
6d).
After 7 d in differentiation media, muscle-like structures with
aligned
myotubes were observed (Fig. 6g and Supplementary Fig. 5c).
To study whether these structures could mature into functional
muscle in vivo, we implanted 7-d differentiated structures
subcutane-
ously (ectopically) in 14- to 16-week-old nude rats (n = 6). The
dis-
sected distal end of the proximal stump of the common peroneal
nerve
(CPN) was embedded within the constructs to promote
integration
(Fig. 6h,i). Adequate innervation of implanted muscle is
essential to
achieve and maintain muscle function. Our model allowed us to
eval-
uate nerve integration of the implanted muscle construct
independ-
ent of the surrounding muscle tissue. After 2 weeks of
implantation,
the retrieved muscle constructs showed well-organized muscle
fiber
structures (Fig. 6j), the presence of acetylcholine receptor
(AChR)
clusters on the muscle fibers (MHC+ and α-BTX+) (Fig. 6k), as
well
as nerve (neurofilament) contacts with α-BTX+ structures
within the
implants (Fig. 6l), indicating that the printed muscle constructs
were
robust enough to maintain their structural characteristics and
induce
nerve integration in vivo. In addition, vascularization
throughout the
muscle constructs was indicated by endothelial cell marker
expression
(Fig. 6m). To examine muscle function, we performed
electromyogra-
phy to evaluate electrical and neurological activation of the
constructs
2 weeks after implantation. Compound muscle action potential,
which
is evoked by motor nerves and measures muscle function, was
3.6 mV,
compared to 10.7 mV for the control gastrocnemius muscle, and
Red: cells
Green: PCL
Blue: Pluronic F-127
Printing process
Printing
nozzle
Cell-laden
hydrogel
Microchannel
Safranin-O Collagen II
P
ri
n
te
d
e
a
r
H
u
m
a
n
e
a
r
500 µm 500 µm
100 µm 100 µm
500 µm
100 µm
100 µm100 µm100 µm
PCL
Microchannel
10 mm 16 mm
3
2
m
m
Pluronic
F-127
Printed ear
construct
PCL
PCL
a b c d e f
g
Pre-implantation
B
e
n
d
in
g
F
in
a
l
1 month
Collagen IISafranin-O
500 µm
1
m
o
n
th
2
m
o
n
th
s
500 µm 200 µm
200 µm 100 µm
100 µm
PCL
Implantation (month)
*5
4
3
G
A
G
c
o
n
te
n
t
(µ
g
/m
g
)
2
1
0
1 2
5 mm
Cycle 1
Cycle 2
Cycle 3
Cycle 4
N
o
rm
a
liz
e
d
lo
a
d
(
N
/m
2
)
80,000
60,000
40,000
20,000
0 N
o
rm
a
liz
e
d
lo
a
d
(
N
/m
2
)
80,000
60,000
40,000
20,000
0
0 0.5 1.0 1.5 2.0
Extension (mm)
0 0.5 1.0 1.5 2.0
Extension (mm)
Cycle 1
Cycle 2
Cycle 3
Cycle 4
h i
j
k l m
500 µm 500 µm
Alcian Blue
Figure 5 Ear cartilage reconstruction. (a–f) In vitro bioprinted
ear construct. (a) 3D CAD of a human ear. (b) Visualized
motion program used to
print 3D architecture of human ear. The motion program was
generated by using 3D CAD model. Lines of green, blue and red
indicate dispensing
paths of PCL, Pluronic F-127 and cell-laden hydrogel,
respectively. (c) 3D printing process using the integrated organ
printing system (Supplementary
Movie 1). The image shows patterning of a layer of the
construct. (d,e) Photographs of the 3D printed ear cartilage
construct with sacrificial Pluronic
F-127 (d) and after removing sacrificial material by dissolving
with cold medium (e). (f) Safranin-O staining of the 3D printed
cartilage constructs
with microchannels (porous; left) and without microchannels
(nonporous; right) after culture in chondrogenic medium for 5
weeks in vitro.
The constructs with microchannels showed the production of
new cartilaginous matrix throughout the entire constructs,
whereas the constructs
without microchannels showed limited tissue formation due to
limited diffusion of nutrients and oxygen. The staining indicates
the production of GAGs.
(g) Safranin-O staining, Alcian Blue staining and
immunohistochemistry for type II collagen of the 3D printed ear
cartilage constructs after culture
in chondrogenic medium for 5 weeks in vitro. Histological
images of the samples showed the production of a new
cartilaginous matrix within the 3D
printed constructs. The chondrocytes in the newly formed tissue
demonstrated similar morphological characteristics to those in
native cartilage,
with cells located within typical chondrocyte lacunae,
surrounded by cartilaginous matrix. The newly formed matrix
generated in the constructs
stained intensely with Safranin-O and Alcian Blue, showing the
presence of sulfated proteoglycans. Immunohistochemical
staining indicated the
presence of type II collagen in the constructs. Human ear was
used a positive control. (h–m) In vivo bioprinted ear construct.
(h,i) Gross appearance
at 1 month after implantation (h), Safranin-O staining and
collagen type II immunostaining (i) of the retrieved ear
construct at 1 month and 2 months
after implantation. (j) GAG contents of the bioprinted ear
cartilage tissues after 1 and 2 months of implantation. Error
bars, mean ± s.d. (k) Gross
examination of bending testing of the bioprinted ear constructs:
pre-implantation vs. 1-month implantation. (l,m) Stress-strain
curve of pre-implanted
construct (l) and1-month implanted construct under four-cycle
three-point bending test (m).
np
g
©
2
01
6
N
at
ur
e
A
m
er
ic
a,
In
c.
A
ll
ri
gh
ts
r
es
er
ve
d.
nature biotechnology VOLUME 34 NUMBER 3 MARCH
2016 3 1 7
A r t i c l e s
0 mV for the negative controls (subcutaneous tissue), indicating
that
the implanted muscle constructs responded to electrical
stimulation
to an extent consistent with immature, developing muscle (Fig.
6n).
DIScUSSIOn
Bioprinters based on jetting, extrusion and LIFT methods can
deliver
viable cells, biomaterials and macromolecules to generate 3D
tissue
structures. However, in general they are limited in their ability
to gen-
erate large biological constructs with sufficient structural
integrity for
surgical implantation28–30, and the few in vivo studies of
bioprinted
tissue structures tested less complex constructs with low
mechanical
stability35,36. The ITOP can address the limitations of size and
sta-
bility by sequentially printing cell-laden hydrogels with a
synthetic
polymer and a temporary scaffolding, creating tissue constructs
with
the structural integrity needed for surgical implantation. A
computer-
generated 3D tissue model can be converted to a motion
program that
operates and guides the dispensing nozzles to take defined paths
for
delivery of cells and materials. The cell-laden hydrogel protects
cell
viability and promotes growth and expansion, whereas the
adjacent
sacrificial scaffolding provides the initial structural and
architectural
integrity. As the cells anchored three dimensionally within the
hydro-
gel initiate the transition to tissue formation, they start to
secrete their
own matrix, replacing the hydrogel as it slowly degrades over
time.
The system’s modular design enables printing of a wide array of
tissue
constructs. Here we used up to four material repositories, but
many
additional repositories could be installed to print constructs
contain-
ing multiple cell types and biomaterials.
Cell carriers for bioprinting must provide adequate mechanical
support, cell-specific cues and negligible cytotoxicity. As few
such
materials are available37,38, we fulfilled these requirements
with a
mixture of gelatin, fibrinogen, HA and glycerol. …
R E V I E W
T I S S UE E N G I N EE R I N G
Engineering Complex Tissues
Anthony Atala,1 F. Kurtis Kasper,2 Antonios G. Mikos2*
D
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 chal-
lenges 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.
o
w
n
lo
a
t U
N
IV
O
F
H
O
U
S
T
O
N
o
n
A
p
ril 1
4
, 2
0
2
0
h
ttp
://stm
.scie
n
ce
m
a
g
.o
rg
/
a
d
e
d
fro
m
INTRODUCTION
A tremendous clinical need exists for the development of
technologies
to facilitate the regeneration of injured or diseased tissues and
organs.
The unrelenting prevalence of trauma, congenital defects, and
diseases
such as cancer drives the demand, which becomes increasingly
urgent
as the global population expands and ages. A wide variety of
tissues
and organs would benefit from engineering-based repair or
regenera-
tion, from musculoskeletal tissues, such as bone and cartilage,
to entire
organs, including the bladder and liver. The field of tissue
engineering
is at the interface of bioengineering, materials science,
chemistry, biol-
ogy, and medicine, poised to meet these unmet clinical needs
through
the development of new technologies and refinement of existing
ones.
Increasing levels of complexity in the tissues or organs targeted
for
repair generally necessitate a concomitant increase in the
complexity
of the associated tissue engineering approach. For example,
solid
organs, such as the kidney, would require several essential
structures
to restore function, whereas tubular hollow organs, such as the
urethra,
are more easily recreated from basic cells and materials (1).
Similarly,
complexity can be found at the interfaces between tissues, such
as the
transition from cartilage to bone in the osteochondral interface
in ar-
ticulating joints. Such interfaces are receiving increasing
attention as
targets for repair, given the prevalence of injuries affecting
them (2, 3).
Indeed, a complex tissue injury or defect may involve multiple
tissue
types, may be associated with compromised vascularity, or may
be at
risk for infection.
Regardless of the complexity of the target for repair, tissue
engi-
neering strategies generally involve the application of
combinations
of biomaterials, cells, and biologically active factors to effect
tissue for-
mation. This process can involve de novo growth in tissue
culture (in
vitro, ex vivo) or induction of tissue regeneration in vivo at
sites or
under conditions where it otherwise would not occur. Increasing
em-
phasis is being placed on the development of tissue engineering
ap-
proaches within the context of the injury or disease underlying
the
defect. For example, traumatic injuries to the extremities may
involve
multiple tissue types (bone, muscle, vasculature, lymphatics,
nerve),
and biomaterial-based approaches for regeneration are being
devel-
1Wake Forest Institute for Regenerative Medicine, Wake Forest
School of Medicine,
Winston Salem, NC 27157, USA. 2Department of
Bioengineering, Rice University, Houston,
TX 77251, USA.
*To whom correspondence should be addressed. E-mail:
[email protected]
www.ScienceT
oped and evaluated using preclinical composite tissue defect
models
(4). The present review will focus on advances in tissue
engineering
and regenerative medicine that may enable the repair of tissues
with
high complexity, while highlighting bottlenecks to clinical
translation
of these technologies.
TISSUE ENGINEERING SCAFFOLDS
Biomaterials can provide a three-dimensional (3D) structure to
sup-
port tissue growth. These scaffolds define and maintain the
space in
which the target tissue will form and can be tailored to support
the
attachment and proliferation of cells to effect the desired tissue
forma-
tion (5). Ideally, a scaffold should serve as a transient structure
that
will degrade or resorb with time, such that it is replaced with
the tissue
of interest. Advances in biomaterials science combined with
increas-
ing knowledge of extracellular matrix (ECM) biology and the
role of
environmental factors in tissue formation have led to the
development
of scaffolds tailored to provide appropriate structural support
and, in
some cases, biological and mechanical cues to promote tissue
regen-
eration in vivo (6–9). Moreover, scaffold biomaterials can be
modified
to present biologically active signals, including cell-adhesion
peptides
and growth factors, to facilitate cell attachment and to direct
tissue
formation (10–12). In some instances, the scaffolds depend
entirely
on the migration of cells from the body into the defect for tissue
for-
mation to occur, whereas other approaches leverage the
scaffolds for
the transplantation of cell populations to supplement the body.
In either
case, tissue engineering scaffolds seek to mimic key elements of
the
ECM and local microenvironment to support and perhaps induce
tis-
sue formation.
Naturally derived polymeric materials, including polypeptides
(for
example, collagen) and polysaccharides (for example,
hyaluronic acid),
have been explored extensively in the development of tissue
engineering
scaffolds for applications ranging from cartilage repair to
functional
pancreatic replacements (13). Indeed, a key advantage
associated with
naturally derived polymers is the general capacity of these
materials
to support the attachment, proliferation, and differentiation of
cells
(14, 15). Although naturally derived polymers are typically
enzymati-
cally degradable, the kinetics of degradation may not be easily
con-
trolled or predicted. The generally weak mechanical strength
associated
with naturally derived polymers is also a limitation, but it may
be
ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160
160rv12 1
http://stm.sciencemag.org/
R E V I E W
a
t U
N
IV
O
F
H
O
U
S
T
O
N
o
n
A
p
ril 1
4
, 2
0
2
0
h
ttp
://stm
.scie
n
ce
m
a
g
.o
rg
/
D
o
w
n
lo
a
d
e
d
fro
m
improved through the introduction of intermolecular cross-links
(16).
However, cross-linking may prolong the degradation of the
materials
(17). A concern with naturally derived polymeric materials is
the var-
iability inherent in the production of the materials as well as the
po-
tential, albeit small, of the materials to evoke an immune
response.
Synthetic polymers present several key advantages relative to
nat-
urally derived polymers. Synthetic polymers can be
reproducibly manu-
factured with a wide range of mechanical properties and
degradation
kinetics to enable the production of scaffolds with properties
tailored
for a particular application (18). For example, scaffolds
comprising
poly(lactic-co-glycolic acid) (PLGA) have been investigated for
the re-
generation of tissues ranging from blood vessels to bone. Many
syn-
thetic polymers undergo hydrolytic degradation, which may be
more
readily predicted and controlled than enzymatic degradation in
vivo,
given the lack of dependence on local enzyme concentrations.
Certain
classes of synthetic polymers, such as poly(a-hydroxy esters),
produce
acidic products upon degradation (19), which may elicit a
prolonged
inflammatory response (20). Nevertheless, synthetic polymers
them-
selves typically do not carry a risk for inducing an immune
response
owing to a lack of biologically functional domains. This is also
a lim-
itation because synthetic polymers cannot facilitate cell
attachment or
direct phenotypic expression as a natural polymer would.
However, a
variety of synthesis techniques have been developed to
incorporate bi-
ologically active domains into synthetic polymer scaffolds,
thereby
enabling the production of biomimetic scaffolds with a defined
and
tunable composition (21).
Materials derived from the native ECM have also been explored
as
scaffolds. Tissues like the urinary bladder submucosa or the
small in-
testinal submucosa can be processed through mechanical and
chemical
manipulation to remove the cellular components, yielding a
collagen-
rich matrix, in a process called “decellularization.” The
structures and
arrangement of the various ECM proteins in the resulting
acellular
matrices are largely conserved, which results in a general
retention
of the mechanical properties of the original tissue (22).
Moreover, acel-
lular tissue matrices have been shown to support the ingrowth
of cells
and tissues in several applications, without inducing a gross
immune
response (23, 24). Indeed, given the natural origin of the
matrices, the
materials degrade slowly after implantation and are replaced or
re-
modeled with matrix produced by cells (25). Decellularized
matrices
may also be processed to form particulates that can be used
either
alone or in combination with other materials to promote tissue
re-
pair (26).
In other cases, synthetic polymeric scaffolds have been
fabricated
and modified through covalent immobilization of ECM-derived
moi-
eties to control presentation of growth factors, promote cell
attachment,
and enhance directed differentiation of progenitor cell
populations (27).
Additional methods to introduce an ECM-mimetic coating on
scaf-
folds have been explored, including coating of synthetic
polymeric scaf-
folds with naturally derived polymers (collagen or gelatin) and
ceramics
(calcium phosphate) for bone tissue engineering applications
(28, 29).
Loai et al. (30) combined particles of acellular tissue matrices
(urinary
bladder submucosa) with polymeric materials in the fabrication
of
scaffolds with biological activity and tunable properties for
generation
of a vascularized bladder in murine and porcine preclinical
models.
Alternative approaches have seeded cell populations onto
scaffolds
and leveraged culture conditions to drive the differentiation of
cells
and the concomitant production of ECM. Recently, this was
demon-
strated in the production of bone-like ECM (31, 32).
www.ScienceT
To support tissue formation, a tissue engineering scaffold must
present an interconnected porosity or be capable of resorbing as
a func-
tion of time to create space for new tissues. Many fabrication
techniques
have been developed to enable the fabrication of 3D scaffolds
with an
interconnected porosity, ranging from particulate leaching
techniques
to electrospinning methods (33, 34). Although traditional
methods for
scaffold fabrication can enable introduction of interconnected
pores
with a tunable pore size, control of pore architecture has been a
chal-
lenge (35). Three-dimensional printing methods have emerged
to en-
able the fabrication of scaffolds with precise control of the
architecture
throughout the structure (36, 37). Printing techniques have even
been
used to produce scaffolds with controlled gradients in
mechanical prop-
erties and gradients of biologically active factors (38). With this
technol-
ogy, scaffolds with spatially controlled properties have been
created for
the regeneration of complex tissue structures, such as bone and
cartilage
(39–41). However, in some cases, printing of complex scaffolds
of di-
mensions of clinical relevance, such as whole kidneys or livers,
may be
too time-consuming for widespread application.
Tissue engineering scaffolds should support the attachment and
proliferation of cells and the subsequent formation of the tissue
of in-
terest. However, scaffold materials alone often lack the
biological cues
to induce tissue formation. Accordingly, scaffolds are
commonly used
for the presentation or controlled delivery of biologically active
factors
to induce tissue regeneration. Growth factors, ranging from
angiogenic
factors, such as vascular endothelial growth factor, to
osteogenic fac-
tors, such as bone morphogenetic protein-2, have been
incorporated
into scaffolds to promote tissue formation (42, 43). Key
challenges as-
sociated with growth factor delivery in tissue engineering
include not
only selection of the appropriate factor or combination of
factors neces-
sary to induce the desired response but also the dose and
spatiotemporal
delivery needed for proper tissue development (44–46). Another
chal-
lenge has been the maintenance of the biological activity of the
factor,
especially once released from the scaffold.
CELLS IN ENGINEERED COMPLEX TISSUES
Scaffolds used in tissue engineering approaches are commonly
divided
into two general categories, namely, acellular scaffolds, which
depend
on cells in the recipient to effect tissue formation, and cellular
scaf-
folds, which serve as cell transplantation vehicles. In both
cases, the
success of a scaffold technology toward achieving tissue growth
de-
pends largely on the action of the cells. Accordingly, many
current ef-
forts in tissue engineering seek to identify and optimize cell
populations
that can be leveraged for delivery with a scaffold to promote
tissue
repair where it otherwise might not occur.
Autologous cell populations have been of great interest for
appli-
cation in tissue engineering approaches because they have
minimal
risk of rejection. Some early efforts in the field focused on
isolating
primary cells from a biopsy of the tissue or organ of interest
and grow-
ing the cells ex vivo for subsequent introduction back into the
patient
in a tissue engineering therapy. However, a major limitation
encountered
in this area has been the difficulty in expanding cells to
sufficient num-
bers for clinical application. As an alternative, precursor cells
and their
necessary culture conditions for tissue engineering have been
identified
for several tissues and organs. For instance, urothelial cells
have been
grown and expanded in vivo, but traditionally, the expansion
has
been limited (47, 48). Methods have been developed in recent
years to
ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160
160rv12 2
http://stm.sciencemag.org/
C
R
E
D
IT
:
C
.
B
IC
K
E
L/
S
C
IE
N
C
E
T
R
A
N
S
LA
T
IO
N
A
L
M
E
D
IC
IN
E
R E V I E W
a
t U
N
IV
O
F
H
O
U
S
T
h
ttp
://stm
.scie
n
ce
m
a
g
.o
rg
/
D
o
w
n
lo
a
d
e
d
fro
m
identify undifferentiated cells within the urothelial cell
population and
to maintain the undifferentiated state even through the growth
phase
to obtain sufficient numbers of cells for seeding of scaffolds
(47, 49).
These methods have enabled the isolation of urothelial cells
from a
single specimen with dimensions of 1 cm2 and the expansion of
these
cells over a period of 8 weeks to sufficient numbers to cover the
equivalent of a football field (4202 m2) (47).
Although advances in cell culture protocols have allowed for
ex-
pansion of autologous cells to sufficient numbers for clinical
applica-
tion, expansion of primary cells from some tissues and organs,
such as
the pancreas, remains a challenge. Additionally, in some cases,
tissue
engineering strategies rely on autologous cells derived from
diseased
tissues or organs, which may not yield a sufficient number of
normal
cells for clinical application. As a result, tissue engineers seek
to leverage
autologous stem and progenitor cell populations, such as bone
marrow–
derived mesenchymal stem cells (MSCs) and adipose-derived
stem
cells (50). Although MSCs have received a great deal of
attention in
the tissue engineering literature, advances with other adult-
derived stem
cell populations have generally progressed slowly, owing in part
to diffi-
culties associated with maintaining the stem cells in culture or
achiev-
ing attachment of the cells to scaffolds (51). Nevertheless, some
clinical
strategies have involved seeding of patient-derived stem and
progen-
itor cells on biomaterial scaffolds and then leveraging the body
as a
bioreactor for tissue growth. For example, a ceramic scaffold
within
a titanium mesh was seeded with bone marrow as a source of
stem
cells and implanted in the latissimus dorsi of a patient to grow a
man-
dibular replacement ectopically (52).
Other types of stem cells have been included in biomaterial
scaf-
folds for tissue engineering applications. These cell-based
therapies are
beyond the scope of this review, but the reader is referred to
(53–55).
It should be noted that a vascular network is generally needed to
sup-
port the viability of cells throughout a larger, more complex
tissue-
engineered construct. Accordingly, a variety of methods have
been
developed to promote vascularization of tissue-engineered
constructs,
ranging from functionalization of scaffolds with bioactive
factors to
www.ScienceT
development of bioreactor systems to promote vessel formation
ex vivo
(56–58). A detailed discussion of vascularization strategies is
provided
in (59, 60).
CREATING COMPLEX ORGANS
An expansive toolbox of biomaterial- and cell-based
technologies stands
ready to contribute to the production of tissue engineering
solutions
to meet clinical needs. However, immense complexity can be
found
in the various tissues and organs targeted for replacement.
More-
over, the injury or disease driving the need for tissue repair or
replace-
ment can add levels of complexity. A common challenge
encountered
in the development of tissue engineering technologies is the
need to
repair tissue defects or to regenerate organs that have intricate
3D
structures. Furthermore, it is challenging to integrate the
regenerating
tissue with surrounding tissues and to maintain cell viability in
large
constructs.
To better understand the structural design of human tissues and
organs that regenerative medicine attempts to replicate, it may
be
helpful to categorize them into four levels according to their
increasing
complexity: flat tissue structures; tubular structures; hollow,
nontubu-
lar, viscus structures; and complex solid organs (Fig. 1). Within
these
levels of complexity, there are several strategies used to achieve
resto-
ration of function. We also consider the unmet clinical needs in
these
areas and the barriers to translation in existing demonstrations.
Flat structures
Sheets of cells consisting of multiple layers of predominantly
one cell
type represent the simplest architectural subtype in the body.
This
level of tissue complexity is exemplified by the integument
system,
which represents one of the earliest attempts at culturing
autologous
cells in vitro for repair purposes (61). The effects of substantial
loss
of skin surface area are detrimental, as can be seen in burn
patients.
Traditional treatments, such as skin grafts harvested from
unburned
O
N
o
n
A
p
ril 1
4
, 2
0
2
0
Kidney
Bladder
Solid organsHollow, viscus structuresTubular structuresFlat
tissue structures
Cornea
Trachea
Fig. 1. Four structural levels of complex tissues and organs. Hu-
man tissues and organs can be categorized generally into four
tures, such as the bladder; and solid organs, such as the kidney.
The
complexity of a tissue engineering approach generally increases
with
levels of structural complexity: flat tissue structures, such as
the
cornea; tubular structures, such as the trachea; hollow, viscus
struc-
the structure and metabolic functions of the tissue or organ
targeted
for repair.
ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160
160rv12 3
http://stm.sciencemag.org/
R E V I E W
a
t U
N
IV
O
F
H
O
U
S
T
O
N
o
n
A
p
ril 1
4
, 2
0
2
0
h
ttp
://stm
.scie
n
ce
m
a
g
.o
rg
/
D
o
w
n
lo
a
d
e
d
fro
m
portions of the body or allogeneic grafts that provide temporary
pro-
tection, are the current clinical “gold standard.” However, skin
auto-
grafts require harvesting healthy tissue, which may not be
available in
adequate supply in some clinical cases, such as severe burns
affecting
large surface areas. Likewise, skin allografts present a risk of
immuno-
logic rejection and disease transmission.
Accordingly, several technologies are currently being used to
engi-
neer adequate skin for human replacement. Normal skin cells
are being
harvested from the patient in the operating room and are then
sprayed
over the burn area (62, 63). Patient-derived skin cells are also
being ex-
panded and layered ex vivo, with subsequent implantation over
the
burn area, thereby reducing the donor site morbidity required
for burn
coverage (64, 65). Although coverage of a burn with a tissue-
engineered
skin construct can facilitate repair, the size and severity of the
burn play
an important role in determining the ultimate outcome. Large,
full-
thickness burns present a greater clinical challenge for repair
than
small, superficial, partial-thickness burns because blood vessels
and
regenerative epithelial elements of the dermis are destroyed in
full-
thickness wounds. Nevertheless, clinical and commercial
success has
been realized with tissue engineering approaches for functional
repair
of skin in several applications (66, 67). The functional and
cosmetic
outcomes, however, may be improved through ongoing efforts
to re-
capitulate more fully with the tissue-engineered constructs the
com-
plex strata; vascular, lymphatic, and neural elements; pigment;
hair
follicles; and secretory glands of natural skin.
The cornea represents another flat tissue structure that has been
the target of biomaterial-based tissue engineering approaches
for re-
pair in the clinic. It performs a fundamental function in the
refraction
of light for vision and depends on maintenance of its
characteristic
transparency for efficacy. The cornea retains its transparency in
vivo
through maintenance of the shape and organization of a highly
aligned
collagen matrix and active pumping for continuous removal of
aqueous
humor from the tissue. A variety of disorders can disrupt proper
cor-
neal function, and surgical transplantation of donor corneal
tissue has
long served as a clinical standard of treatment for such
conditions.
However, transplantation requires procurement of donor tissue
matched
to the specific requirements of the recipient. To address this
challenge,
biomaterial-based tissue engineering approaches have been
developed
and translated clinically to enable corneal repair without the
need for
human donor tissue (68).
Tubular structures
Regenerative medicine has been able to successfully replicate
many
types of tubular structures, including the urethra, trachea, and
esoph-
agus, in both animals and humans. In general, these structures
consist
of two different cell types arranged as sheets of cells. These
sheets form
into circular, bilayered tissues, which usually serve as means of
trans-
porting fluid or air throughout the body. The tubular tissue
structures
are composed of an inner layer of epithelial or endothelial cells
that
provide a functional barrier and an outer layer of smooth muscle
and
connective tissue to provide support. Whereas the single cell–
layered
skin constructs do not require a complex foundation, tubular
structures
need to incorporate a matrix of synthetic or naturally derived
scaffold-
ing for support.
Tubular, tissue-engineered urethral constructs comprising
synthetic,
biodegradable poly(glycolic acid) (PGA)/PLGA scaffolds
seeded with
autologous urethral muscle and epithelial cells were implanted
into five
patients needing complex urethral reconstruction, and the
engineered
www.ScienceT
urethras remained functional over the clinical follow-up period
of up
to 6 years (69). For blood vessels, autologously derived cells
cultured
from peripheral vein biopsies have been grown in both
biodegradable
collagen and synthetic scaffolds and successfully used as
pulmonary
artery transplants (70). With a different method, vascular access
grafts
for patients with end-stage renal disease requiring hemodialysis
have
been engineered and implanted in humans (71). To accomplish
this,
fibroblasts and endothelial cells were harvested from patients,
ex-
panded ex vivo as sheets of cells, and then wrapped around a
stainless
steel cylinder to allow for fusion. In both situations (70, 71),
clinical
trials have yielded functional implants. Synthetic materials have
also
been seeded with cells to create new blood vessels for
implantation.
For example, human and canine smooth muscle cells were
cultured
on PGA tubular scaffolds, then treated with detergents to
produce
acellular vascular grafts capable of long-term storage. These
vessels
demonstrated patency in both baboon and canine models (72).
Decellularized scaffolds have been used to create tracheas. In
animal models, autologous chondrocytes cultured from cartilage
bi-
opsies were seeded in biodegradable collagen scaffolds and
success-
fully implanted in the pulmonary tree (73). Autologously
derived
chondrocytes have been differentiated from bone marrow MSCs,
and epithelial cells were isolated from a bronchial mucosa
biopsy.
The cells were seeded in the decellularized donor trachea and
cultured
in a bioreactor (74).
Hollow, viscus structures
Like tubular structures, hollow, viscus organs, such as the
bladder and
vagina, generally consist of an inner layer of epithelial-type
cells sur-
rounded by an outer layer of smooth muscle and/or connective
tissue
to provide functional capacity and to anchor the structure in
place.
Whereas tubular structures generally serve as conduits for air or
fluid,
viscus, nontubular organs have wider functional parameters,
higher
metabolic requirements, and more complex intracellular and
inter-
organ interactions. Similar to tubular organs, the biofabrication
pro-
cess depends on a scaffold seeded with at least two different
cell types.
However, the scaffold design is more complex in terms of both
its ar-
chitecture and its predetermined anatomical space limitations,
which
are often patient-specific. In addition, once the engineered
construct is
completed, there are special considerations for implanting the
engi-
neered construct and for connecting it with other tissues and
organs.
Regeneration of bladder tissue has been accomplished in
patients by
using autologously derived urothelial and smooth muscle cells
(75). A
computed tomography scan was performed on patients before
tissue
biopsy to determine the size of the organ to be constructed.
Thus, the
scaffold architecture and size were individualized for each
patient. The
cells were harvested, cultured in vitro, and seeded onto
biodegradable
collagen-PGA composite scaffolds, and the cell-scaffold
constructs
were placed in bioreactors to develop the tissues.
Similarly, a construct for vaginal replacement was synthesized
by
combining PGA fibers with a coating of PLGA, seeding this
scaffold
with rabbit epithelial cells, and culturing within a perfusion
bioreactor.
After implantation as a total vaginal replacement in a rabbit
model,
the biomaterial constructs yielded organs that were successfully
inte-
grated by the host animal and subsequently demonstrated
histological
characteristics similar to natural tissue after several months of
growth
(76). The engineered constructs had to be designed with
determined
specifications that would allow a patent connection with both
the uter-
us superiorly and the introitus opening inferiorly. As a result of
these
ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160
160rv12 4
http://stm.sciencemag.org/
R E V I E W
h
ttp
://stm
.scie
n
ce
m
a
g
.o
r
D
o
w
n
lo
a
d
e
d
fro
m
experiments, human clinical trials for vaginal regeneration are
under
way (COFEPRIS HIM87120BSO).
Solid organs
Solid organs, such as the kidney, heart, pancreas, and liver,
have the
highest level of tissue complexity. The traditional treatment for
end-
stage solid organ disease is either temporary supportive
treatment with
drugs or devices or whole-organ transplantation. Conventional
trans-
plantation allows select patients to regain a functional organ,
yet it is
exceptionally complicated to obtain a histocompatible match
that does
not require the use of immunosuppressive agents. The ultimate
goal of
regenerative medicine is to bioengineer and transplant complex,
solid
organs composed of cells derived from the patient in need. This
ob-
jective, however, presents an exceedingly difficult and
challenging task
given the tissue complexity and developmental process of these
organs.
Complete regeneration of these whole organs requires
incorporation of
extensive vascular networks to support the viability of cells
throughout
the organ as well as precise organization of multiple cell
types—two
challenges traditionally not faced in the biofabrication process
of sim-
pler tissues like the skin. Whereas creation of flat, tubular, and
hollow
viscus organs primarily uses cell-seeded scaffolds, replication
of solid
organ function must incorporate other methods to be successful
because
these organs have complex architecture that extends beyond
simple lay-
ers. To this end, efforts are focused on the development of
biomaterial-
based approaches that incorporate gradients of growth factors,
hybrid
composite materials (77), and use 3D printing methods (37).
Patients with end-stage renal disease suffer major medical
sequelae
secondary to loss of the many physiological duties carried out
by
a
t U
N
IV
O
F
H
O
U
S
T
O
N
o
n
A
p
ril 1
4
, 2
0
2
0
g
/
the kidneys. With complete renal fail-
ure, these patients must undergo me-
chanical dialysis to replace the waste
disposal function of the kidneys and
must be closely monitored for electro-
lyte and …
BIOMEDICINE
3D bioprinting of collagen to rebuild
components of the human heart
A. Lee1*, A. R. Hudson1*, D. J. Shiwarski1, J. W. Tashman1,
T. J. Hinton1, S. Yerneni1,
J. M. Bliley1, P. G. Campbell1,2, A. W. Feinberg1,3†
Collagen is the primary component of the extracellular matrix in
the human body. It has proved
challenging to fabricate collagen scaffolds capable of
replicating the structure and function
of tissues and organs.We present a method to 3D-bioprint
collagen using freeform reversible
embedding of suspended hydrogels (FRESH) to engineer
components of the human heart
at various scales, from capillaries to the full organ. Control of
pH-driven gelation provides
20-micrometer filament resolution, a porous microstructure that
enables rapid cellular
infiltration and microvascularization, and mechanical strength
for fabrication and perfusion
of multiscale vasculature and tri-leaflet valves. We found that
FRESH 3D-bioprinted
hearts accurately reproduce patient-specific anatomical
structure as determined by
micro–computed tomography.Cardiac ventricles printed with
human cardiomyocytes showed
synchronized contractions, directional action potential
propagation, and wall thickening
up to 14% during peak systole.
F
or biofabrication, the goal is to engineer
tissue scaffolds to treat diseases for which
there are limited options, such as end-stage
organ failure. Three-dimensional (3D) bio-
printing has achieved important milestones
including microphysiological devices (1), pat-
terned tissues (2), perfusable vascular-like net-
works (3–5), and implantable scaffolds (6).
However, direct printing of living cells and soft
biomaterials such as extracellular matrix (ECM)
proteins has proved difficult (7). A key obstacle
is the problem of supporting these soft and dy-
namic biological materials during the printing
process to achieve the resolution and fidelity
required to recreate complex 3D structure and
function. Recently, Dvir and colleagues 3D-
printed a decellularized ECM hydrogel into a
heart-like model and showed that human car-
diomyocytes and endothelial cells could be in-
tegrated into the print and were present as
spherical nonaligned cells after 1 day in culture
(8). However, no further structural or functional
analysis was performed.
We report the ability to directly 3D-bioprint
collagen with precise control of composition and
microstructure to engineer tissue components
of the human heart at multiple length scales.
Collagen is an ideal material for biofabrication
because of its critical role in the ECM, where it
provides mechanical strength, enables struc-
tural organization of cell and tissue compart-
ments, and serves as a depot for cell adhesion
and signaling molecules (9). However, it is dif-
ficult to 3D-bioprint complex scaffolds using
collagen in its native unmodified form because
gelation is typically achieved using thermally
driven self-assembly, which is difficult to control.
Researchers have used approaches including
RESEARCH
Lee et al., Science 365, 482–487 (2019) 2 August 2019 1 of 5
1Department of Biomedical Engineering, Carnegie
Mellon University, Pittsburgh, PA 15213, USA. 2Engineering
Research Accelerator, Carnegie Mellon University, Pittsburgh,
PA 15213, USA. 3Department of Materials Science and
Engineering, Carnegie Mellon University, Pittsburgh, PA
15213, USA.
*These authors contributed equally to this work.
†Corresponding author. Email: [email protected]
Fig. 1. High-resolution
3D bioprinting of
collagen using FRESH
v2.0. (A) Time-lapse
sequence of 3D bioprint-
ing of the letters “CMU”
using FRESH v2.0.
(B) Schematic of acidified
collagen solution extruded
into the FRESH support
bath buffered to pH 7.4,
where rapid neutralization
causes gelation and
formation of a collagen
filament. (C and D) Rep-
resentative images of
the gelatin microparticles
in the support bath
for (C) FRESH v1.0 and
(D) v2.0, showing the
decrease in size and
polydispersity. (E) Histo-
gram of Feret diameter
distribution for gelatin
microparticles in FRESH
v1.0 (blue) and v2.0
(red). (F) Mean Feret
diameter of gelatin micro-
particles for FRESH v1.0
and v2.0 [N > 1200, data are means ± SD, ****P < 0.0001
(Student t test)]. (G) Storage (Gʹ) and loss (Gʺ) moduli for
FRESH v1.0 and v2.0 support
baths showing yield stress fluid behavior. (H) A “window-
frame” print construct with single filaments across the middle,
comparing G-code (left), FRESH v1.0
(center), and FRESH v2.0 (right). (I) Single filaments of
collagen showing the variability of the smallest diameter (~250
mm) that can be printed using
FRESH v1.0 (top) compared to relatively smooth filaments 20
to 200 mm in diameter using FRESH v2.0 (bottom). (J)
Collagen filament Feret diameter as a
function of extrusion needle internal diameter for FRESH v2.0,
showing a linear relationship.
o
n
A
u
g
u
st 1
, 2
0
1
9
h
ttp
://scie
n
ce
.scie
n
ce
m
a
g
.o
rg
/
D
o
w
n
lo
a
d
e
d
fro
m
http://science.sciencemag.org/
chemically modifying collagen into an ultraviolet
(UV)–cross-linkable form (10), adjusting pH,
temperature, and collagen concentration to con-
trol gelation and print fidelity (11, 12), and/or
denaturing it into gelatin (13) to make it ther-
moreversible. However, these hydrogels are typ-
ically soft and tend to sag, and they are difficult
to print with high fidelity beyond a few layers
in height. Instead, we developed an approach
that uses rapid pH change to drive collagen self-
assembly within a buffered support material,
enabling us to (i) use chemically unmodified
collagen as a bio-ink, (ii) enhance mechanical
properties by using high collagen concentra-
tions of 12 to 24 mg/ml, and (iii) create complex
structural and functional tissue architectures.
To accomplish this, we developed a substantially
improved second generation of the freeform
reversible embedding of suspended hydrogels
(FRESH v2.0) 3D-bioprinting technique used
in combination with our custom-designed open-
source hardware platforms (fig. S1) (14, 15).
FRESH works by extruding bio-inks within a
thermoreversible support bath composed of
a gelatin microparticle slurry that provides
support during printing and is subsequently
melted away at 37°C (Fig. 1, A and B, and
movie S1) (16).
The original version of the FRESH support
bath, termed FRESH v1.0, consisted of irregularly
shaped microparticles with a mean diameter of
~65 mm created by mechanical blending of a
large gelatin block (Fig. 1C) (16). In FRESH v2.0,
we developed a coacervation approach to gen-
erate gelatin microparticles with (i) uniform
spherical morphology (Fig. 1D), (ii) reduced poly-
dispersity (Fig. 1E), (iii) decreased particle diam-
eter of ~25 mm (Fig. 1F), and (iv) tunable storage
modulus and yield stress (Fig. 1G and fig. S2).
FRESH v2.0 improves resolution with the ability
to precisely generate collagen filaments and ac-
curately reproduce complex G-code, as shown
with a window-frame calibration print (Fig. 1H).
Using FRESH v1.0, the smallest collagen filament
reliably printed was ~250 mm in mean diameter,
with highly variable morphology due to the rela-
tively large and polydisperse gelatin micropar-
ticles (Fig. 1I). In contrast, FRESH v2.0 improves
the resolution by an order of magnitude, with
collagen filaments reliably printed from 200 mm
down to 20 mm in diameter (Fig. 1, I and J).
Filament morphology from solid-like to highly
porous was controlled by tuning the collagen
gelation rate using salt concentration and buffer-
ing capacity of the gelatin support bath (fig. S3).
A pH 7.4 support bath with 50 mM HEPES was
the optimal balance between individual strand
resolution and strand-to-strand adhesion and
was versatile, enabling FRESH printing of mul-
tiple bio-inks with orthogonal gelation mech-
anisms including collagen-based inks, alginate,
fibrinogen, and methacrylated hyaluronic acid in
the same print by adding CaCl2, thrombin, and
UV light exposure (fig. S4) (15).
We first focused on FRESH-printing a sim-
plified model of a small coronary artery–scale
linear tube from collagen type I for perfusion with
a custom-designed pulsatile perfusion system
(Fig. 2A and fig. S5). The linear tube had an inner
diameter of 1.4 mm (fig. S6A) and a wall thick-
ness of ~300 mm (fig. S6B), and was patent and
manifold as determined by dextran perfusion
(fig. S6, C to E, and movie S2) (15). C2C12 cells
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article
TitleABC123 Version X1Leadership Newsletter Article

More Related Content

Similar to TitleABC123 Version X1Leadership Newsletter Article

2014 Acta Biomaterialia
2014 Acta Biomaterialia2014 Acta Biomaterialia
2014 Acta Biomaterialia
Helen Cox
 
Surface characterization and osteoblast like cells culture on collagen modifi...
Surface characterization and osteoblast like cells culture on collagen modifi...Surface characterization and osteoblast like cells culture on collagen modifi...
Surface characterization and osteoblast like cells culture on collagen modifi...
Grupo de Pesquisa em Nanoneurobiofisica
 
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
Alejandro Roldan
 
Thesis Final Report - Shendu Ma
Thesis Final Report - Shendu MaThesis Final Report - Shendu Ma
Thesis Final Report - Shendu Ma
Shendu Marshell Ma
 
nanocellulose based medicine 2017 Rev 3rd Article.pdf
nanocellulose based medicine 2017 Rev 3rd Article.pdfnanocellulose based medicine 2017 Rev 3rd Article.pdf
nanocellulose based medicine 2017 Rev 3rd Article.pdf
tanveercdr
 

Similar to TitleABC123 Version X1Leadership Newsletter Article (20)

kinetics microalgae-3BT
kinetics microalgae-3BTkinetics microalgae-3BT
kinetics microalgae-3BT
 
Bc
BcBc
Bc
 
2014 Acta Biomaterialia
2014 Acta Biomaterialia2014 Acta Biomaterialia
2014 Acta Biomaterialia
 
Surface characterization and osteoblast like cells culture on collagen modifi...
Surface characterization and osteoblast like cells culture on collagen modifi...Surface characterization and osteoblast like cells culture on collagen modifi...
Surface characterization and osteoblast like cells culture on collagen modifi...
 
Writing for publication 2017 writing an abstract
Writing for publication 2017 writing an abstractWriting for publication 2017 writing an abstract
Writing for publication 2017 writing an abstract
 
MICROENCAPSULATION PROJECT
MICROENCAPSULATION PROJECTMICROENCAPSULATION PROJECT
MICROENCAPSULATION PROJECT
 
Chapter1 useofediblefilms
Chapter1 useofediblefilmsChapter1 useofediblefilms
Chapter1 useofediblefilms
 
Resume
ResumeResume
Resume
 
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
Micro - algae cultivation for biofuels: cost, energy balance, environmental i...
 
Layer by-layer decorated herbal cell compatible scaffolds for bone tissue eng...
Layer by-layer decorated herbal cell compatible scaffolds for bone tissue eng...Layer by-layer decorated herbal cell compatible scaffolds for bone tissue eng...
Layer by-layer decorated herbal cell compatible scaffolds for bone tissue eng...
 
Enzyme immobilization an overview on techniques and support
Enzyme immobilization an overview on techniques and supportEnzyme immobilization an overview on techniques and support
Enzyme immobilization an overview on techniques and support
 
Thesis Final Report - Shendu Ma
Thesis Final Report - Shendu MaThesis Final Report - Shendu Ma
Thesis Final Report - Shendu Ma
 
Dentine bonding agents comprising calcium-silicates to support proactive dent...
Dentine bonding agents comprising calcium-silicates to support proactive dent...Dentine bonding agents comprising calcium-silicates to support proactive dent...
Dentine bonding agents comprising calcium-silicates to support proactive dent...
 
nanocellulose based medicine 2017 Rev 3rd Article.pdf
nanocellulose based medicine 2017 Rev 3rd Article.pdfnanocellulose based medicine 2017 Rev 3rd Article.pdf
nanocellulose based medicine 2017 Rev 3rd Article.pdf
 
doppalapudi2014.pdf
doppalapudi2014.pdfdoppalapudi2014.pdf
doppalapudi2014.pdf
 
Isolation, Characterization, and Application of Calcite Producing Bacteria fo...
Isolation, Characterization, and Application of Calcite Producing Bacteria fo...Isolation, Characterization, and Application of Calcite Producing Bacteria fo...
Isolation, Characterization, and Application of Calcite Producing Bacteria fo...
 
Energy-Dispersive X-Ray Spectroscopy Procedure for Analysing Cellular Element...
Energy-Dispersive X-Ray Spectroscopy Procedure for Analysing Cellular Element...Energy-Dispersive X-Ray Spectroscopy Procedure for Analysing Cellular Element...
Energy-Dispersive X-Ray Spectroscopy Procedure for Analysing Cellular Element...
 
History and-applications-of-hydrogels
History and-applications-of-hydrogelsHistory and-applications-of-hydrogels
History and-applications-of-hydrogels
 
Fibrous Scaffold Produced By Rotary Jet Spinning Technique
Fibrous Scaffold Produced By Rotary Jet Spinning TechniqueFibrous Scaffold Produced By Rotary Jet Spinning Technique
Fibrous Scaffold Produced By Rotary Jet Spinning Technique
 
J of mat chem A
J of mat chem AJ of mat chem A
J of mat chem A
 

More from marilynnhoare

1 of 20Male erection is an example of ______. No thought or emotio.docx
1 of 20Male erection is an example of ______. No thought or emotio.docx1 of 20Male erection is an example of ______. No thought or emotio.docx
1 of 20Male erection is an example of ______. No thought or emotio.docx
marilynnhoare
 
1 Final Project I Guidelines and Rubric Overview.docx
1 Final Project I Guidelines and Rubric Overview.docx1 Final Project I Guidelines and Rubric Overview.docx
1 Final Project I Guidelines and Rubric Overview.docx
marilynnhoare
 
1 2 3 Brief Cont.docx
1 2 3 Brief Cont.docx1 2 3 Brief Cont.docx
1 2 3 Brief Cont.docx
marilynnhoare
 
1 A company should evaluate there flow of information and departmen.docx
1 A company should evaluate there flow of information and departmen.docx1 A company should evaluate there flow of information and departmen.docx
1 A company should evaluate there flow of information and departmen.docx
marilynnhoare
 
.Assignment 1 Careers in AccountingAccounting is the study of h.docx
.Assignment 1 Careers in AccountingAccounting is the study of h.docx.Assignment 1 Careers in AccountingAccounting is the study of h.docx
.Assignment 1 Careers in AccountingAccounting is the study of h.docx
marilynnhoare
 

More from marilynnhoare (20)

1 of 20Male erection is an example of ______. No thought or emotio.docx
1 of 20Male erection is an example of ______. No thought or emotio.docx1 of 20Male erection is an example of ______. No thought or emotio.docx
1 of 20Male erection is an example of ______. No thought or emotio.docx
 
1 Describe the six elements of Max Weber’s model of bureaucracy. Ex.docx
1 Describe the six elements of Max Weber’s model of bureaucracy. Ex.docx1 Describe the six elements of Max Weber’s model of bureaucracy. Ex.docx
1 Describe the six elements of Max Weber’s model of bureaucracy. Ex.docx
 
1 Final Project I Guidelines and Rubric Overview.docx
1 Final Project I Guidelines and Rubric Overview.docx1 Final Project I Guidelines and Rubric Overview.docx
1 Final Project I Guidelines and Rubric Overview.docx
 
1 2 3 Brief Cont.docx
1 2 3 Brief Cont.docx1 2 3 Brief Cont.docx
1 2 3 Brief Cont.docx
 
1 and half page please How has deviance been redefined in modern.docx
1 and half page please How has deviance been redefined in modern.docx1 and half page please How has deviance been redefined in modern.docx
1 and half page please How has deviance been redefined in modern.docx
 
1 A company should evaluate there flow of information and departmen.docx
1 A company should evaluate there flow of information and departmen.docx1 A company should evaluate there flow of information and departmen.docx
1 A company should evaluate there flow of information and departmen.docx
 
.        What factors lay behind the upscaling of Hollywood film g.docx
.        What factors lay behind the upscaling of Hollywood film g.docx.        What factors lay behind the upscaling of Hollywood film g.docx
.        What factors lay behind the upscaling of Hollywood film g.docx
 
.  The Daytona International Airport has decided Embry-Riddle is no .docx
.  The Daytona International Airport has decided Embry-Riddle is no .docx.  The Daytona International Airport has decided Embry-Riddle is no .docx
.  The Daytona International Airport has decided Embry-Riddle is no .docx
 
.You will identify a problem with PESTICIDES and you will show t.docx
.You will identify a problem with PESTICIDES and you will show t.docx.You will identify a problem with PESTICIDES and you will show t.docx
.You will identify a problem with PESTICIDES and you will show t.docx
 
.Write a 7-9 page paper with a References List. Your written assig.docx
.Write a 7-9 page paper with a References List. Your written assig.docx.Write a 7-9 page paper with a References List. Your written assig.docx
.Write a 7-9 page paper with a References List. Your written assig.docx
 
.three pages minimum topic Life Long Learning of Engineering i.docx
.three pages minimum topic  Life Long Learning of Engineering i.docx.three pages minimum topic  Life Long Learning of Engineering i.docx
.three pages minimum topic Life Long Learning of Engineering i.docx
 
.Question 1Figures and beliefs of Greek mythology (including de.docx
.Question 1Figures and beliefs of Greek mythology (including de.docx.Question 1Figures and beliefs of Greek mythology (including de.docx
.Question 1Figures and beliefs of Greek mythology (including de.docx
 
.NEED THIS ON 5162015Have to type a paper about victimology3.docx
.NEED THIS ON 5162015Have to type a paper about victimology3.docx.NEED THIS ON 5162015Have to type a paper about victimology3.docx
.NEED THIS ON 5162015Have to type a paper about victimology3.docx
 
.Family, Sex, and Gender1.Acquaintance rape is a major p.docx
.Family, Sex, and Gender1.Acquaintance rape is a major p.docx.Family, Sex, and Gender1.Acquaintance rape is a major p.docx
.Family, Sex, and Gender1.Acquaintance rape is a major p.docx
 
.Assignment 1 Careers in AccountingAccounting is the study of h.docx
.Assignment 1 Careers in AccountingAccounting is the study of h.docx.Assignment 1 Careers in AccountingAccounting is the study of h.docx
.Assignment 1 Careers in AccountingAccounting is the study of h.docx
 
.Arrogance and ignorance may be called the twin causes of the Am.docx
.Arrogance and ignorance may be called the twin causes of the Am.docx.Arrogance and ignorance may be called the twin causes of the Am.docx
.Arrogance and ignorance may be called the twin causes of the Am.docx
 
. Why are energy transformation important for an ecosystems stabili.docx
. Why are energy transformation important for an ecosystems stabili.docx. Why are energy transformation important for an ecosystems stabili.docx
. Why are energy transformation important for an ecosystems stabili.docx
 
... Social Media PaperWrite 700- to 1,050-word paper and addre.docx
... Social Media PaperWrite 700- to 1,050-word paper and addre.docx... Social Media PaperWrite 700- to 1,050-word paper and addre.docx
... Social Media PaperWrite 700- to 1,050-word paper and addre.docx
 
. Briefly discuss why 1 theory is more convincing than the others..docx
. Briefly discuss why 1 theory is more convincing than the others..docx. Briefly discuss why 1 theory is more convincing than the others..docx
. Briefly discuss why 1 theory is more convincing than the others..docx
 
-          1 Describe Taoism. What are its origins and main b.docx
-          1 Describe Taoism. What are its origins and main b.docx-          1 Describe Taoism. What are its origins and main b.docx
-          1 Describe Taoism. What are its origins and main b.docx
 

Recently uploaded

Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
EADTU
 

Recently uploaded (20)

PANDITA RAMABAI- Indian political thought GENDER.pptx
PANDITA RAMABAI- Indian political thought GENDER.pptxPANDITA RAMABAI- Indian political thought GENDER.pptx
PANDITA RAMABAI- Indian political thought GENDER.pptx
 
How to Manage Call for Tendor in Odoo 17
How to Manage Call for Tendor in Odoo 17How to Manage Call for Tendor in Odoo 17
How to Manage Call for Tendor in Odoo 17
 
Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
Transparency, Recognition and the role of eSealing - Ildiko Mazar and Koen No...
 
UGC NET Paper 1 Unit 7 DATA INTERPRETATION.pdf
UGC NET Paper 1 Unit 7 DATA INTERPRETATION.pdfUGC NET Paper 1 Unit 7 DATA INTERPRETATION.pdf
UGC NET Paper 1 Unit 7 DATA INTERPRETATION.pdf
 
80 ĐỀ THI THỬ TUYỂN SINH TIẾNG ANH VÀO 10 SỞ GD – ĐT THÀNH PHỐ HỒ CHÍ MINH NĂ...
80 ĐỀ THI THỬ TUYỂN SINH TIẾNG ANH VÀO 10 SỞ GD – ĐT THÀNH PHỐ HỒ CHÍ MINH NĂ...80 ĐỀ THI THỬ TUYỂN SINH TIẾNG ANH VÀO 10 SỞ GD – ĐT THÀNH PHỐ HỒ CHÍ MINH NĂ...
80 ĐỀ THI THỬ TUYỂN SINH TIẾNG ANH VÀO 10 SỞ GD – ĐT THÀNH PHỐ HỒ CHÍ MINH NĂ...
 
FICTIONAL SALESMAN/SALESMAN SNSW 2024.pdf
FICTIONAL SALESMAN/SALESMAN SNSW 2024.pdfFICTIONAL SALESMAN/SALESMAN SNSW 2024.pdf
FICTIONAL SALESMAN/SALESMAN SNSW 2024.pdf
 
Economic Importance Of Fungi In Food Additives
Economic Importance Of Fungi In Food AdditivesEconomic Importance Of Fungi In Food Additives
Economic Importance Of Fungi In Food Additives
 
Simple, Complex, and Compound Sentences Exercises.pdf
Simple, Complex, and Compound Sentences Exercises.pdfSimple, Complex, and Compound Sentences Exercises.pdf
Simple, Complex, and Compound Sentences Exercises.pdf
 
VAMOS CUIDAR DO NOSSO PLANETA! .
VAMOS CUIDAR DO NOSSO PLANETA!                    .VAMOS CUIDAR DO NOSSO PLANETA!                    .
VAMOS CUIDAR DO NOSSO PLANETA! .
 
Model Attribute _rec_name in the Odoo 17
Model Attribute _rec_name in the Odoo 17Model Attribute _rec_name in the Odoo 17
Model Attribute _rec_name in the Odoo 17
 
FSB Advising Checklist - Orientation 2024
FSB Advising Checklist - Orientation 2024FSB Advising Checklist - Orientation 2024
FSB Advising Checklist - Orientation 2024
 
COMMUNICATING NEGATIVE NEWS - APPROACHES .pptx
COMMUNICATING NEGATIVE NEWS - APPROACHES .pptxCOMMUNICATING NEGATIVE NEWS - APPROACHES .pptx
COMMUNICATING NEGATIVE NEWS - APPROACHES .pptx
 
What is 3 Way Matching Process in Odoo 17.pptx
What is 3 Way Matching Process in Odoo 17.pptxWhat is 3 Way Matching Process in Odoo 17.pptx
What is 3 Way Matching Process in Odoo 17.pptx
 
Accessible Digital Futures project (20/03/2024)
Accessible Digital Futures project (20/03/2024)Accessible Digital Futures project (20/03/2024)
Accessible Digital Futures project (20/03/2024)
 
How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17How to Create and Manage Wizard in Odoo 17
How to Create and Manage Wizard in Odoo 17
 
Details on CBSE Compartment Exam.pptx1111
Details on CBSE Compartment Exam.pptx1111Details on CBSE Compartment Exam.pptx1111
Details on CBSE Compartment Exam.pptx1111
 
How to setup Pycharm environment for Odoo 17.pptx
How to setup Pycharm environment for Odoo 17.pptxHow to setup Pycharm environment for Odoo 17.pptx
How to setup Pycharm environment for Odoo 17.pptx
 
How to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSHow to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POS
 
Graduate Outcomes Presentation Slides - English
Graduate Outcomes Presentation Slides - EnglishGraduate Outcomes Presentation Slides - English
Graduate Outcomes Presentation Slides - English
 
21st_Century_Skills_Framework_Final_Presentation_2.pptx
21st_Century_Skills_Framework_Final_Presentation_2.pptx21st_Century_Skills_Framework_Final_Presentation_2.pptx
21st_Century_Skills_Framework_Final_Presentation_2.pptx
 

TitleABC123 Version X1Leadership Newsletter Article

  • 1. Title ABC/123 Version X 1 Leadership Newsletter Article LDR/300 Version 7 1 University of Phoenix Material Headline [The title or overall description of the newsletter article] Newsletter Tips 1. Interview your individual and research any information necessary. 2. Carefully consider your individual and audience so you offer an article that is engaging and worthwhile. 3. Create a compelling headline and opening to get your audience’s attention. 4. Use clear and concise sentences to generate your individual’s leadership profile. 5. Add any pictures, tables, or graphs that offer insight or clarification for the reader. 6. Review newsletter template websites for assistance. For example, open your web browser and search for either “smore
  • 2. newsletter” or “templates cakemail”. Opening [A statement that describes the individual, their position, and organization] Content [An interpretation of the individual’s leadership style based on the Five-Factor personality model, and an explanation of an incident in which the individual had to solve a difficult problem or situation – including examples] Wrap-Up Conclusion [A statement that summarizes your overall message] Citations (APA) [References for any citations or additional resources]
  • 3. Leadership Newsletter Article Template Copyright © XXXX by University of Phoenix. All rights reserved. Copyright © 2017 by University of Phoenix. All rights reserved. REPORT ◥ BIOMEDICINE Multivascular networks and functional intravascular topologies within biocompatible hydrogels Bagrat Grigoryan1*, Samantha J. Paulsen1*, Daniel C. Corbett2,3*, Daniel W. Sazer1, Chelsea L. Fortin3,4, Alexander J. Zaita1, Paul T. Greenfield1, Nicholas J. Calafat1, John P. Gounley5†, Anderson H. Ta1, Fredrik Johansson2,3, Amanda Randles5, Jessica E. Rosenkrantz6, Jesse D. Louis-Rosenberg6, Peter A. Galie7, Kelly R. Stevens2,3,4‡, Jordan S. Miller1‡ Solid organs transport fluids through distinct vascular networks that are biophysically and biochemically entangled, creating complex three-dimensional (3D) transport regimes that have remained difficult to produce and study.We establish
  • 4. intravascular andmultivascular design freedoms with photopolymerizable hydrogels by using food dye additives as biocompatible yet potent photoabsorbers for projection stereolithography.We demonstrate monolithic transparent hydrogels, produced in minutes, comprising efficient intravascular 3D fluid mixers and functional bicuspid valves.We further elaborate entangled vascular networks from space-filling mathematical topologies and explore the oxygenation and flow of human red blood cells during tidal ventilation and distension of a proximate airway. In addition, we deploy structured biodegradable hydrogel carriers in a rodent model of chronic liver injury to highlight the potential translational utility of this materials innovation. T he morphologies of the circulatory and pulmonary systems are physically and evo- lutionarily entangled (1). In air-breathing vertebrates, these bounded and conserved vessel topologies interact to enable the oxygen-dependent respiration of the entire or- ganism (2–4). To build and interrogate soft hy- drogels containing such prescribed biomimetic and multivascular architectures, we sought to use stereolithography (fig. S1) (5), commonly em- ployed to efficiently convert photoactive liquid resins into structured plastic parts through lo- calized photopolymerization (6, 7). Comparedwith extrusion 3D printing, which deposits voxels in a serial fashion (8–12), photocrosslinking can be highly parallelized via image projection to
  • 5. simultaneously and independently address mil- lions of voxels per time step. In stereolithography, xy resolution isdeterminedby the lightpath,whereas z resolution is dictated by light-attenuating ad- ditives that absorb excess light and confine the polymerization to the desired layer thickness, thereby improving pattern fidelity. In the ab- sence of suitable photoabsorber additives, 3D photopatterning of soft hydrogels has been lim- ited in the types of patterns that can be generated (13–16) or has required complex, expensive, and low-throughput microscopy to enhance z reso- lution via the multiphoton effect (17–19). How- ever, common light-blocking chemicals used for photoresist patterning or plastic part fabrication, such as Sudan I, are not suitable for biomanu- facturing owing to their known genotoxic and carcinogenic characteristics (20). Therefore, we hypothesized that the identification of nontoxic light blockers for projection stereolithography could provide a major advance to the architec- tural richness available for the design and gen- eration of widely used biocompatible hydrogels. Here, we establish that synthetic and natural food dyes, widely used in the food industry, can be applied as potent biocompatible photoabsorbers to enable the stereolithographic production of hydrogels containing intricate and functional vascular architectures. We identified candidate photoabsorbers among food additives whose ab- sorbance spectra encompass visible light wave- lengths that can be used for biocompatible photopolymerization. We initially sought to gen- eratemonolithic hydrogels, composed primarily
  • 6. of water and poly(ethylene glycol) diacrylate [PEGDA, 6 kDa, 20weight% (wt%)], with a 1-mm cylindrical channel oriented perpendicular to the light-projection axis. The fabrication of even this trivial design cannot be easily realized because of the dilute nature of such aqueous formulations, in which the low mass fraction of crosslinkable groups and the requisite longer polymerization times result in inadvertent polymerization and solidification within the narrow void spaces that were designed to be hollowperfusable vasculature (figs. S2 to S4). We determined that aqueous pre-hydrogel solutions containing tartrazine (yellow food coloring FD&C Yellow 5, E102), curcumin (from turmeric), or anthocyanin (from blueberries) can each yield hydrogels with a patent vessel (figs. S2 to S5). In addition to these organic molecules, inorganic gold nanoparticles (50 nm), widely regarded for their biocompatibility and light- attenuating properties (21), also function as an effective photoabsorbing additive to generate perfusable hydrogels (fig. S4). To understand how these photoabsorbers affect the gelation kinetics of photopolymeriz- able hydrogels, we performed photorheological characterization with short-duration light ex- posures, which indicate that these additives cause a dose-dependent delay in the induction of photo- crosslinking (figs. S2D and S4E). Saturating light exposures that extend beyond the reaction termi- nation point demonstrate that suitable additives did not ultimately interfere with the reaction be-
  • 7. cause hydrogels eventually reached an equivalent storage modulus independent of the additive concentration (figs. S2D and S4E). We selected tartrazine as a photoabsorber for further studies. In addition to its low toxicity in humans and broadutility in the food industry (22), we observed that this hydrophilic dye is easily washed out of generated hydrogels (70% elutes within 3 hours for small gels), resulting in nearly transparent constructs suitable for imaging (fig. S2E). Some tartrazine may also be degraded during poly- merization, as tartrazine is known to be sensitive to free radicals (23). Submerging gels in water or saline solution to remove soluble tartrazine also flushes the vascular topology and removes unreacted pre-hydrogel solution. In contrast to tartrazine, curcumin is lipophilic and does not wash out in aqueous solutions; anthocyanin has a peak absorbance far fromour intended 405-nm light source, requiring high concentrations for suitable potency; and gold nanoparticles are physically entrapped and make transmission or fluorescence microscopy impractical (fig. S4E). We assessed whether this materials insight could similarly impart new architectural freedoms tomore-advanced photoactivematerials. Photo- absorber additives are necessary and sufficient to enable vessel construction in thiol-ene step- growth photopolymerization (24) of hydrogels and in a continuous liquid interface production (6) workflow for the generation of hydrogels (fig. S5). We observed strong lamination between ad- jacent fabricated layers and a rapid response of the patterned hydrogel tomechanical deforma- tions (fig. S6). This facile generation of soft
  • 8. hydrogels with patent cylindrical vessels oriented orthogonal to the light-projection axis sug- gests an extensive design flexibility toward the generation of complex vascular topologies, and RESEARCH Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 1 of 7 1Department of Bioengineering, Rice University, Houston, TX 77005, USA. 2Department of Bioengineering, University of Washington, Seattle, WA 98195, USA. 3Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA. 4Department of Pathology, University of Washington, Seattle, WA 98195, USA. 5Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA. 6Nervous System, Somerville, MA 02143, USA. 7Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA. *These authors contributed equally to this work. †Present address: Computational Science and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA. ‡Corresponding author. Email: [email protected] (K.R.S.); [email protected] (J.S.M.) on A pril 14, 2020 http://science.sciencem ag.org/ D
  • 9. ow nloaded from http://science.sciencemag.org/ the optical clarity of resultant hydrogels implies imaging methodologies suitable for characteri- zation and validation of fluid flows. Next, we investigated the ability to form hydrogels containing functional intravascular topologies. We first explored chaotic mixers: intravascular topologies that homogenize fluids as a result of interactions between fluid flow streams and the vessel geometry (25, 26). Where- as macroscale static mixers have found broad utility in industrial processes (27) because of their unparalleled efficiency, translation of intravascular static mixers into microfluidic systems has been difficult to implement, owing to their complex 3D topology. To this end, we generated mono- lithic hydrogels with an integrated static mixer composed of 3D twisted-fin elements (150 mm thick) of alternating chirality inside a 1-mm cylindrical channel. We applied laminar fluid streams to the static mixer at a low Reynolds number (0.002) and observed rapid mixing per unit length (Fig. 1A) and as a function of fin number (fig. S7). The elasticity and compliance of PEG-based hydrogels (fig. S6) enabled the facile generation of a 3D functional bicuspid venous valve (Fig. 1B).We observed that the valve leaflets
  • 10. are dynamic, respond rapidly to pulsatile antero- grade and retrograde flows, and promote the for- mation of stable mirror image vortices in the valve sinuses (Fig. 1B and movie S1) according to established mappings of native tissue (28, 29). Solid organs contain distinct fluid networks that are physically and chemically entangled, providing the rich extracellular milieu that is a hallmark of multicellular life. The ability to fabricate such multivascular topologies within biocompatible and aqueous environments could enable a step change in the fields of biomaterials and tissue engineering. A first objective is the development of an efficient framework to design entangled networks that can provide suitable blueprints for their fabrication within hydrogels. Separate vascular networks must not make a direct fluid connection or they would topolog- ically reduce to a single connected network. We find that mathematical space-filling and fractal topology algorithms provide an efficient para- metric language to design complex vascular blue- prints and a mathematical means to design a second vascular architecture that does not inter- sect the first (Fig. 2). We demonstrate a selection of hydrogels (20 wt %, 6-kDa PEGDA) contain- ing entangled vascular networks based on 3D mathematical algorithms (Fig. 2, A to D): a helix surrounding an axial vessel, 1° and 2° Hilbert curves, a bicontinuous cubic lattice (based on a Schwarz P surface), and a torus entangled with a torus knot. Perfusion with colored dyes and micro-computed tomography (mCT) analysis de- monstrate pattern fidelity, vascular patency, and
  • 11. Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 2 of 7 Fig. 1. Monolithic hydrogels with functional intravascular topologies. (A) Monolithic hydrogels with a perfusable channel containing integrated fin elements of alternating chirality. These static elements rapidly promote fluid dividing and mixing (as shown by fluorescence imaging), consistent with a computational model of flow (scale bars, 1 mm). (B) Hydrogels with a functional 3D bicuspid valve integrated into the vessel wall under anterograde and retrograde flows (scale bars, 500 mm). Particle image velocimetry demonstrates stable mirror image vortices in the sinus region behind open valve leaflets. RESEARCH | REPORT on A pril 14, 2020 http://science.sciencem ag.org/ D ow nloaded from http://science.sciencemag.org/
  • 12. fluidic independence between the two networks (Fig. 2, A to D, and movie S2). We sought to evaluate the efficiency of inter- vascular interstitial transport by measuring the delivery of oxygen from a source vessel to per- fused human red blood cells (RBCs) flowing in an adjacent 3D topology. We tessellated the en- tangled helical topology shown in Fig. 2A along a serpentine path while maintaining the inter- vessel distance at 300 mm (Fig. 2E). Perfusion of deoxygenated RBCs [oxygen partial pressure (PO2) ≤ 40 mmHg; oxygen saturation (SO2) ≤ 45%] into the helical channel during ventilation of the serpentine channel with humidified gaseous oxygen (7 kPa) caused a noticeable color change of RBCs fromdark red at the inlet to bright red at the outlet (Fig. 2, E and F). Collection of perfused RBCs showed significantly higher SO2 and PO2 relative to deoxygenated RBCs loaded at the inlet and negative control gels ventilated with humidified nitrogen gas (Fig. 2G and fig. S8). Although this serpentine-helix design demon- strates the feasibility of intervascular oxygen trans- port between 3D entangled networks, we sought to introduce additional structural features of native distal lung into a bioinspired model of alveolar morphology and oxygen transport. In particular, the realization of 3D hydrogels that contain branching networks and that can sup- port mechanical distension during cyclic ventila-
  • 13. tion of a pooled airway could enable investigations of the performance of lung morphologies derived from native structure (30) and could provide a complete workflow for the development and ex- amination of new functional topologies. Over the past several decades, alveolar morphology has been approximated mathematically as 3D space- filling tessellations of polyhedra (31–34). However, the translation of these ideas into useful blue- prints has remained nontrivial because of the need for efficient space-filling tessellations and an ensheathing vasculature that closely tracks the curvature of the 3D airway topography. Our solution is to calculate a 3D topological offset of the airway (moving each face in its local normal Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 3 of 7 Fig. 2. Entangled vascular networks. (A to D) Adaptations of mathematical space-filling curves to entangled vessel topologies within hydrogels (20 wt % PEGDA, 6 kDa): (A) axial vessel and helix, (B) interpenetrating Hilbert curves, (C) bicontinuous cubic lattice, and (D) torus and (3,10) torus knot (scale bars, 3 mm). (E) Tessellation of the axial vessel and its encompassing helix along a serpentine pathway. The photograph is a top-down view of a fabricated hydrogel with oxygen and RBC delivery to respective vessels. During perfusion, RBCs change color from dark red
  • 14. (at the RBC inlet) to bright red (at the RBC outlet) (scale bar, 3 mm). Boxed regions are magnified in (F) (scale bar, 1 mm). (G) Perfused RBCs were collected at the outlet and quantified for SO2 and PO2. Oxygen flow increased SO2 and PO2 of perfused RBCs compared with deoxygenated RBCs perfused at the inlet (dashed line) and a nitrogen flow negative control (N ≥ 3 replicates, data are mean ± SD, *P < 2 × 10−7 by Student’s t test). RESEARCH | REPORT on A pril 14, 2020 http://science.sciencem ag.org/ D ow nloaded from http://science.sciencemag.org/ Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 4 of 7 Fig. 3. Tidal ventilation and oxygenation in hydrogels with vascularized
  • 15. alveolar model topologies. (A) (Top) Architectural design of an alveolar model topology based on a Weaire-Phelan 3D tessellation and topologic offset to derive an ensheathing vasculature. (Bottom) Cutaway view illustrates the model alveoli (alv.) with a shared airway atrium. Convex (blue) and concave (green) regions of the airway are highlighted. (B) Photograph of a printed hydrogel during RBC perfusion while the air sac was ventilated with O2 (scale bar, 1 mm). (C) Upon airway inflation with oxygen, concave regions of the airway (dashed black circles) squeeze adjacent blood vessels and cause RBC clearance (scale bar, 500 mm). (D) A computational model of airway inflation demonstrates increased displacement at concave regions (dashed yellow circles). (E) Oxygen saturation of RBCs increased with decreasing RBC flow rate (N = 3, data aremean ± SD, *P < 9 × 10−4 by Student’s t test).The dashed line indicates SO2 of deoxygenated RBCs perfused at the inlet. (F) Elaboration of a lung-mimetic design through generative growth of the airway, offset growth of opposing inlet and outlet vascular networks, and population of branch tips with a distal lung subunit. (G) The distal lung subunit is composed of a concave and convex airway ensheathed in vasculature by 3D offset and anisotropicVoronoi tessellation. (H) Photograph of a printed
  • 16. hydrogel containing the distal lung subunit during RBC perfusion while the air sac was ventilated with O2 (scale bar, 1 mm). (I) Threshold view of the area enclosed by the dashed box in (H) demonstrates bidirectional RBC flow during ventilation. (J) Distal lung subunit can stably withstand ventilation for more than 10,000 cycles (24 kPa, 0.5 Hz) and demonstrates RBC sensitivity to ventilation gas (N2 or O2). RESEARCH | REPORT on A pril 14, 2020 http://science.sciencem ag.org/ D ow nloaded from http://science.sciencemag.org/ Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 5 of 7 Fig. 4. Engraftment of functional hepatic hydrogel carriers. (A to C) Albumin promoter activity was enhanced in hydrogel carriers
  • 17. containing hepatic aggregates after implantation in nude mice. Data from all time points for each condition are shown in (B) [N = 4, *P < 0.05 by two- way analysis of variance (ANOVA) followed by Tukey’s post-hoc test]. Cumulative bio- luminescence for each condition is shown in (C) (N = 4, *P < 0.05 by one-way ANOVA followed by Tukey’s post-hoc test). Error bars indicate SEM. GelMA, gelatin methacrylate. (D) Gross images of hydrogels upon resection (scale bars, 5 mm). (E) (Left) Prevascularized hepatic hydrogel carriers are created by seeding endothelial cells (HUVECs) in the vascular network after printing. (Right) Confocal microscopy observations show that hydrogel anchors physically entrap fibrin gel containing the hepatocyte aggregates (Hep) (scale bar, 1 mm). (F) Hepatocytes in prevascularized hepatic hydrogel carriers exhibit albumin promoter activity after implantation in mice with chronic liver injury.Graft sections stainedwithH&Eshowpositioning of hepatic aggregates (black arrows) relative to printed (case, anchor) and nonprinted (fibrin) components of the carrier system (scale bar, 50 mm). (G) Hydrogel carriers are infiltrated with host blood (gross, H&E). Carriers contain aggregates that express the marker cytokeratin-18 (Ck-18) and are in close
  • 18. proximity to Ter-119–positive RBCs (scale bars, 40 mm). RESEARCH | REPORT on A pril 14, 2020 http://science.sciencem ag.org/ D ow nloaded from http://science.sciencemag.org/ direction) and have the new surface serve as the template on which a vascular skeleton is built. With this approach, we developed a bioinspired alveolar model with an ensheathing vasculature from 3D tessellations of the Weaire-Phelan foam topology (35) (Fig. 3 and fig. S9). Although the fundamental units of the Weaire-Phelan foam are convex polyhedra (fig. S9), 3D tessellations can produce a surface containing both convex and concave regions reminiscent of native alveolar air sacs (30) with a shared airway atrium sup- porting alveolar buds (Fig. 3A). We extended the manifold air surface in the normal direc- tion, removed faces, and ensheathed edges in a smoothed polygonal mesh to form a highly branched vascular network (containing 185 vessel
  • 19. segments and 113 fluidic branch points) that encloses the airway and tracks its curvature (fig. S9B). We printed hydrogels (20wt%, 6-kDa PEGDA) patterned with the alveolar model topology at a voxel resolution of 5 pl and a print time of 1 hour (Fig. 3B). Cyclic ventilation of the pooled airway with humidified oxygen gas (10 kPa, 0.5Hz) led to noticeable distension and an apparent change in the curvature of concave airway regions (fig. S9C). Perfusion of deoxygenated RBCs at the blood vessel inlet (10 to 100 mm/min) during cyclic ventilation led to observable compression and RBC clearance from vessels adjacent to con- cave airway regions (Fig. 3, B and C). By observing dilute RBC streams at the early stages of per- fusion, we also discerned that the cyclic com- pression of RBC vessels—actuated by the concave airway regions upon each inflation cycle—acts as switching valves to redirect fluid streams to neighboring vessel segments (movie S3). We im- plemented a simplified 2D computational model of airway inflation (fig. S9D), which predicts an- isotropic distension of the airway and compres- sion of adjacent blood vessels, corresponding to local curvature (fig. S9E). In addition, analysis from a 3D computational model supports an- isotropic distension of the concave regions of the airway during inflation (Fig. 3D). Despite the volume of the alveolar model hydrogel (0.8 ml) being <25% of that of the serpentine-helix model (3.5 ml), we measured similar oxygenation ef- ficiencies for the two designs (Fig. 3E). Our data suggest that branching topology, hydrogel dis- tension, and redirection of fluid streams during
  • 20. ventilation may boost intravascular mixing and allow faster volumetric uptake of oxygen by the well-mixed RBCs. Vascular constriction during breathing has been previously described as an important fluid control mechanism in the mam- malian lung (36), and here we provide a means to actualize these ideas in completely defined and biocompatiblematerials andwithin aqueous environments. To extend this work toward a coherent ap- proximation of scalable lung-mimetic design, we must consolidate the location of the vascular inlet, vascular outlet, and air duct, such that dis- tal lung subunits can be populated on the tips of multiscale branching architecture. Therefore, within a given computational bounding volume,we first derive a branching airway (Fig. 3F). Next, the centerlines of inlet and outlet blood vessel networks are grown 180° opposite each other across and topologically offset from the airway, and the blood vessels traverse down to the tips of all daughter branches. The final step is to pop- ulate the tips of each distal lung with an alveolar unit cell (Fig. 3G andmovie S4) whose ensheath- ing vasculature (containing 354 vessel segments and 233 fluidic branch points) itself is an an- isotropic Voronoi surface tessellation along a topological offset of its local airway (fig. S9, F and G).We found that hydrogels (20 wt %, 6-kDa PEGDA) could withstand more than 10,000 ven- tilation cycles (at 24 kPa and a frequency of 0.5 Hz) over 6 hours during RBC perfusion and while switching the inflow gas between humidi- fied oxygen and humidified nitrogen (Fig. 3, H
  • 21. to J). Color-filtered views of the early stages of RBC perfusion (Fig. 3I) indicate that ventilation promotes RBC mixing and bidirectional flows within selected vessel segments near the mid- point of the distal lung subunit (movie S4). We use our custom stereolithography appa- ratus for tissue engineering (SLATE) to demon- strate production of tissue constructs containing mammalian cells (figs. S1, S10, and S11 andmovie S5). Lung-mimetic architectures can also be pop- ulatedwith human lung fibroblasts in the bulk of the interstitial space and human epithelial-like cells in the airway (fig. S12), which could facilitate the development of a hydrogel analog of a lab- on-a-chip lung design (37). Finally, we subjected primary humanmesenchymal stem cells (hMSCs) to SLATE fabrication (with mixtures of PEGDA and gelatinmethacrylate) and show that the cells within cylindrical fabricated hydrogels remain viable and can undergo osteogenic differentia- tion (fig. S13D). In related multiweek perfusion tissue culture of hMSCs with osteogenic differ- entiation media, osteogenic marker–positive hMSCs were visible throughout the gel (fig. S14). These studies indicate that SLATE fabrication supports rapid biomanufacturing, can maintain the viability of mammalian cell lines, supports the normal function and differentiation of pri- mary human stem cells, and provides an ex- perimentally tractablemeans to explore stem cell differentiation as a function of soluble factor delivery via vascular perfusion. We next sought to establish the utility of this process for fabricating structurally complex and
  • 22. functional tissues for therapeutic transplanta- tion. In particular, the liver is the largest solid organ in the human body, carrying out hundreds of essential tasks in a manner thought to be dependent on its structural topology. We created complex structural features in hydrogel within the expanded design space imparted by SLATE to assemblemultimaterial liver tissues. Bioprinted single-cell tissues andbioprintedhydrogel carriers containing hepatocyte aggregates were fabricated (Fig. 4, A to C). The albumin promoter activity of tissue carriers loaded with aggregates was enhanced by more than a factor of 60 compared with that of implanted tissues containing single cells (Fig. 4, B and C). Furthermore, upon gross examination of tissues after resection, hydrogel carrier tissues appeared to havemore integration with host tissue and blood (Fig. 4D). Despite the improved utility of hepatic aggregates over single cells, aggregate size puts substantial architectural limitations on 3D printing because aggregates are larger in size than our lowest voxel resolution (50 mm). To accommodate these design con- straints, we built a more advanced carrier that can deliver hepatic aggregates within natural fibrin gel, has a vascular compartment that can be seeded with endothelial cells, and incorpo- rates structural hydrogel anchors to physically, rather than chemically, retain the fibrin gel and facilitate remodeling between the graft and host tissue (Fig. 4E and fig. S15). Microchannel net- works were seeded with human umbilical vein endothelial cells (HUVECs) because our previous studies demonstrated that inclusion of endothelial cords improved tissue engraftment (38). We then
  • 23. evaluated whether optimized bioengineered liver tissueswould survive transplantation in a rodent model of chronic liver injury. After 14 days of engraftment in mice with chronic liver injury, hepatic hydrogel carriers exhibited albumin pro- moter activity indicative of surviving functional hepatocytes (Fig. 4F). Immunohistological char- acterization revealed the presence of hepatic aggregates adhered to printed hydrogel com- ponents that stained positively for the marker cytokeratin-18 (Fig. 4, F and G). Further charac- terization through gross examination and higher- magnification images of slides stained with hematoxylin and eosin (H&E) indicated the pres- ence of host blood in explanted tissues. Immu- nostaining using a monoclonal antibody against Ter-119 confirmed the erythroid identity of cells in microvessels adjacent to hepaticmicroaggregates in explanted tissues (Fig. 4G, right). This work provides an approach to address long-standing design limitations in tissue engineering that have hindered progress of preclinical studies. We have identified readily available food dyes that can serve as potent photoabsorbers for bio- compatible and cytocompatible production of hydrogels containing functional vascular to- pologies for studies of fluidmixers, valves, inter- vascular transport, nutrient delivery, and host engraftment.With our stereolithographic process, there is potential for simultaneous and orthog- onal control over tissue architecture and bio- materials for the design of regenerative tissues. REFERENCES AND NOTES
  • 24. 1. R. Monahan-Earley, A. M. Dvorak, W. C. Aird, J. Thromb. Haemost. 11 (suppl. 1), 46–66 (2013). 2. G. R. Scott, J. Exp. Biol. 214, 2455–2462 (2011). 3. E. R. Schachner, J. R. Hutchinson, C. Farmer, PeerJ 1, e60 (2013). 4. C. G. Farmer, Physiology 30, 260–272 (2015). 5. Supplementary figures, as well as materials and methods, are available as supplementary materials. 6. J. R. Tumbleston et al., Science 347, 1349–1352 (2015). 7. B. E. Kelly et al., Science 363, 1075–1079 (2019). 8. J. S. Miller et al., Nat. Mater. 11, 768–774 (2012). 9. T. J. Hinton et al., Sci. Adv. 1, e1500758 (2015). 10. T. Bhattacharjee et al., Sci. Adv. 1, e1500655 (2015). 11. D. B. Kolesky, K. A. Homan, M. A. Skylar-Scott, J. A. Lewis, Proc. Natl. Acad. Sci. U.S.A. 113, 3179–3184 (2016). 12. H.-W. Kang et al., Nat. Biotechnol. 34, 312–319 (2016). Grigoryan et al., Science 364, 458–464 (2019) 3 May 2019 6 of 7 RESEARCH | REPORT on A pril 14, 2020 http://science.sciencem ag.org/ D ow
  • 25. nloaded from http://science.sciencemag.org/ 13. V. Liu Tsang et al., FASEB J. 21, 790–801 (2007). 14. H. Lin et al., Biomaterials 34, 331–339 (2013). 15. J. A. S. Neiman et al., Biotechnol. Bioeng. 112, 777–787 (2015). 16. X. Ma et al., Proc. Natl. Acad. Sci. U.S.A. 113, 2206–2211 (2016). 17. M. S. Hahn, J. S. Miller, J. L. West, Adv. Mater. 18, 2679– 2684 (2006). 18. C. A. DeForest, K. S. Anseth, Nat. Chem. 3, 925–931 (2011). 19. K. A. Heintz et al., Adv. Healthc. Mater. 5, 2153–2160 (2016). 20. T. M. Fonovich, Drug Chem. Toxicol. 36, 343–352 (2013). 21. S. Kumar, J. Aaron, K. Sokolov, Nat. Protoc. 3, 314–320 (2008). 22. L. J. Stevens, J. R. Burgess, M. A. Stochelski, T. Kuczek, Clin. Pediatr. 54, 309–321 (2015). 23. M. Li et al., J. Agric. Food Chem. 62, 12052–12060 (2014). 24. C. A. DeForest, B. D. Polizzotti, K. S. Anseth, Nat. Mater. 8, 659–664 (2009). 25. …
  • 26. 3 1 2 VOLUME 34 NUMBER 3 MARCH 2016 nature biotechnology A r t i c l e s The demand for engineered tissues has risen rapidly owing to the limited availability of donor tissues and organs for transplantation. Despite some initial successes in engineering relatively simple tissues, many challenges remain in developing tissues and organs suitable for clinical translation1,2. Three-dimensional (3D) printing technology shows promise for creating complex composite tissue constructs3–8 through precise placement of cell-laden hydrogels in a layer-by- layer fashion7,9–17. The most commonly used bioprinting systems are based on jetting, extrusion and laser-induced forward transfer (LIFT)6,18,19. The jetting method produces picoliter scale drops with a printing resolution of 20~100 µm. However, because the hydrogel concen- tration is low20–23, the thickness of printed constructs may be lim- ited because of inadequate structural support24. Extrusion methods, which use a syringe and piston system to dispense material through microscale nozzles, can produce more stable 3D cell-laden structures using high concentrations of hydrogels such as alginate, fibrin
  • 27. and Pluronic F-127 (refs. 18,25–27). However, it is difficult to construct large free-form tissue structures owing to inadequate structural integ- rity, mechanical stability and printability28–30. The LIFT method can precisely print cells in relatively small constructs31 but requires rapid gelation of hydrogels to achieve high resolution of the printed pat- terns, resulting in low flow rates. Here we describe a system that deposits cell-laden hydrogels together with synthetic biodegradable polymers that impart mechanical strength, thereby overcoming previous limitations on the size, shape, structural integrity and vascularization of bioprinted tissue con- structs. This was accomplished by designing multidispensing modules for delivering various cell types and polymers in a single construct; by developing an optimized carrier material for delivering cells to discrete locations in the 3D structure in a liquid form; by designing sophisticated nozzle systems with a resolution down to 2 µm for bio- materials and down to 50 µm for cells; by cross-linking cell- laden hydrogels after passage though the nozzle system; by simultaneously printing an outer sacrificial acellular hydrogel mold that is dissolved
  • 28. after the tissue construct acquires enough rigidity to retain its shape; and by creating a lattice of microchannels permissive to nutrient and oxygen diffusion into the printed tissue constructs. These properties, all designed to work in a coordinated manner, make up the ITOP. We demonstrate the printer by fabricating human-scale mandible bone, ear-shaped cartilage and organized skeletal muscle. Evaluation of the characteristics and function of these tissues in vitro and in vivo showed tissue maturation and organization that may be sufficient for translation to patients. RESULTS Design of the ITOP system Multiple cartridges (Fig. 1a and Supplementary Fig. 1) are used to deliver and pattern multiple cell-laden composite hydrogels, support- ing poly(ε-caprolactone) (PCL) polymer and a sacrificial Pluronic F-127 hydrogel (Fig. 1b). The end of each cartridge is connected to a microscale nozzle, and the top is connected to an air pressure controller for precisely controlling the dispensing volume. A heating unit ensures that the PCL remains easily dispensable. A three- axis motorized stage system enables 3D patterning of multiple cells and
  • 29. biomaterials. The system resides in a humidified and temperature- controlled (18 °C) enclosure. The correct shape of a tissue construct is obtained from a human body by processing computed tomography (CT) or magnetic resonance imaging (MRI) data in computer-aided design (CAD) software (Fig. 1c). A custom nozzle motion program is generated by incorporating A 3D bioprinting system to produce human-scale tissue constructs with structural integrity Hyun-Wook Kang, Sang Jin Lee, In Kap Ko, Carlos Kengla, James J Yoo & Anthony Atala A challenge for tissue engineering is producing three- dimensional (3D), vascularized cellular constructs of clinically relevant size, shape and structural integrity. We present an integrated tissue–organ printer (ITOP) that can fabricate stable, human- scale tissue constructs of any shape. Mechanical stability is achieved by printing cell-laden hydrogels together with biodegradable polymers in integrated patterns and anchored on sacrificial hydrogels. The correct shape of the tissue construct is achieved by representing clinical imaging data as a computer model of the anatomical defect and translating the model into a program that controls the motions of the printer nozzles, which dispense cells to discrete locations. The incorporation of microchannels into the tissue constructs facilitates diffusion of nutrients to printed cells, thereby overcoming the diffusion limit of 100–200 mm for cell survival in engineered tissues. We demonstrate capabilities of the ITOP by fabricating mandible and calvarial bone, cartilage
  • 30. and skeletal muscle. Future development of the ITOP is being directed to the production of tissues for human applications and to the building of more complex tissues and solid organs. Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA. Correspondence should be addressed to A.A. ([email protected]). Received 27 July 2015; accepted 19 October 2015; published online 15 February 2016; doi:10.1038/nbt.3413 np g © 2 01 6 N at ur e A m er ic a,
  • 31. In c. A ll ri gh ts r es er ve d. http://dx.doi.org/10.1038/nbt.3413 http://www.nature.com/naturebiotechnology/ nature biotechnology VOLUME 34 NUMBER 3 MARCH 2016 3 1 3 A r t i c l e s the printing pattern in combination with fabrication conditions (for example, scan speed, temperature, material information and air pressure). The ITOP uses a text-based motion program consisting of a command list for XYZ stage movements and air pressure actuation (Supplementary Source Code). The motion program is then transferred to
  • 32. the main computer of the 3D printing system that effects the biofabrication process. The composite hydrogel for cell delivery consisted of gelatin, fibrinogen, hyaluronic acid (HA) and glycerol mixed into DMEM (high glucose). We tested various concentrations of each component to achieve proper print- ing resolution and dispensing uniformity, mechanical properties (before and after cross-linking with thrombin) and cell viability (Supplementary Fig. 2). The optimized concentrations of hydro- gel ingredients and the numbers of cells needed for fabrication of individual tissue constructs are listed in Table 1. Patterning synthetic polymers confers mechanical strength Our first series of studies tested the ability of the ITOP to generate uni- form two-dimensional (2D) and 3D cell patterns of multiple cell types. Using 3T3 fibroblasts labeled with two fluorescent dyes, Dil (red) and DiO (green), we demonstrated delivery of the two populations in a unique 2D pattern (Fig. 2a,b). To create 3D constructs, we combined the fluorescently labeled 3T3 fibroblasts in composite hydrogels with supporting PCL and printed them in two patterns—type I (Fig. 2c) and type II (Fig. 2f). These patterns differ in the placement of PCL and thus in the mechanical strength of the printed construct. The type I pattern creates multiple PCL frames in each layer throughout
  • 33. the con- struct, and places cells and gel materials in between the frames. The type II pattern consists of cell-laden hydrogel and porous structures, surrounded by a PCL framework on the outer layers and corners of each layer, thus protecting the contents from external load. Type I constructs (Fig. 2d,e) maintained a more stable structure than type II constructs (Fig. 2g,h), owing to the abundance of uniformly distrib- uted PCL frames. Therefore, we used the type I pattern to fabricate mandible bone and ear-shaped cartilage structures and the type II pattern to print organized skeletal muscle constructs. Next, we produced 3D structures by placing either type I or type II pat- terns of cell-laden hydrogel and PCL (~130 µm for type I and ~250 µm wide for type II). The microchannels (type I: 500 × 300 µm2; type II: 650 × 450 µm2), formed by the PCL patterns, were designed to maxi- mize diffusion of nutrients and oxygen. In addition, we used Pluronic F-127 hydrogel as a sacrificial outer layer to support the 3D architecture of the dispensed cell-laden structures before crosslinking. After cross- linking of fibrinogen using thrombin, the uncross-linked components
  • 34. (gelatin, HA, glycerol and Pluronic F-127) were washed out. To determine cell viability during printing, we examined survival of 3T3 fibroblasts at 60 min (day 0), 3 d and 6 d after printing. Live/dead cell assays showed ≥95% cell viability on day 0, which was maintained through days 3 and 6 (Fig. 2i). Cell proliferation, assessed using the AlamarBlue assay system, increased over a 15-d period, similar to the proliferation of control cells encapsulated in a fibrin construct (Fig. 2j). These data indicate that the optimized composite hydrogel system maintained cell viability during the printing process and provided a favorable microenvironment for cell proliferation. Mandible bone reconstruction To demonstrate construction of a human-sized bone structure, we fab- ricated a mandible fragment in a size and shape similar to what would be needed for facial reconstruction after traumatic injury (Fig. 3). The cell type used was human amniotic fluid–derived stem cells Main computer 3-axis stage controller Pressure
  • 35. controller Multi- cartridge module 3D printed construct PCL (gray) A: cell A (red) B: cell B (green) S: sacrificial material Closed chamber Medical imaging (CT, MRI) Visualized motion program 3D printing process 3D bioprinted tissue product 10 mm DICOM format STL format Text-based command list 3D printed construct PCL
  • 36. Cell A Cell B XYZ stage Heating unit PCL A B S Po re (m icr oc ha nn el) 3D CAD model a b c Figure 1 ITOP system. (a) The ITOP system consists of three major units: (i) 3-axis stage/ controller, (ii) dispensing module including multi-cartridge and pneumatic pressure
  • 37. controller and (iii) a closed acrylic chamber with temperature controller and humidifier. (b) Illustration of basic patterning of 3D architecture including multiple cell-laden hydrogels and supporting PCL polymer. (c) CAD/CAM process for automated printing of 3D shape imitating target tissue or organ. A 3D CAD model developed from medical image data generates a visualized motion program, which includes instructions for XYZ stage movements and actuating pneumatic pressure to achieve 3D printing. Table 1 Preparation of the cell-laden composite hydrogels for 3D bioprinted tissue constructs Composite hydrogel Gelatin Fibrinogen HA Glycerol Cell type & density Cell viability Remark Bone (type I) 35 mg/ml 20 mg/ml 3 mg/ml 10% v/v Human AFSCs, 5 × 106 cells/ml 91 ± 2% (day 1) Figs. 3 and 4 Cartilage (Type I) 45 mg/ml 30 mg/ml 3 mg/ml 10% v/v Rabbit ear chondrocytes, 40 × 106 cells/ml 91 ± 8% (day 1) Fig. 5 Skeletal muscle (type II) 35 mg/ml 20 mg/ml 3 mg/ml 10% v/v Mouse C2C12 myoblasts, 3 × 106 cells/ml 97 ± 6% (day 1) Fig. 6 np g © 2 01 6
  • 39. biotechnology A r t i c l e s (hAFSCs), which can give rise to osteogenic lineages in appropriate media32,33. Mandible bone defects have an arbitrary shape. We used data from a CT scan of a human mandible defect in combination with Mimics software (Materialise, Leuven, Belgium) to produce a CAD model of the defect shape, with dimensions of 3.6 cm × 3.0 cm × 1.6 cm (Fig. 3a). A text-based command motion program, gener- ated from the CAD model with custom CAM software, determined the required dispensing paths of cell-laden hydrogel, a mixture of PCL and tricalcium phosphate (TCP), and Pluronic F127 (Fig. 3b). PCL/TCP and hAFSCs mixed with the composite hydrogel (Table 1) were printed in a type I pattern with a Pluronic F127 temporary sup- port (Fig. 3c). At 1 d of culture, cell viability in the printed bone structures was 91 ± 2% (n = 3, Table 1), confirming that the printing process did not adversely affect cell viability. After induction of osteo- genic differentiation using an established protocol32,33 for 28 d (n = 5, Fig. 3d), we stained the structures with Alizarin Red S; staining at the
  • 40. surface of the 3D bone structures indicated calcium deposition in the hAFSC-laden hydrogel (Fig. 3e). 3D constructs before differentiation showed no Alizarin Red S staining (data not shown). Calvarial bone reconstruction To study maturation of the bioprinted bone in vivo, we fabricated rat calvarial bone constructs in a circular shape (8 mm diameter × 1.2 mm thickness) with hAFSCs (Fig. 4a,b and Supplementary Fig. 3), cultured them in osteogenic media for 10 d, implanted them in a calvarial bone defect region of Sprague Dawley rats (n = 4) and analyzed them 5 months after implantation (Fig. 4c). Type l Pore 1 mm 1 mm 1 mm 1 mm Type l Top Type l - top Type ll PCL
  • 41. PCL Cell-laden hydrogel Cell-laden hydrogel Side Top Side Type ll Type ll – top 500 µm 120 100 80 60 40 20 0 0 3 6 Culture time (d)
  • 43. in te n si ty Control Printed 500 µm 1 3 7 15 a b c d e f g h i j Figure 2 2D/3D patterning using the ITOP system. (a,b) 2D patterning of ‘WFIRM’ characters written by cell-laden hydrogels through the integrated organ printing. Microscopic (a) and fluorescent images (b) of ‘WFIRM’ characters, which were produced using cells labeled with Dil and DiO. (c–h) Two basic types of 3D patterning: type I pattern (c–e) and type II pattern (f–h). Two types of 3D patterning, including cell-A (red), cell-B (blue) and PCL (green), were fabricated by the integrated organ printing (c,f); photographs (d,g) and fluorescent image (e,h) of the 3D printed
  • 44. patterns. (i) Cell viability was over 95% on day 0 and then maintained on days 3 and 6 (n = 3). (j) Cell proliferation results showed that the number of cells continuously increased over a 15-d period, and no significant differences between the control and the printed constructs were observed (n = 5). Error bars, mean ± s.d. Bony defect Red: cells Green: PCL Blue: Pluronic F-127 Pluronic F-127 (sacrificial material) Printing nozzle Pore PCL Cell-laden hydrogel 30 mm 3 6 m m P
  • 45. o re PCL Cell-laden hydrogel 1 mm a b c d e Figure 3 Mandible bone reconstruction. (a) 3D CAD model recognized a mandible bony defect from human CT image data. (b) Visualized motion program was generated to construct a 3D architecture of the mandible bone defect using CAM software developed by our laboratory. Lines of green, blue and red colors indicate the dispensing paths of PCL, Pluronic F-127 and cell-laden hydrogel, respectively. (c) 3D printing process using the integrated organ printing system. The image shows patterning of a layer of the construct. (d) Photograph of the 3D printed mandible bone defect construct, which was cultured in osteogenic medium for 28 d. (e) Osteogenic differentiation of hAFSCs in the printed construct was confirmed by Alizarin Red S staining, indicating calcium deposition. np g
  • 47. d. nature biotechnology VOLUME 34 NUMBER 3 MARCH 2016 3 1 5 A r t i c l e s The bioprinted constructs showed newly formed vascularized bone tissue through- out the implants, including the central por- tion, with no necrosis (Fig. 4j), whereas the untreated defect and scaffold-only treated control groups showed fibrotic tis- sue ingrowth (Fig. 4d) and minimal bone tissue formation restricted to the periphery of the implant (Fig. 4g), respectively. The modified Tetrachrome staining confirmed mature bone (red) and osteoid (blue) for- mation (Fig. 4e,h,k). Von Willebrand factor (vWF) immunostaining showed large blood vessel formation within newly formed bone tissue throughout the bioprinted bone con- structs, including the central portion (Fig. 4l), whereas the nontreated (Fig. 4f) and scaffold-only (Fig. 4i) groups had only limited vascu- larization restricted to the periphery of the implant. Ear cartilage reconstruction Next, we tested the ability of the ITOP to fabricate tissue constructs of complex shape by making human-sized external ears, as the frame- work of an auricle consists of a single piece of cartilage with a
  • 48. com- plicated geometry of ridges. A CT image of an ear (Fig. 5a) was used to develop a motion program (Fig. 5b) to print a chondrocyte- laden hydrogel, PCL and Pluronic F-127. Using rabbit ear chondrocytes (passages 3 and 4) mixed with the composite hydrogel (Table 1), we fabricated human ear–shaped cartilage constructs with dimensions of 3.2 cm × 1.6 cm × 0.9 cm (Fig. 5c–e) in the type I pattern. Cell viabil- ity was 91 ± 8% at 1 d after printing (n = 3, Table 1). After 5 weeks in the culture medium, the constructs were stained with Safranin-O and showed production of a new cartilaginous matrix (Fig. 5f). The constructs with microchannels showed enhanced tissue forma- tion as evidenced by the production of new viable cartilaginous matrix throughout the entire ear constructs. In contrast, the constructs without microchannels showed only limited tissue formation restricted to the peripheral region, likely owing to the diffusion limits of nutrients and oxygen. The cells in the newly formed tissues demonstrated similar morphological characteristics to those in native ear cartilage, with cells located within typical chondrocyte lacunae, surrounded by a cartilaginous matrix (Fig. 5g). Native human ear tissue served as a positive control. To determine whether the printed ear constructs would mature
  • 49. in vivo, we implanted them in the dorsal subcutaneous space of athymic mice and retrieved them 1 and 2 months after implantation (n = 4). The shape was well maintained, with substantial cartilage formation upon gross examination (Fig. 5h). Histological analysis showed the for- mation of cartilage tissue (Fig. 5i). The glycosaminoglycan (GAG) content (2.7 ± 0.2 µg/mg at 1 month and 4.2 ± 0.3 µg/mg at 2 months) increased over time, reaching 20% of that of native ear GAG content (Fig. 5j). Vascularization of the printed constructs in the outer region was suggested by endothelial cell marker expression at 1 and 2 months after implantation (Supplementary Fig. 4). The inner regions were avascular (Supplementary Fig. 4), as in native cartilage, but the car- tilage cells were viable, suggesting adequate nutrient diffusion during development. Biomechanical analyses (n = 4, Fig. 5k) showed that maturation in vivo strengthened the tissue constructs, resulting in a higher normalized load during bending compared with pre- implant constructs. In addition, resilience, measured by the ∆Load%, was tested by repeated bending and relaxation cycles. Resilience between the repeated bending cycles was much higher in the constructs
  • 50. implanted for 1 month (Fig. 5m and Supplementary Table 1) than in the con- structs before implantation (Fig. 5l) . These results demonstrate the generation of ear-shaped cartilage with resilience properties similar to those of native cartilage (rabbit ear) (Supplementary Table 1). Skeletal muscle reconstruction Finally, we applied the ITOP to fabricate an organized soft tissue—a 3D muscle construct 15 mm × 5 mm × 1 mm in dimension containing Figure 4 Calvarial bone reconstruction. (a) Visualized motion program (top) used to print a 3D architecture of calvarial bone construct. Green and red color lines indicate the dispensing paths of the PCL/TCP mixture and cell-laden hydrogel, respectively. Photograph of the printed calvarial bone construct (bottom). (b) Scanning electron microscope images of the printed bone constructs. (c) Photographs of the printed bone constructs at day 0 (top) and 5 months (bottom) after implantation. (d–l) Histological and immunohistological images of nontreated (d–f), scaffold only without cells (g–i) and hAFSCs-printed construct at 5 months after implantation (j–l). H&E staining (d,g,j), modified tetrachrome staining (e,h,k) and vWF immunostaining (f,i,l). Tetrachrome staining: red, mature bone; blue, osteoid and lining of lacunae. vWF immunofluorescent image: red, blood vessel. NB: new bone; PCL/TCP: remaining scaffold.
  • 51. Red: cells Green: PCL/TCP 200 µm2 mm Top view 530 µm Side view Day 0 5 months 5 mm NB NB PCL/ TCP PCL/ TCP PCL/ TCP PCL/ TCP NB NB
  • 52. NB NB NB NB NB NB 5 mm 100 µm 100 µm 100 µm a b c d e f g h i j k l np g © 2 01 6 N
  • 53. at ur e A m er ic a, In c. A ll ri gh ts r es er ve d. 3 1 6 VOLUME 34 NUMBER 3 MARCH 2016 nature biotechnology
  • 54. A r t i c l e s mouse myoblasts (Table 1) printed in the type II pattern (Fig. 6a,b). Immediately after printing, the printed structures contained mus- cle fiber–like bundles (~400 µm width), supporting PCL pillars and Pluronic F-127 hydrogel as a temporary structure (Fig. 6c and Supplementary Fig. 5a). Notably, the printed cells began stretch- ing along the longitudinal axis of the constructs at day 3 in growth media (Fig. 6e and Supplementary Fig. 5b) with high cell viability (Fig. 6f), and the constructs underwent compaction34, keeping the fibers taut during cell growth and differentiation, whereas the printed cells without PCL support did not show cellular alignment (Fig. 6d). After 7 d in differentiation media, muscle-like structures with aligned myotubes were observed (Fig. 6g and Supplementary Fig. 5c). To study whether these structures could mature into functional muscle in vivo, we implanted 7-d differentiated structures subcutane- ously (ectopically) in 14- to 16-week-old nude rats (n = 6). The dis- sected distal end of the proximal stump of the common peroneal nerve (CPN) was embedded within the constructs to promote integration (Fig. 6h,i). Adequate innervation of implanted muscle is
  • 55. essential to achieve and maintain muscle function. Our model allowed us to eval- uate nerve integration of the implanted muscle construct independ- ent of the surrounding muscle tissue. After 2 weeks of implantation, the retrieved muscle constructs showed well-organized muscle fiber structures (Fig. 6j), the presence of acetylcholine receptor (AChR) clusters on the muscle fibers (MHC+ and α-BTX+) (Fig. 6k), as well as nerve (neurofilament) contacts with α-BTX+ structures within the implants (Fig. 6l), indicating that the printed muscle constructs were robust enough to maintain their structural characteristics and induce nerve integration in vivo. In addition, vascularization throughout the muscle constructs was indicated by endothelial cell marker expression (Fig. 6m). To examine muscle function, we performed electromyogra- phy to evaluate electrical and neurological activation of the constructs 2 weeks after implantation. Compound muscle action potential, which is evoked by motor nerves and measures muscle function, was 3.6 mV, compared to 10.7 mV for the control gastrocnemius muscle, and Red: cells Green: PCL Blue: Pluronic F-127
  • 57. 500 µm 500 µm 100 µm 100 µm 500 µm 100 µm 100 µm100 µm100 µm PCL Microchannel 10 mm 16 mm 3 2 m m Pluronic F-127 Printed ear construct PCL PCL a b c d e f g Pre-implantation
  • 59. 500 µm 200 µm 200 µm 100 µm 100 µm PCL Implantation (month) *5 4 3 G A G c o n te n t (µ g /m g )
  • 60. 2 1 0 1 2 5 mm Cycle 1 Cycle 2 Cycle 3 Cycle 4 N o rm a liz e d lo a d ( N /m 2 )
  • 62. 0 0 0.5 1.0 1.5 2.0 Extension (mm) 0 0.5 1.0 1.5 2.0 Extension (mm) Cycle 1 Cycle 2 Cycle 3 Cycle 4 h i j k l m 500 µm 500 µm Alcian Blue Figure 5 Ear cartilage reconstruction. (a–f) In vitro bioprinted ear construct. (a) 3D CAD of a human ear. (b) Visualized motion program used to print 3D architecture of human ear. The motion program was generated by using 3D CAD model. Lines of green, blue and red indicate dispensing paths of PCL, Pluronic F-127 and cell-laden hydrogel, respectively. (c) 3D printing process using the integrated organ printing system (Supplementary Movie 1). The image shows patterning of a layer of the construct. (d,e) Photographs of the 3D printed ear cartilage
  • 63. construct with sacrificial Pluronic F-127 (d) and after removing sacrificial material by dissolving with cold medium (e). (f) Safranin-O staining of the 3D printed cartilage constructs with microchannels (porous; left) and without microchannels (nonporous; right) after culture in chondrogenic medium for 5 weeks in vitro. The constructs with microchannels showed the production of new cartilaginous matrix throughout the entire constructs, whereas the constructs without microchannels showed limited tissue formation due to limited diffusion of nutrients and oxygen. The staining indicates the production of GAGs. (g) Safranin-O staining, Alcian Blue staining and immunohistochemistry for type II collagen of the 3D printed ear cartilage constructs after culture in chondrogenic medium for 5 weeks in vitro. Histological images of the samples showed the production of a new cartilaginous matrix within the 3D printed constructs. The chondrocytes in the newly formed tissue demonstrated similar morphological characteristics to those in native cartilage, with cells located within typical chondrocyte lacunae, surrounded by cartilaginous matrix. The newly formed matrix generated in the constructs stained intensely with Safranin-O and Alcian Blue, showing the presence of sulfated proteoglycans. Immunohistochemical staining indicated the presence of type II collagen in the constructs. Human ear was used a positive control. (h–m) In vivo bioprinted ear construct. (h,i) Gross appearance at 1 month after implantation (h), Safranin-O staining and collagen type II immunostaining (i) of the retrieved ear construct at 1 month and 2 months after implantation. (j) GAG contents of the bioprinted ear cartilage tissues after 1 and 2 months of implantation. Error
  • 64. bars, mean ± s.d. (k) Gross examination of bending testing of the bioprinted ear constructs: pre-implantation vs. 1-month implantation. (l,m) Stress-strain curve of pre-implanted construct (l) and1-month implanted construct under four-cycle three-point bending test (m). np g © 2 01 6 N at ur e A m er ic a, In c. A ll ri
  • 65. gh ts r es er ve d. nature biotechnology VOLUME 34 NUMBER 3 MARCH 2016 3 1 7 A r t i c l e s 0 mV for the negative controls (subcutaneous tissue), indicating that the implanted muscle constructs responded to electrical stimulation to an extent consistent with immature, developing muscle (Fig. 6n). DIScUSSIOn Bioprinters based on jetting, extrusion and LIFT methods can deliver viable cells, biomaterials and macromolecules to generate 3D tissue structures. However, in general they are limited in their ability to gen- erate large biological constructs with sufficient structural integrity for surgical implantation28–30, and the few in vivo studies of
  • 66. bioprinted tissue structures tested less complex constructs with low mechanical stability35,36. The ITOP can address the limitations of size and sta- bility by sequentially printing cell-laden hydrogels with a synthetic polymer and a temporary scaffolding, creating tissue constructs with the structural integrity needed for surgical implantation. A computer- generated 3D tissue model can be converted to a motion program that operates and guides the dispensing nozzles to take defined paths for delivery of cells and materials. The cell-laden hydrogel protects cell viability and promotes growth and expansion, whereas the adjacent sacrificial scaffolding provides the initial structural and architectural integrity. As the cells anchored three dimensionally within the hydro- gel initiate the transition to tissue formation, they start to secrete their own matrix, replacing the hydrogel as it slowly degrades over time. The system’s modular design enables printing of a wide array of tissue constructs. Here we used up to four material repositories, but many additional repositories could be installed to print constructs contain- ing multiple cell types and biomaterials.
  • 67. Cell carriers for bioprinting must provide adequate mechanical support, cell-specific cues and negligible cytotoxicity. As few such materials are available37,38, we fulfilled these requirements with a mixture of gelatin, fibrinogen, HA and glycerol. … R E V I E W T I S S UE E N G I N EE R I N G Engineering Complex Tissues Anthony Atala,1 F. Kurtis Kasper,2 Antonios G. Mikos2* D 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 chal- lenges 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. o w n lo
  • 69. ://stm .scie n ce m a g .o rg / a d e d fro m INTRODUCTION A tremendous clinical need exists for the development of technologies to facilitate the regeneration of injured or diseased tissues and organs. The unrelenting prevalence of trauma, congenital defects, and diseases such as cancer drives the demand, which becomes increasingly urgent as the global population expands and ages. A wide variety of tissues and organs would benefit from engineering-based repair or
  • 70. regenera- tion, from musculoskeletal tissues, such as bone and cartilage, to entire organs, including the bladder and liver. The field of tissue engineering is at the interface of bioengineering, materials science, chemistry, biol- ogy, and medicine, poised to meet these unmet clinical needs through the development of new technologies and refinement of existing ones. Increasing levels of complexity in the tissues or organs targeted for repair generally necessitate a concomitant increase in the complexity of the associated tissue engineering approach. For example, solid organs, such as the kidney, would require several essential structures to restore function, whereas tubular hollow organs, such as the urethra, are more easily recreated from basic cells and materials (1). Similarly, complexity can be found at the interfaces between tissues, such as the transition from cartilage to bone in the osteochondral interface in ar- ticulating joints. Such interfaces are receiving increasing attention as targets for repair, given the prevalence of injuries affecting them (2, 3). Indeed, a complex tissue injury or defect may involve multiple tissue types, may be associated with compromised vascularity, or may be at
  • 71. risk for infection. Regardless of the complexity of the target for repair, tissue engi- neering strategies generally involve the application of combinations of biomaterials, cells, and biologically active factors to effect tissue for- mation. This process can involve de novo growth in tissue culture (in vitro, ex vivo) or induction of tissue regeneration in vivo at sites or under conditions where it otherwise would not occur. Increasing em- phasis is being placed on the development of tissue engineering ap- proaches within the context of the injury or disease underlying the defect. For example, traumatic injuries to the extremities may involve multiple tissue types (bone, muscle, vasculature, lymphatics, nerve), and biomaterial-based approaches for regeneration are being devel- 1Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157, USA. 2Department of Bioengineering, Rice University, Houston, TX 77251, USA. *To whom correspondence should be addressed. E-mail: [email protected] www.ScienceT oped and evaluated using preclinical composite tissue defect models (4). The present review will focus on advances in tissue engineering
  • 72. and regenerative medicine that may enable the repair of tissues with high complexity, while highlighting bottlenecks to clinical translation of these technologies. TISSUE ENGINEERING SCAFFOLDS Biomaterials can provide a three-dimensional (3D) structure to sup- port tissue growth. These scaffolds define and maintain the space in which the target tissue will form and can be tailored to support the attachment and proliferation of cells to effect the desired tissue forma- tion (5). Ideally, a scaffold should serve as a transient structure that will degrade or resorb with time, such that it is replaced with the tissue of interest. Advances in biomaterials science combined with increas- ing knowledge of extracellular matrix (ECM) biology and the role of environmental factors in tissue formation have led to the development of scaffolds tailored to provide appropriate structural support and, in some cases, biological and mechanical cues to promote tissue regen- eration in vivo (6–9). Moreover, scaffold biomaterials can be modified to present biologically active signals, including cell-adhesion peptides and growth factors, to facilitate cell attachment and to direct tissue formation (10–12). In some instances, the scaffolds depend
  • 73. entirely on the migration of cells from the body into the defect for tissue for- mation to occur, whereas other approaches leverage the scaffolds for the transplantation of cell populations to supplement the body. In either case, tissue engineering scaffolds seek to mimic key elements of the ECM and local microenvironment to support and perhaps induce tis- sue formation. Naturally derived polymeric materials, including polypeptides (for example, collagen) and polysaccharides (for example, hyaluronic acid), have been explored extensively in the development of tissue engineering scaffolds for applications ranging from cartilage repair to functional pancreatic replacements (13). Indeed, a key advantage associated with naturally derived polymers is the general capacity of these materials to support the attachment, proliferation, and differentiation of cells (14, 15). Although naturally derived polymers are typically enzymati- cally degradable, the kinetics of degradation may not be easily con- trolled or predicted. The generally weak mechanical strength associated with naturally derived polymers is also a limitation, but it may be ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160
  • 74. 160rv12 1 http://stm.sciencemag.org/ R E V I E W a t U N IV O F H O U S T O N o n A p ril 1 4 , 2
  • 76. improved through the introduction of intermolecular cross-links (16). However, cross-linking may prolong the degradation of the materials (17). A concern with naturally derived polymeric materials is the var- iability inherent in the production of the materials as well as the po- tential, albeit small, of the materials to evoke an immune response. Synthetic polymers present several key advantages relative to nat- urally derived polymers. Synthetic polymers can be reproducibly manu- factured with a wide range of mechanical properties and degradation kinetics to enable the production of scaffolds with properties tailored for a particular application (18). For example, scaffolds comprising poly(lactic-co-glycolic acid) (PLGA) have been investigated for the re- generation of tissues ranging from blood vessels to bone. Many syn- thetic polymers undergo hydrolytic degradation, which may be more readily predicted and controlled than enzymatic degradation in vivo, given the lack of dependence on local enzyme concentrations. Certain classes of synthetic polymers, such as poly(a-hydroxy esters), produce acidic products upon degradation (19), which may elicit a prolonged
  • 77. inflammatory response (20). Nevertheless, synthetic polymers them- selves typically do not carry a risk for inducing an immune response owing to a lack of biologically functional domains. This is also a lim- itation because synthetic polymers cannot facilitate cell attachment or direct phenotypic expression as a natural polymer would. However, a variety of synthesis techniques have been developed to incorporate bi- ologically active domains into synthetic polymer scaffolds, thereby enabling the production of biomimetic scaffolds with a defined and tunable composition (21). Materials derived from the native ECM have also been explored as scaffolds. Tissues like the urinary bladder submucosa or the small in- testinal submucosa can be processed through mechanical and chemical manipulation to remove the cellular components, yielding a collagen- rich matrix, in a process called “decellularization.” The structures and arrangement of the various ECM proteins in the resulting acellular matrices are largely conserved, which results in a general retention of the mechanical properties of the original tissue (22). Moreover, acel- lular tissue matrices have been shown to support the ingrowth of cells
  • 78. and tissues in several applications, without inducing a gross immune response (23, 24). Indeed, given the natural origin of the matrices, the materials degrade slowly after implantation and are replaced or re- modeled with matrix produced by cells (25). Decellularized matrices may also be processed to form particulates that can be used either alone or in combination with other materials to promote tissue re- pair (26). In other cases, synthetic polymeric scaffolds have been fabricated and modified through covalent immobilization of ECM-derived moi- eties to control presentation of growth factors, promote cell attachment, and enhance directed differentiation of progenitor cell populations (27). Additional methods to introduce an ECM-mimetic coating on scaf- folds have been explored, including coating of synthetic polymeric scaf- folds with naturally derived polymers (collagen or gelatin) and ceramics (calcium phosphate) for bone tissue engineering applications (28, 29). Loai et al. (30) combined particles of acellular tissue matrices (urinary bladder submucosa) with polymeric materials in the fabrication of scaffolds with biological activity and tunable properties for generation
  • 79. of a vascularized bladder in murine and porcine preclinical models. Alternative approaches have seeded cell populations onto scaffolds and leveraged culture conditions to drive the differentiation of cells and the concomitant production of ECM. Recently, this was demon- strated in the production of bone-like ECM (31, 32). www.ScienceT To support tissue formation, a tissue engineering scaffold must present an interconnected porosity or be capable of resorbing as a func- tion of time to create space for new tissues. Many fabrication techniques have been developed to enable the fabrication of 3D scaffolds with an interconnected porosity, ranging from particulate leaching techniques to electrospinning methods (33, 34). Although traditional methods for scaffold fabrication can enable introduction of interconnected pores with a tunable pore size, control of pore architecture has been a chal- lenge (35). Three-dimensional printing methods have emerged to en- able the fabrication of scaffolds with precise control of the architecture throughout the structure (36, 37). Printing techniques have even been used to produce scaffolds with controlled gradients in mechanical prop- erties and gradients of biologically active factors (38). With this technol- ogy, scaffolds with spatially controlled properties have been
  • 80. created for the regeneration of complex tissue structures, such as bone and cartilage (39–41). However, in some cases, printing of complex scaffolds of di- mensions of clinical relevance, such as whole kidneys or livers, may be too time-consuming for widespread application. Tissue engineering scaffolds should support the attachment and proliferation of cells and the subsequent formation of the tissue of in- terest. However, scaffold materials alone often lack the biological cues to induce tissue formation. Accordingly, scaffolds are commonly used for the presentation or controlled delivery of biologically active factors to induce tissue regeneration. Growth factors, ranging from angiogenic factors, such as vascular endothelial growth factor, to osteogenic fac- tors, such as bone morphogenetic protein-2, have been incorporated into scaffolds to promote tissue formation (42, 43). Key challenges as- sociated with growth factor delivery in tissue engineering include not only selection of the appropriate factor or combination of factors neces- sary to induce the desired response but also the dose and spatiotemporal delivery needed for proper tissue development (44–46). Another chal- lenge has been the maintenance of the biological activity of the factor,
  • 81. especially once released from the scaffold. CELLS IN ENGINEERED COMPLEX TISSUES Scaffolds used in tissue engineering approaches are commonly divided into two general categories, namely, acellular scaffolds, which depend on cells in the recipient to effect tissue formation, and cellular scaf- folds, which serve as cell transplantation vehicles. In both cases, the success of a scaffold technology toward achieving tissue growth de- pends largely on the action of the cells. Accordingly, many current ef- forts in tissue engineering seek to identify and optimize cell populations that can be leveraged for delivery with a scaffold to promote tissue repair where it otherwise might not occur. Autologous cell populations have been of great interest for appli- cation in tissue engineering approaches because they have minimal risk of rejection. Some early efforts in the field focused on isolating primary cells from a biopsy of the tissue or organ of interest and grow- ing the cells ex vivo for subsequent introduction back into the patient in a tissue engineering therapy. However, a major limitation encountered in this area has been the difficulty in expanding cells to sufficient num- bers for clinical application. As an alternative, precursor cells
  • 82. and their necessary culture conditions for tissue engineering have been identified for several tissues and organs. For instance, urothelial cells have been grown and expanded in vivo, but traditionally, the expansion has been limited (47, 48). Methods have been developed in recent years to ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160 160rv12 2 http://stm.sciencemag.org/ C R E D IT : C . B IC K E L/ S C IE N C E T R
  • 83. A N S LA T IO N A L M E D IC IN E R E V I E W a t U N IV O F H O U S
  • 84. T h ttp ://stm .scie n ce m a g .o rg / D o w n lo a d e d fro m identify undifferentiated cells within the urothelial cell population and to maintain the undifferentiated state even through the growth
  • 85. phase to obtain sufficient numbers of cells for seeding of scaffolds (47, 49). These methods have enabled the isolation of urothelial cells from a single specimen with dimensions of 1 cm2 and the expansion of these cells over a period of 8 weeks to sufficient numbers to cover the equivalent of a football field (4202 m2) (47). Although advances in cell culture protocols have allowed for ex- pansion of autologous cells to sufficient numbers for clinical applica- tion, expansion of primary cells from some tissues and organs, such as the pancreas, remains a challenge. Additionally, in some cases, tissue engineering strategies rely on autologous cells derived from diseased tissues or organs, which may not yield a sufficient number of normal cells for clinical application. As a result, tissue engineers seek to leverage autologous stem and progenitor cell populations, such as bone marrow– derived mesenchymal stem cells (MSCs) and adipose-derived stem cells (50). Although MSCs have received a great deal of attention in the tissue engineering literature, advances with other adult- derived stem cell populations have generally progressed slowly, owing in part to diffi- culties associated with maintaining the stem cells in culture or achiev-
  • 86. ing attachment of the cells to scaffolds (51). Nevertheless, some clinical strategies have involved seeding of patient-derived stem and progen- itor cells on biomaterial scaffolds and then leveraging the body as a bioreactor for tissue growth. For example, a ceramic scaffold within a titanium mesh was seeded with bone marrow as a source of stem cells and implanted in the latissimus dorsi of a patient to grow a man- dibular replacement ectopically (52). Other types of stem cells have been included in biomaterial scaf- folds for tissue engineering applications. These cell-based therapies are beyond the scope of this review, but the reader is referred to (53–55). It should be noted that a vascular network is generally needed to sup- port the viability of cells throughout a larger, more complex tissue- engineered construct. Accordingly, a variety of methods have been developed to promote vascularization of tissue-engineered constructs, ranging from functionalization of scaffolds with bioactive factors to www.ScienceT development of bioreactor systems to promote vessel formation ex vivo (56–58). A detailed discussion of vascularization strategies is provided in (59, 60).
  • 87. CREATING COMPLEX ORGANS An expansive toolbox of biomaterial- and cell-based technologies stands ready to contribute to the production of tissue engineering solutions to meet clinical needs. However, immense complexity can be found in the various tissues and organs targeted for replacement. More- over, the injury or disease driving the need for tissue repair or replace- ment can add levels of complexity. A common challenge encountered in the development of tissue engineering technologies is the need to repair tissue defects or to regenerate organs that have intricate 3D structures. Furthermore, it is challenging to integrate the regenerating tissue with surrounding tissues and to maintain cell viability in large constructs. To better understand the structural design of human tissues and organs that regenerative medicine attempts to replicate, it may be helpful to categorize them into four levels according to their increasing complexity: flat tissue structures; tubular structures; hollow, nontubu- lar, viscus structures; and complex solid organs (Fig. 1). Within these levels of complexity, there are several strategies used to achieve resto- ration of function. We also consider the unmet clinical needs in
  • 88. these areas and the barriers to translation in existing demonstrations. Flat structures Sheets of cells consisting of multiple layers of predominantly one cell type represent the simplest architectural subtype in the body. This level of tissue complexity is exemplified by the integument system, which represents one of the earliest attempts at culturing autologous cells in vitro for repair purposes (61). The effects of substantial loss of skin surface area are detrimental, as can be seen in burn patients. Traditional treatments, such as skin grafts harvested from unburned O N o n A p ril 1 4 , 2 0 2 0 Kidney Bladder
  • 89. Solid organsHollow, viscus structuresTubular structuresFlat tissue structures Cornea Trachea Fig. 1. Four structural levels of complex tissues and organs. Hu- man tissues and organs can be categorized generally into four tures, such as the bladder; and solid organs, such as the kidney. The complexity of a tissue engineering approach generally increases with levels of structural complexity: flat tissue structures, such as the cornea; tubular structures, such as the trachea; hollow, viscus struc- the structure and metabolic functions of the tissue or organ targeted for repair. ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160 160rv12 3 http://stm.sciencemag.org/ R E V I E W a t U N IV O
  • 91. m a g .o rg / D o w n lo a d e d fro m portions of the body or allogeneic grafts that provide temporary pro- tection, are the current clinical “gold standard.” However, skin auto- grafts require harvesting healthy tissue, which may not be available in adequate supply in some clinical cases, such as severe burns affecting large surface areas. Likewise, skin allografts present a risk of immuno- logic rejection and disease transmission. Accordingly, several technologies are currently being used to
  • 92. engi- neer adequate skin for human replacement. Normal skin cells are being harvested from the patient in the operating room and are then sprayed over the burn area (62, 63). Patient-derived skin cells are also being ex- panded and layered ex vivo, with subsequent implantation over the burn area, thereby reducing the donor site morbidity required for burn coverage (64, 65). Although coverage of a burn with a tissue- engineered skin construct can facilitate repair, the size and severity of the burn play an important role in determining the ultimate outcome. Large, full- thickness burns present a greater clinical challenge for repair than small, superficial, partial-thickness burns because blood vessels and regenerative epithelial elements of the dermis are destroyed in full- thickness wounds. Nevertheless, clinical and commercial success has been realized with tissue engineering approaches for functional repair of skin in several applications (66, 67). The functional and cosmetic outcomes, however, may be improved through ongoing efforts to re- capitulate more fully with the tissue-engineered constructs the com- plex strata; vascular, lymphatic, and neural elements; pigment; hair follicles; and secretory glands of natural skin.
  • 93. The cornea represents another flat tissue structure that has been the target of biomaterial-based tissue engineering approaches for re- pair in the clinic. It performs a fundamental function in the refraction of light for vision and depends on maintenance of its characteristic transparency for efficacy. The cornea retains its transparency in vivo through maintenance of the shape and organization of a highly aligned collagen matrix and active pumping for continuous removal of aqueous humor from the tissue. A variety of disorders can disrupt proper cor- neal function, and surgical transplantation of donor corneal tissue has long served as a clinical standard of treatment for such conditions. However, transplantation requires procurement of donor tissue matched to the specific requirements of the recipient. To address this challenge, biomaterial-based tissue engineering approaches have been developed and translated clinically to enable corneal repair without the need for human donor tissue (68). Tubular structures Regenerative medicine has been able to successfully replicate many types of tubular structures, including the urethra, trachea, and esoph- agus, in both animals and humans. In general, these structures
  • 94. consist of two different cell types arranged as sheets of cells. These sheets form into circular, bilayered tissues, which usually serve as means of trans- porting fluid or air throughout the body. The tubular tissue structures are composed of an inner layer of epithelial or endothelial cells that provide a functional barrier and an outer layer of smooth muscle and connective tissue to provide support. Whereas the single cell– layered skin constructs do not require a complex foundation, tubular structures need to incorporate a matrix of synthetic or naturally derived scaffold- ing for support. Tubular, tissue-engineered urethral constructs comprising synthetic, biodegradable poly(glycolic acid) (PGA)/PLGA scaffolds seeded with autologous urethral muscle and epithelial cells were implanted into five patients needing complex urethral reconstruction, and the engineered www.ScienceT urethras remained functional over the clinical follow-up period of up to 6 years (69). For blood vessels, autologously derived cells cultured from peripheral vein biopsies have been grown in both biodegradable collagen and synthetic scaffolds and successfully used as pulmonary
  • 95. artery transplants (70). With a different method, vascular access grafts for patients with end-stage renal disease requiring hemodialysis have been engineered and implanted in humans (71). To accomplish this, fibroblasts and endothelial cells were harvested from patients, ex- panded ex vivo as sheets of cells, and then wrapped around a stainless steel cylinder to allow for fusion. In both situations (70, 71), clinical trials have yielded functional implants. Synthetic materials have also been seeded with cells to create new blood vessels for implantation. For example, human and canine smooth muscle cells were cultured on PGA tubular scaffolds, then treated with detergents to produce acellular vascular grafts capable of long-term storage. These vessels demonstrated patency in both baboon and canine models (72). Decellularized scaffolds have been used to create tracheas. In animal models, autologous chondrocytes cultured from cartilage bi- opsies were seeded in biodegradable collagen scaffolds and success- fully implanted in the pulmonary tree (73). Autologously derived chondrocytes have been differentiated from bone marrow MSCs, and epithelial cells were isolated from a bronchial mucosa biopsy. The cells were seeded in the decellularized donor trachea and cultured
  • 96. in a bioreactor (74). Hollow, viscus structures Like tubular structures, hollow, viscus organs, such as the bladder and vagina, generally consist of an inner layer of epithelial-type cells sur- rounded by an outer layer of smooth muscle and/or connective tissue to provide functional capacity and to anchor the structure in place. Whereas tubular structures generally serve as conduits for air or fluid, viscus, nontubular organs have wider functional parameters, higher metabolic requirements, and more complex intracellular and inter- organ interactions. Similar to tubular organs, the biofabrication pro- cess depends on a scaffold seeded with at least two different cell types. However, the scaffold design is more complex in terms of both its ar- chitecture and its predetermined anatomical space limitations, which are often patient-specific. In addition, once the engineered construct is completed, there are special considerations for implanting the engi- neered construct and for connecting it with other tissues and organs. Regeneration of bladder tissue has been accomplished in patients by using autologously derived urothelial and smooth muscle cells (75). A computed tomography scan was performed on patients before
  • 97. tissue biopsy to determine the size of the organ to be constructed. Thus, the scaffold architecture and size were individualized for each patient. The cells were harvested, cultured in vitro, and seeded onto biodegradable collagen-PGA composite scaffolds, and the cell-scaffold constructs were placed in bioreactors to develop the tissues. Similarly, a construct for vaginal replacement was synthesized by combining PGA fibers with a coating of PLGA, seeding this scaffold with rabbit epithelial cells, and culturing within a perfusion bioreactor. After implantation as a total vaginal replacement in a rabbit model, the biomaterial constructs yielded organs that were successfully inte- grated by the host animal and subsequently demonstrated histological characteristics similar to natural tissue after several months of growth (76). The engineered constructs had to be designed with determined specifications that would allow a patent connection with both the uter- us superiorly and the introitus opening inferiorly. As a result of these ranslationalMedicine.org 14 November 2012 Vol 4 Issue 160 160rv12 4 http://stm.sciencemag.org/
  • 98. R E V I E W h ttp ://stm .scie n ce m a g .o r D o w n lo a d e d fro m experiments, human clinical trials for vaginal regeneration are under
  • 99. way (COFEPRIS HIM87120BSO). Solid organs Solid organs, such as the kidney, heart, pancreas, and liver, have the highest level of tissue complexity. The traditional treatment for end- stage solid organ disease is either temporary supportive treatment with drugs or devices or whole-organ transplantation. Conventional trans- plantation allows select patients to regain a functional organ, yet it is exceptionally complicated to obtain a histocompatible match that does not require the use of immunosuppressive agents. The ultimate goal of regenerative medicine is to bioengineer and transplant complex, solid organs composed of cells derived from the patient in need. This ob- jective, however, presents an exceedingly difficult and challenging task given the tissue complexity and developmental process of these organs. Complete regeneration of these whole organs requires incorporation of extensive vascular networks to support the viability of cells throughout the organ as well as precise organization of multiple cell types—two challenges traditionally not faced in the biofabrication process of sim- pler tissues like the skin. Whereas creation of flat, tubular, and hollow viscus organs primarily uses cell-seeded scaffolds, replication
  • 100. of solid organ function must incorporate other methods to be successful because these organs have complex architecture that extends beyond simple lay- ers. To this end, efforts are focused on the development of biomaterial- based approaches that incorporate gradients of growth factors, hybrid composite materials (77), and use 3D printing methods (37). Patients with end-stage renal disease suffer major medical sequelae secondary to loss of the many physiological duties carried out by a t U N IV O F H O U S T O N o
  • 101. n A p ril 1 4 , 2 0 2 0 g / the kidneys. With complete renal fail- ure, these patients must undergo me- chanical dialysis to replace the waste disposal function of the kidneys and must be closely monitored for electro- lyte and … BIOMEDICINE 3D bioprinting of collagen to rebuild components of the human heart A. Lee1*, A. R. Hudson1*, D. J. Shiwarski1, J. W. Tashman1, T. J. Hinton1, S. Yerneni1, J. M. Bliley1, P. G. Campbell1,2, A. W. Feinberg1,3† Collagen is the primary component of the extracellular matrix in the human body. It has proved
  • 102. challenging to fabricate collagen scaffolds capable of replicating the structure and function of tissues and organs.We present a method to 3D-bioprint collagen using freeform reversible embedding of suspended hydrogels (FRESH) to engineer components of the human heart at various scales, from capillaries to the full organ. Control of pH-driven gelation provides 20-micrometer filament resolution, a porous microstructure that enables rapid cellular infiltration and microvascularization, and mechanical strength for fabrication and perfusion of multiscale vasculature and tri-leaflet valves. We found that FRESH 3D-bioprinted hearts accurately reproduce patient-specific anatomical structure as determined by micro–computed tomography.Cardiac ventricles printed with human cardiomyocytes showed synchronized contractions, directional action potential propagation, and wall thickening up to 14% during peak systole. F or biofabrication, the goal is to engineer tissue scaffolds to treat diseases for which there are limited options, such as end-stage organ failure. Three-dimensional (3D) bio- printing has achieved important milestones including microphysiological devices (1), pat- terned tissues (2), perfusable vascular-like net- works (3–5), and implantable scaffolds (6). However, direct printing of living cells and soft biomaterials such as extracellular matrix (ECM) proteins has proved difficult (7). A key obstacle
  • 103. is the problem of supporting these soft and dy- namic biological materials during the printing process to achieve the resolution and fidelity required to recreate complex 3D structure and function. Recently, Dvir and colleagues 3D- printed a decellularized ECM hydrogel into a heart-like model and showed that human car- diomyocytes and endothelial cells could be in- tegrated into the print and were present as spherical nonaligned cells after 1 day in culture (8). However, no further structural or functional analysis was performed. We report the ability to directly 3D-bioprint collagen with precise control of composition and microstructure to engineer tissue components of the human heart at multiple length scales. Collagen is an ideal material for biofabrication because of its critical role in the ECM, where it provides mechanical strength, enables struc- tural organization of cell and tissue compart- ments, and serves as a depot for cell adhesion and signaling molecules (9). However, it is dif- ficult to 3D-bioprint complex scaffolds using collagen in its native unmodified form because gelation is typically achieved using thermally driven self-assembly, which is difficult to control. Researchers have used approaches including RESEARCH Lee et al., Science 365, 482–487 (2019) 2 August 2019 1 of 5 1Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA. 2Engineering
  • 104. Research Accelerator, Carnegie Mellon University, Pittsburgh, PA 15213, USA. 3Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA. *These authors contributed equally to this work. †Corresponding author. Email: [email protected] Fig. 1. High-resolution 3D bioprinting of collagen using FRESH v2.0. (A) Time-lapse sequence of 3D bioprint- ing of the letters “CMU” using FRESH v2.0. (B) Schematic of acidified collagen solution extruded into the FRESH support bath buffered to pH 7.4, where rapid neutralization causes gelation and formation of a collagen filament. (C and D) Rep- resentative images of the gelatin microparticles in the support bath for (C) FRESH v1.0 and (D) v2.0, showing the decrease in size and polydispersity. (E) Histo- gram of Feret diameter distribution for gelatin microparticles in FRESH v1.0 (blue) and v2.0 (red). (F) Mean Feret diameter of gelatin micro- particles for FRESH v1.0 and v2.0 [N > 1200, data are means ± SD, ****P < 0.0001
  • 105. (Student t test)]. (G) Storage (Gʹ) and loss (Gʺ) moduli for FRESH v1.0 and v2.0 support baths showing yield stress fluid behavior. (H) A “window- frame” print construct with single filaments across the middle, comparing G-code (left), FRESH v1.0 (center), and FRESH v2.0 (right). (I) Single filaments of collagen showing the variability of the smallest diameter (~250 mm) that can be printed using FRESH v1.0 (top) compared to relatively smooth filaments 20 to 200 mm in diameter using FRESH v2.0 (bottom). (J) Collagen filament Feret diameter as a function of extrusion needle internal diameter for FRESH v2.0, showing a linear relationship. o n A u g u st 1 , 2 0 1 9 h ttp ://scie n ce
  • 106. .scie n ce m a g .o rg / D o w n lo a d e d fro m http://science.sciencemag.org/ chemically modifying collagen into an ultraviolet (UV)–cross-linkable form (10), adjusting pH, temperature, and collagen concentration to con- trol gelation and print fidelity (11, 12), and/or denaturing it into gelatin (13) to make it ther-
  • 107. moreversible. However, these hydrogels are typ- ically soft and tend to sag, and they are difficult to print with high fidelity beyond a few layers in height. Instead, we developed an approach that uses rapid pH change to drive collagen self- assembly within a buffered support material, enabling us to (i) use chemically unmodified collagen as a bio-ink, (ii) enhance mechanical properties by using high collagen concentra- tions of 12 to 24 mg/ml, and (iii) create complex structural and functional tissue architectures. To accomplish this, we developed a substantially improved second generation of the freeform reversible embedding of suspended hydrogels (FRESH v2.0) 3D-bioprinting technique used in combination with our custom-designed open- source hardware platforms (fig. S1) (14, 15). FRESH works by extruding bio-inks within a thermoreversible support bath composed of a gelatin microparticle slurry that provides support during printing and is subsequently melted away at 37°C (Fig. 1, A and B, and movie S1) (16). The original version of the FRESH support bath, termed FRESH v1.0, consisted of irregularly shaped microparticles with a mean diameter of ~65 mm created by mechanical blending of a large gelatin block (Fig. 1C) (16). In FRESH v2.0, we developed a coacervation approach to gen- erate gelatin microparticles with (i) uniform spherical morphology (Fig. 1D), (ii) reduced poly- dispersity (Fig. 1E), (iii) decreased particle diam- eter of ~25 mm (Fig. 1F), and (iv) tunable storage modulus and yield stress (Fig. 1G and fig. S2).
  • 108. FRESH v2.0 improves resolution with the ability to precisely generate collagen filaments and ac- curately reproduce complex G-code, as shown with a window-frame calibration print (Fig. 1H). Using FRESH v1.0, the smallest collagen filament reliably printed was ~250 mm in mean diameter, with highly variable morphology due to the rela- tively large and polydisperse gelatin micropar- ticles (Fig. 1I). In contrast, FRESH v2.0 improves the resolution by an order of magnitude, with collagen filaments reliably printed from 200 mm down to 20 mm in diameter (Fig. 1, I and J). Filament morphology from solid-like to highly porous was controlled by tuning the collagen gelation rate using salt concentration and buffer- ing capacity of the gelatin support bath (fig. S3). A pH 7.4 support bath with 50 mM HEPES was the optimal balance between individual strand resolution and strand-to-strand adhesion and was versatile, enabling FRESH printing of mul- tiple bio-inks with orthogonal gelation mech- anisms including collagen-based inks, alginate, fibrinogen, and methacrylated hyaluronic acid in the same print by adding CaCl2, thrombin, and UV light exposure (fig. S4) (15). We first focused on FRESH-printing a sim- plified model of a small coronary artery–scale linear tube from collagen type I for perfusion with a custom-designed pulsatile perfusion system (Fig. 2A and fig. S5). The linear tube had an inner diameter of 1.4 mm (fig. S6A) and a wall thick- ness of ~300 mm (fig. S6B), and was patent and manifold as determined by dextran perfusion (fig. S6, C to E, and movie S2) (15). C2C12 cells