You are on page 1of 18

Recent advances in 3D printing: vascular

network for tissue and organ regeneration

SUNG YUN HANN, HAITAO CUI, TIMOTHY ESWORTHY, SHIDA MIAO, XUAN ZHOU, SE-JUN LEE,
JOHN P. FISHER, and LIJIE GRACE ZHANG
WASHINGTON, DC; AND COLLEGE PARK, MARYLAND

Over the past years, the fabrication of adequate vascular networks has remained
the main challenge in engineering tissues due to technical difficulties, while the ulti-
mate objective of tissue engineering is to create fully functional and sustainable
organs and tissues to transplant in the human body. There have been a number of
studies performed to overcome this limitation, and as a result, 3D printing has
become an emerging technique to serve in a variety of applications in constructing
vascular networks within tissues and organs. 3D printing incorporated technical
approaches allow researchers to fabricate complex and systematic architecture of
vascular networks and offer various selections for fabrication materials and printing
techniques. In this review, we will discuss materials and strategies for 3D printed vas-
cular networks as well as specific applications for certain vascularized tissue and
organ regeneration. We will also address the current limitations of vascular tissue
engineering and make suggestions for future directions research may take. (Trans-
lational Research 2019; 211:46 63)

Abbreviations: a-SMA = a-smooth muscle actin; BMP-2 = Bone morphogenetic protein-2;


CAD = Computer-aided design; cBCG = Crosslinked bioactive nanocoating with growth fac-
tors; dECM = Decellularized extracellular matrix; ECM = Extracellular matrix; ECs = Endothelial
cells; EPCs = Endothelial progenitor cells; FDM = Fused deposition modeling; GelMA = Gelatin
methacryloyl; HA = Hyaluronic acid; hMSCs = Human mesenchymal stem cells; HUVECs =
Human umbilical vein endothelial cells; MMP = Matrix metalloprotease; PCL = Polycaprolac-
tone; PEGDA = Polyethylene glycol diacrylate; PLA = Polylactic acid; PLGA = Polylactic-gly-
colic acid; SLA = Stereolithography apparatus; SMCs = Smooth muscle cells; UV = Ultraviolet;
VEGF = Vascular endothelial growth factor

INTRODUCTION
From the Department of Mechanical and Aerospace Engineering, The
George Washington University, Washington, DC; Fischell Department The primary aim of tissue engineering is to develop
of Bioengineering, University of Maryland, College Park, Maryland; fully functional and sustainable tissues and organs in
Center for Engineering Complex Tissues, University of Maryland, Col-
lege Park, Maryland; Department of Electrical and Computer Engineer-
vitro and in vivo for repairing or replacing damaged tis-
ing, The George Washington University, Washington, DC; Department sues in the body.1 4 Approaches involved in tissue
of Biomedical Engineering, The George Washington University, Wash- engineering have varied among their specific applica-
ington, DC; Department of Medicine, The George Washington Univer- tions such as regeneration of bone, skin, heart, and
sity Medical Center, Washington, DC. others.5 Although there have been many studies per-
Submitted for Publication January 30, 2019; received submitted formed in that regard, only a few of them have pre-
March 31, 2019; accepted for publication April 2, 2019.
sented successful results from the in vitro level to
Reprint requests: Lijie Grace Zhang, Department of Mechanical and
Aerospace Engineering, The George Washington University, Science
clinical transplantation.6,7 The lack of acquired data in
and Engineering, Hall 3590, 800 22nd Street NW, Washington DC tissue engineering mainly originates from insufficient
20052. E-mail address: lgzhang@email.gwu.edu. technical advancement in the creation of blood vessels,
1931-5244/$ - see front matter which are referred to as vascularized networks.8 Vas-
Ó 2019 Elsevier Inc. All rights reserved. cularization is essential for the supplementation of oxy-
https://doi.org/10.1016/j.trsl.2019.04.002 gen and nutrients, as well as the removal of waste,

46
Translational Research
Volume 211 Hann et al 47

which is necessary for tissues and organs to maintain writing. The FDM technique employs ample heating
their functions.5,9 In general, vascular networks can be for the transition of materials from solid to the
created via vasculogenesis and angiogenesis.9 Vasculo- semi-molten state before solidification at the time of
genesis involves the generation of new blood vessels printing.10 It is compatible with a wide selection of
from endothelial cells (ECs), whereas angiogenesis is plastic biomaterials and offers great versatility in
assisted by germination from existing vessels.9 the study of 3D printing in general. However, FDM
Many techniques have been adopted to fabricate vas- also possesses some drawbacks such as a notably
cular networks with complex, unique structures and limited printing resolution and restriction of hydro-
functionality for mimicking blood vessels in the human gel use. For example, the minimum feature size of
body so far; however, 3D printing techniques have 100 mm and compulsory “molten-solid” state transi-
attracted researchers particularly during the past years tion for the extrusion step are considerable down-
due to outstanding advantages including controlla- sides compared to other 3D printing strategies.14
bility, reproducibility, and repeatability.5 Since 3D Comparatively, bioplotting is a fluid-dispensing sys-
printing was first developed in the 1980s, there have tem, capable of employing a wide selection of cell-
been enormous advancements in tissue and organ laden bioinks such as cell aggregates, hydrogels,
regeneration.10,11 As a result, it has become common micro-carriers, and decellularized matrices.5
to generate vascular networks in organs or tissues by SLA involves a surface photopolymerization of a
utilizing several different 3D printing techniques with liquid or gel state polymer induced by an ultraviolet
a varied selection of materials due to its quickness (UV) laser. SLA primarily utilizes laser energy to gen-
compared to microcirculation in vivo.5 However, 3D erate subsequently thick and solidified layers in the
printed vascularization still remains a challenge since bioink reservoir. Although scaffolds created by SLA
it has been unable to fully mimic the complex structure are not mechanically robust, this strategy offers precise
and function of natural vasculature. control of resolution accompanied by a microscale
In this review, we focus on 3D printing techniques, laser tip. Unlike FDM, SLA offers a limited selection
materials, and other elements that are taken into for bioinks that must be photopolymerizable. To date,
account for the successful formation of vascular net- modification of polymers has been investigated to
works with relevant applications as a means to discuss overcome this restriction, but the number of bioinks is
the current limitations of vascular tissue engineering still limited compared to FDM.10 Also, an excessive
and to propose future research directions. exposure to the UV light can result in geometrical dis-
tortion or shrinkage.10
Here, we focus on the methods to create perfusable
3D PRINTING METHODS FOR VASCULAR 3D vascular networks by classifying them into 2 differ-
FABRICATION ent regimes, direct and indirect printing systems, where
Although the fabrication of perfectly functional vas- the most important aspect in 3D printing vascular net-
cular networks remains a challenge for most tissue works is biomimicry and implementing their complex
engineers,12 there are considerable types of 3D printing and hierarchical design (Fig 1 and Table 1).
methods that are expected to overcome current limita- Direct printing. The first approach is direct patterning
tions. Commonly, many researchers have categorized to implement internal tubular shapes within a scaffold.
3D printing into 2 types by cell seeding method: This approach requires fewer fabrication steps as it per-
Direct-Cell-Seeding and Post-Cell-Seeding by incorpo- forms the creation of blood vessels and tissue in a con-
rating laser-assisted, droplet-based, and extrusion- tinuous, layer-by-layer process. All printing systems
based manufacturing processes.5,13 Regarding the mentioned above can be utilized for direct printing.
methodologies to create vascular constructs, there are Therefore, to date, there have been a variety of studies
2 major approaches that can be taken into consider- performed to demonstrate vascularized tissue and
ation. First, vascularization promoting cells can be organ regenerations using this method. For example,
cocultured within the scaffolds to generate vascular the study done by Suri et al utilized a method in which
networks. Second, perfusable blood vessels can be cre- partially UV cured photopolymerizable glycidyl meth-
ated via direct fabrication. Specifically, in the fabrica- acrylate hyaluronic acid was used to fabricate a micro-
tion of 3D vascular networks, it is conventional to channel incorporated 3D scaffold. Selective reflection
utilize extrusion-based and stereolithography apparatus of UV light using the prepatterned substrate under the
(SLA) printing techniques. SLA system led to partial photopolymerization of the
Extrusion-based techniques including fused depo- ink. A sequential deposition-wash off-deposition pro-
sition modeling (FDM) and bioplotting are the most cess between each layer was incorporated.15 Also, dif-
universally used 3D printing platforms for direct ferent microscale structures with various inner shapes
Translational Research
48 Hann et al September 2019

Fig 1. A diagram tree of the 3D printing techniques for vascular network fabrication.

were printable in polyethylene glycol diacrylate an impermanent template to generate vascular channels
(PEGDA) and lithium phenyl-2, 4, 6-trimethylbenzoyl- within collagen-based tissue structures.21,22 In one of
phosphinate using SLA.16 In addition, Laschke et al the earlier studies, Wu et al printed a 3D microvascular
utilized a 3D bioplotting technique to fabricate poly- network using thermal and chemical characteristics of
lactic-glycolic acid (PLGA) based scaffolds in order to a fugitive ink consist of Pluronic F127.23 Although the
create vascular networks capable of angiogenesis in use of a sacrificial template suggests an advantage over
the presence of growth factor.17 The notorious issue the direct printing methods in the aforementioned geo-
with direct horizontal 3D printing is an erratic fluid metrical and microfluidic aspects, as it is still left with
stream within printed internal microchannels due to constraints in fabrication and feasibility during and
blockage or pleated shape caused by gravity.18 Another after printing. First, this approach is surrounding
interesting direct 3D printing platform was recently hydrogel or polymer construct dependent, thereby pre-
introduced by Hinton et al. They modified a thermo- cise control on the outer material is restricted.5 Second,
plastic extrusion-based 3D printer by adding a custom- an efficient method to interconnect the vessels with a
built syringe pump extrusion system to it.19 The system complicated design to mimic the natural vascular net-
incorporated a thermo-reversible support bath with gel- work needs to be resolved. The previously performed
atin microparticles for specialized use in 3D printing of studies are mostly implementable to vascularized net-
hydrogel bioinks in a layer-by-layer fashion. Upon the works which lie horizontally between the layers or sim-
completion of printing, sufficient heating removed the ply stack vertically, which is a limited approach to
gelatin support, leaving only the printed structure. The model the complex design of native vascularized net-
authors showed that this modified extrusion 3D printer works.
was capable of fabricating complex geometrical struc-
tures, such as coronary artery vascular trees and embry-
onic chick heart.19 Thus, this platform can be MATERIAL AND CELL SELECTIONS
considered useful for 3D printing of complex biologi- The next aspect of 3D printing is the selection of
cal structures with hydrogels. versatile bioinks and cells. There are properties
Indirect printing. The utilization of a sacrificial tem- required that biomaterials must fulfill in order to
plate that leads to indirect printing overcomes the main achieve a desired 3D printed construct with its biologi-
drawback of direct patterning on the substrate. Golden cal functionalization. Above all, biomaterials should
et al fabricated a minimum of 6 mm wide internal chan- exhibit suitability to implement innate features of
nel using the liquefaction temperature difference native blood vessels, which are composed of collagen
between the Polydimethylsiloxane substrate and the and elastic fibers, elastic lamellae, and proteoglycans.24
gelatin mesh that they used. In their study, heating at Besides bearing resemblances to the material properties
room temperature for 1 hour was substantial to allow of native blood vessels, a bioink must also have char-
the gelatin to melt. As a result, scaffolds with hollow acteristics which can be explicitly summarized in terms
microchannels consisting of open ends were fabri- of printability, biocompatibility, biodegradability, and
cated.20 Similarly, multiple studies utilized gelatin as suitable mechanical properties. Among various groups
Translational Research
Volume 211 Hann et al 49

and cell damage by UV light


of chemicals, polymers occupy the majority of conven-

Limited selection for bioinks


tional bioinks due to the possession of the listed prop-

Limited 3D resolution and

restricted hydrogel use


erties above. Polymers for bioinks are divided into 2

restricted cell density

Limited resolution and


major categories by their origin, natural and synthetic
(Table 2). The natural polymers are naturally obtained

Slow fabrication
Disadvantages

or extracted from humans, animals, bacteria, or plants,


whereas synthetic polymers are man-made and inten-
tionally synthesized to supplement the drawbacks of
natural polymers, which will be discussed in the fol-
lowing subsections. To formulate a suitable bioink,
there are a number of biological and mechanical char-
acteristics of the materials which must be taken into
consideration. Regardless of the classification, the bio-
Good versatility and a wide
Low cost, least damage to

ink should exhibit degradability and biocompatibility


printed cells, and rapid

Wide selection of bioinks

for cell proliferation and growth as well as printabil-


selection of bioinks

ity.10 Especially in the case of vascular fabrication,


shape fidelity, stability (eg, swelling) and flexibility are
High resolution

fabrication
Advantages

also considerable requirements for material selection as


they are directly relevant to the sustainability of tubular
and pore structures.10 According to the “biofabrication
window” described by Malda et al and Sun et al, the
printability of bioinks is proportional to polymer con-
centration, which affects crosslinkability and shape
fidelity for 3D printing of vascular networks.25,26
Simultaneous use of multiple
Adjustable droplet size and

Heating for state transition

However, a densely cross-linked environment interferes


with cell viability, which means the degree of biocom-
photopolymerization

patibility of the printed construct is constrained by its


Unique properties

mechanical properties.25 Therefore, moderate crosslink-


ing of the biopolymer is necessary to obtain suitable
of materials

biological and mechanical properties.


bioinks
shape
Surface

In addition to the listed properties of bioinks, micro-


environmental conditions must be considered for the
Table 1. List of 3D printing methods for vascular fabrication5,10,13,14,108

initiation of vasculogenesis and angiogenesis. For


example, one of the cell types to be discussed in the
following section, endothelial progenitor cells (EPCs),
have been shown to be influenced by laminar shear
Fused deposition modeling
Stereolithography appara-

stress, which in turn promotes vasculogenesis.27


According to the study by Yamamoto et al, laminar
Printing technique

shear stress was found to stimulate EPC proliferation


and morphological changes. Thus, vasculogenesis was
promoted in dynamic culture conditions, while greater
Bioplotting
tus (SLA)

angiogenesis was observed in static conditions.27


(FDM)

Another microenvironmental condition for ECs to


Inkjet

form vascular networks is the inclusion of bioactive


growth factors, such as the use of vascular endothelial
growth factor (VEGF).28 Although concentrations of
VEGF outside of the ideal range may result in abnor-
Technical classification

mal constructs, proper use of VEGF is crucial to accel-


erate the formation of vascular networks.28
Extrusion-based
Droplet-based

Natural polymers. Natural polymers are widely used


Laser-assisted

as the fundamental elements to obtain biomimetic


characteristics of the extracellular matrix (ECM),
which is considered the biggest advantage. However,
there are noteworthy drawbacks to natural polymers as
50
Table 2. List of natural and synthetic materials for bioinks4,5,14,23,30,35,38,41,42,25,46 48,76 78,109, 110 127

Hann et al
Material classification Printing technique Solidification mechanism Strengths Drawbacks
Protein Collagen and its Droplet, extrusion, and SLA Physical or chemical Nonimmunogenic Low cell adhesion without
derivatives crosslinking modification
Gelatin and its Droplet, extrusion, and SLA Physical or chemical Nonimmunogenic Weak mechanical properties
derivatives crosslinking without modification
Elastin Droplet and extrusion Self-assembling Good biocompatibility Calcification, hydrophobicity,
and insolubility
Fibrin Droplet, extrusion, and SLA Self-assembling Angiogenesis enhancement Low mechanical property
Matrigel Droplet and extrusion Self-assembling Self-renewal and pluripotency Low mechanical property
for stem cells
Polysaccharide Agarose Droplet and extrusion Physical crosslinking Thermo-reversibility Low cell adhesion without
modification
Alginate Droplet and extrusion Physical crosslinking Biocompatibility, fast gelation, Low cell adhesion without
and nonimmunogenic modification
Chitosan Extrusion Chemical crosslinking pH-sensitive solubility, biocom- High viscosity
patibility, and hydrophilicity
Hyaluronic acid and Droplet, extrusion and SLA Physical or chemical The similarity to ECM and cell Poor mechanical property
its derivatives crosslinking viability
Decellularized ECM Droplet and extrusion Physical or chemical Natural ECM biomimicry and Complex preparation steps
crosslinking tissue specificity
Polyethers Poly (ethylene glycol) SLA Covalent crosslinking Nonimmunogenic Low cell adhesion
diacrylate
Polyesters Poly (caprolactone) Extrusion and SLA Temperature or covalent Good biocompatibility Very slow degradation
and its derivatives crosslinking
Poly (lactic acid) Extrusion Temperature Good biocompatibility Immunogenicity
Poly (propylene SLA Covalent crosslinking Good biodegradability Immunogenicity
fumarate)
Poloxamer Pluronic F127 Extrusion Physical crosslinking Nonimmunogenic and high Poor mechanical property
printability and slow gelation

Translational Research
September 2019
Translational Research
Volume 211 Hann et al 51

wells, such as their lack of mechanical strength and photocrosslinkable material such as methacryloyl to
controllability over their chemical structure.5 Selection induce crosslinked formation.39 Agarose is another
of natural polymers for 3D printing varies depending type of polysaccharide-based natural polymer, which is
on which physical and chemical characteristics are to derived from red seaweed and is largely composed
be exploited such as chemical crosslinkability, temper- of (1-3)-b-D-galactopyranose-(1-4)-3, 6-anhydro-a-L-
ature sensitivity, or others. In addition to the inherent galactopyranose units.40,41 Due to its poor capacity to
characteristics of natural polymers, the selection of promote cell adhesion and proliferation, agarose nor-
bioink is printing method dependent. For instance, mally undergoes chemical modification or combination
extrusion-based printing and SLA are capable of print- with other biocompatible polymers to be used for the
ing hydrogels, microcarriers, tissue spheroids, cell pel- scaffold fabrication. One of the reported techniques to
lets, tissue strands, and decellularized matrix promote cellular adhesion and mechanical stability of
components.5,14 Here, we classify natural polymers for agarose is blending agarose with gelatin.42 Chitosan is
3D printing into 2 categories based on their basal com- created by alkaline hydrolysis of chitin from the shells
ponents such as proteins and polysaccharides. Natural of crustaceans and consists of b-(1-4)-linked N-acetyl-
polymers include collagen, gelatin, elastin, fibrinogen, D-glucosamine and D-glucosamine subunits.43 Chito-
and Matrigel are protein derived, while cellulose, algi- san offers multiple options for chemical modifications
nate, hyaluronic acid, agarose, and chitosan are poly- due to its unique characteristics, such as pH-sensitive
saccharide derived polymers. Collagen is one of the solubility, hydrophilicity, and biocompatibility.41 As a
generally used proteins in tissue engineering as it is the result, chitosan has been successfully exploited in
most abundant protein of ECM in the human body.29 many tissue engineering studies.44,45
Among 28 different types of identified collagen, type-I With regards for the aspect of natural ECM biomi-
collagen occupies the largest portion in the mammalian micry, decellularized ECM (dECM) are excellent can-
body and cell culture. Gelatin is a derivative of colla- didates for tissue-specific bioinks. There have been a
gen obtained via partial hydrolysis of collagen.30 In number of dECM based bioink preparation protocols
previous studies, gelatin has demonstrated an impor- reported, and researchers have utilized them for both
tant role in vivo for its biodegradability and a sacrificial vasculature and other cell studies.46 48 To promote the
template in vascularization by using its temperature printability and cell viability of dECM based bioinks,
sensitivity.21,22,31 Fibrinogen is a protein that has been it is a common practice to blend them with synthetic
extracted from the bloodstream. Once fibrinogen is polymeric hydrogels or covalent crosslinkers.46,49,50
combined with the enzyme thrombin, it forms mono- Synthetic polymers. The biggest advantage of syn-
meric fibrin.32 Due to its superb cytocompatibility, thetic polymers over natural polymers is controllability
many researchers have utilized fibrinogen in the form of their physical and chemical properties, which
of fibrin to promote cell adhesion in their studies.32,33 implies they can be custom-formulated for51 particular
Matrigel is a mixture of ECM proteins secreted from printing platforms and methods. However, synthetic
Englebreth-Holm-Swarm mouse sarcoma cells,34,35 polymers also exhibit limitations which inhibit their
and is notable for its ability to mimic the ECM of vari- greater usability such as a lack of biocompatibility,
ous cancer and stem cell lines, as well as its capacity to toxic degradation, and loss of mechanical properties
maintain the stem cells in an undifferentiated state.35 during degradation.13
Similarly, polysaccharide derived polymers possess Here, we focus on the prevalent synthetic polymers
the same biomimicry of natural polymers. Alginate for 3D printing inks that have been used. Among the
(alginic acid) is a polysaccharide refined from brown variety of bioinks available, polylactic acid (PLA)
seaweed and composed of (1-4)-linked b-D-mannur- exhibits biodegradability, nontoxicity, printability,
onic acid and a-L-guluronic acid monomers.36 Algi- easy processability, and biocompatibility so that it has
nate is capable of forming ionic hydrogels with become the outstanding synthetic polymer for use in
moderate cell attraction.5,36 In order to enhance cell FDM printing.51 53 However, high processing temper-
adhesion of native alginate, prior studies introduced ature limits has its availability for cell-laden bioprint-
covalent modification.37 Hyaluronic acid (HA) or hya- ing. Additionally, PLA has insufficient mechanical
luronan is a key component of the ECM, and therefore strength and bioactivity for utilization in bone replace-
has been clinically used in the past decades. HA is ment, so it is necessary to be used in combination with
composed of (1-4)-linked b-D-glucuronic and (1-3)- other biocompatible ceramics.52 Polycaprolactone
linked b-N-acetyl-D-glucosamine residues.38 Unmodi- (PCL) has also attracted many researchers as it offers
fied HA has several limitations to be used as a bioink nontoxicity and low liquefaction temperature, which
due to its high viscosity and weak formability so that it lead to easy blending with other natural polymers and
requires chemical modifications and mixing with a relatively higher cell viability for extrusion-based
Translational Research
52 Hann et al September 2019

printing.54 PEGDA is also used for cell-laden bioprint- they are obtained via reprogramming or differentiation
ing, due to its excellent crosslinkability under UV or of autologous stem cells. However, there are con-
visible light exposure although it is not degradable straints for autologous and many primary cells to be
material. PEGDA is commonly combined with photo- utilized in 3D printed constructs, which involves the
crosslinkable natural polymers such as gelatin metha- isolation of cells to be used in the in vitro environment
cryloyl (GelMA) to improve biodegradability and bio- and their finite lifespan.5,13 Stem cells, such as human
activity.55 Polypropylene fumarate is a biocompatible embryonic stem cells and human induced pluripotent
and biodegradable polymer and possesses photocros- stem cells, are key cell sources to overcome the
slinkability, so it is suitable for SLA based 3D fabrica- obstacles associated with the current cell reservoirs.
tion techniques. In a recent study, polypropylene Embryonic stem cells and induced pluripotent stem
fumarate was used as a favored material for vascular cells exhibit the capability to differentiate into unlim-
study which includes venous and aortic graft models.56 ited lineages, which include ECs, cardiomyocytes,
As mentioned in the previous section, Pluronic F127 is chondrocytes, osteoblasts, hepatocytes, insulin-secret-
one of the promising candidates among synthetic poly- ing beta cells, and neural cells.59 Specifically, in the
mers for sacrificial templates for 3D vascular fabrica- case of vascularization studies, it is critical to under-
tion study. Pluronic F127 belongs to the poloxamer stand that native blood vessels vary in dimension and
class of polymers, which are nonionic triblock copolymers number of layers. Therefore, it is apparent that the cell
composed of polyoxypropylene bridging 2 polyoxyethy- composition of each layer may differ. Among all types
lenes to maintain polyoxyethylenes/polyoxypropylene of cells, ECs are the basic components of the innermost
ratio of 2:1 by weight.57 As a fugitive ink, the concen- layer of native blood vessels.5 The presence of ECs is
tration of aqueous Pluronic F127 solution must be ubiquitous in the entire vascular network from arteries
greater than 20%, which is the transitional point to to capillaries. Also, they play a critical role in angio-
exhibit temperature sensitive gelation behavior.57 genesis, vasoconstriction, and vasodilation. Therefore,
Chemically modifying Pluronic F127 through acryla- ECs are of the most important cell-type to consider for
tion or methacrylation offers greater crosslinkability vascularization study.1 EPCs from bone-marrow are
under UV or visible light exposure, which enhances known as supportive cell sources for angiogenesis and
its benefit as a bioink.5 The combination of the 2 prop- differentiation toward ECs.1,60 Along with ECs, the
erties, thermal reversibility of gelation and photocros- main cell components of the middle layer of large ves-
slinkablity, allows Pluronic F127 to be employed for sels, smooth muscle cells (SMCs), can also be utilized
patterning of 3D vascular networks.23 for angiogenesis due to their self-assembly capability.5
Cell sources. The choice of cell types is extremely In addition, coculture systems using multiple types of
significant to construct the intended tissue with its cells to improve vascular network generation have
associated functionality. As such different tissues and been widely used in related studies. For instance,
organs consist of various types of cells with multiple human mesenchymal stem cells (hMSCs) can be cocul-
biological functions that must be recapitulated in the tured with ECs to promote angiogenesis and cell viabil-
regenerated tissue.5,13 Besides, the primary functional ity in cardiovascular studies.1,61
cells, most tissues contain multiple ancillary functional In 3D printing of tissues or organs, cells can be
cell types to provide supportive, structural or barrier directly encapsulated in bioinks during the printing
functions for vascularization or provide a necessary process or seeded on 3D constructs after printing. For
environment for stem cell maintenance and differentia- direct cell encapsulation, they must present robustness
tion.13 To that end, it is essential to take into account to endure physical and biological stresses including the
that the choice of cells for 3D printing incorporates the existence of external forces, pressure, and others.13 In
functionalities of primary and other cells as well as the post seeding, the positioning of cells at the desired
interactions between them. Cells chosen for 3D print- location of the fabricated 3D construct remains a criti-
ing are expected to mimic their behavior and function- cal challenge.5
alities in vivo such as cellular homeostasis, self-
renewability, responsiveness to tissue damage, and
integration with host tissue.5,13,58 The proper control of
cell proliferation in vivo and in vitro is also significant VASCULAR NETWORK FABRICATION AND
since an insufficient number of cells may result in the APPLICATIONS
loss of tissue and cell viability, whereas the excessive To preserve and mimic the metabolic functions of
proliferation may result in hyperplasia or apoptosis.13 native tissues, including transportation of nutrients and
The preferable source of cells is autologous due to oxygen, as well as the removal of cellular waste and
their adaptability and minimal immune response, as CO2, the robust formation of a perfusable vascular
Translational Research
Volume 211 Hann et al 53

network is significant for 3D printed tissues or organs.9 osteogenesis.67 Since the 3D printing technique has
For example, cells located further than 200 mm from attracted many researchers as a superb substitute for
the closest capillaries experience hypoxia and apopto- traditional bone graft fabrication, various studies have
sis due to limited oxygen and nutrient supply.5,8 It is demonstrated the importance of vascularization for 3D
known that native blood vessels vary in scale from cen- bone printing. As a result, vasculogenesis and angio-
timeters (aorta) to microns (capillaries) with different genesis have become characteristic processes that must
blood pressures.18 As mentioned above, the native be taken into account for the design of 3D bone regen-
blood vessels are classified into 3 major components: eration studies. Wang et al demonstrated a method
(1) arteries (arterioles), (2) capillaries, and (3) veins using a virus-activated matrix with arginine-glycine-
(venules). Arteries hold the highest blood pressures aspartic acid peptide phage nanofibers incorporated 3D
and transport blood from the heart, while veins carry printing to induce vascularized osteogenesis. In their
blood back to the heart, and capillaries bridge them. study, the rat mesenchymal stem cell seeded scaffold
The blood vessels consist of 3 primary layers, tunica was used as a bone void filler and generated new blood
adventitia, tunica media, and tunica intima. The out- vessels in the bone defect area.64 The researchers in
most layer, tunica adventitia, is comprised of collagen, our lab, Holmes et al, previously performed nanocrys-
fibroblasts, and ECM. The tunica media, the middle talline hydroxyapatite incorporated microfeatured vas-
layer is the thickest layer and primarily consists of cularized bone scaffold.68 Temple et al designed and
SMCs and pericytes, which demonstrate distinctive fabricated anatomically shaped vascularized bone
cardioprotective functions such as angiogenic genera- grafts with PCL, which is synthetic and biodegradable,
tion and chronic inflammation reduction. The tunica and human adipose-derived stem cells using 3D print-
intima is the innermost vascular layer and is comprised ing technology.69 Similarly, Yan et al recently reported
of ECs. It is responsible for conducting blood flow reg- a study of PCL-based, vascularized 3D printed bone
ulation, as well as platelet and leukocyte control.62 scaffolds that can control the release of deferoxamine,
Hierarchical integration of the listed features above which is regarded as a desirable material to enhance
within a 3D printed structure is currently incomplete vascularization and bone regeneration.70 72 A PLGA
as it requires precise geometric and functional con- and nanocrystalline hydroxyapatite based 3D bioplot-
trols over fabrication, especially at the submicron ting technique to fabricate hyperelastic vascularized
scale. Herein, we mainly introduce the current techni- bone has also been recently reported.73 In our lab, we
ques for modeling a 3D vascular network incorporated have successfully created an engineered vascularized
into tissues and organs, such as vascularized bone, bone with a fluid perfusble microstructure by 3D FDM
vascularized skeletal muscle, cardiovascular system, printing. A biologically inspired nanocoating technique
liver, and brain to analyze their innovativeness in bio- was incorporated into the 3D printed scaffolds, in order
fabrication of complicated structural and functional to obtain the smart delivery of dual growth factors
characteristics. (BMP-2 and VEGF) with the sequential release in spa-
Vascularized bone. In the human body, bone tissue is tiotemporal coordination (Fig 2a d).2 In another
highly vascularized and metabolically active, and is recent study, we proposed an integrated approach for
comprised of a rigid natural composite of collagen and complex vascularized bone regeneration, which com-
hydroxyapatite. It is classified into 2 mineralized tis- bines an advanced dual 3D printing platform with
sues, cortical bone which has 10% 30% porosity and regional immobilization strategies of bioactive factors.
a hard outer layer, and cancellous bone with a The combination of hard PLA bone scaffolds and soft
30% 90% porosity interior.63 Such a vascularized sys- GelMA vessels provided a promising platform to
tem enables the supply of nutrients, oxygen, and blood, obtain a hierarchically biomimetic construct, with mul-
as well as the removal of waste products to preserve tiphasic characteristics (Fig 2e i).3 These approaches
skeletal coherence.64,65 For this reason, intraosseous present clinical potential to generate biomimetic vascu-
vasculatures in the human body stay within 100 mm larized bone grafts with complex 3D structure and bio-
from their closest capillaries. Otherwise, insufficient compatible functionalities.
blood supply by incomplete vascularization will result Vascularized skeletal muscle. Muscles are primarily
in avascular necrosis. In addition, intraosseous vascula- responsible for the movement control and force genera-
ture offers advanced functions to maintain skeletal tion through tendons by contraction as they are bonded
integrity, such as bone remodeling, development, and to bones or internal organs.74 Muscles including skele-
fracture repair.66 tal muscles, cardiac muscle (myocardium), and smooth
Traditionally, an ideal bone graft model was muscles are mainly comprised of muscle fibers, which
expected to possess a number of properties including consist of myofibrils with myofilaments as subcompo-
osteointegration, osteoconduction, osteoinduction, and sites and interweaved high-density vessels.74 Among
Translational Research
54 Hann et al September 2019

the 3 major muscle types, skeletal muscles are attached supply for autografts and potential risk of unexpected
to bones directly or indirectly via tendons, which have transplant rejection for allografts have accelerated the
been independently investigated for the development need for an alternative solution.80 As a result, a number
of artificial fabrication techniques, while cardiac and of studies for vascularized skin with 3D printing have
smooth muscles are commonly combined with the cor- been performed.47,81 83
responding internal organs. Vascular networks are In the recently reported study by Kim et al, porcine
directly linked to the primary role of skeletal muscles, skin-derived dECM was utilized as a raw material in a
the actuation of contraction. bioink to fabricate a 3D printed prevascularized skin
In order for 3D printed skeletal muscles to possess patch. In their study, extrusion-based and inkjet-based
therapeutic potential, seamless propagation of applied 3D printing techniques were exploited to model the
load must be taken into account, as natural skeletal human dermis and epidermis in a layer-by-layer fash-
muscles are capable of achieving efficient force trans- ion for an in vitro experiment. Simultaneously, human
mission.74 In addition, natural muscle, like bone, is a adipose-derived stem cells and EPCs laden bioinks
highly vascularized tissue type and accounts for most were used in the creation of the prevascularized skin
of the energy consumption in the human body. In other patch for in vivo study.47 Michael et al used a laser-
words, more precise control and even dispersion of the assisted 3D printing technique to engineer skin substi-
vascular network fabrication are required for 3D tutes in vivo. The multiple layers of fibroblast-based
printed skeletal muscles to perform adequate force and keratinocyte-based collagen were 3D printed onto
delivery. a sheet of the stabilization matrix. In their study, vascu-
Many researchers have utilized genetically modified larization was promoted by the presence of keratino-
myoblasts or growth factor delivery matrices to cytes, which are known to generate VEGF.81,84
enhance vascularization on fabricated skeletal muscle Moreover, Skardal et al utilized a direct 3D printing
tissue.75 78 For example, Von Degenfeld et al demon- approach of amniotic fluid-derived stem cells to pro-
strated an approach to generate new blood vessels mote vascularization of skin in vivo.82 These 3D print-
using myoblasts with growth factors including VEGF ing-based experimental results have suggested the use
and fibroblast growth factor.77 After the transition to of different cellular approaches to conventional cell-
the 3D printing era, a number of techniques have been based methods, which typically include ECs or hMSCs
utilized to generate vascularized muscle tissue con- for the fabrication of vascularized skin.
structs. Levenberg et al engineered vascularized skele- Cardiovascular system. The cardiovascular system
tal muscle tissues in vitro by coculturing myoblasts, includes the heart, blood vessels (arteries, veins, arte-
embryonic fibroblasts, and ECs in poly-(L-lactic acid) riovenous shunts, and capillaries), and lymphatic ves-
and PLGA based 3D scaffolds.75 Their results have sels.1 Based on the statistics provided by the American
shown that prevascularized skeletal muscle tissues gen- Heart Association, cardiovascular disease directly or
erated by an endothelial coculture system help induce indirectly accounts for beyond 55% of all deaths in the
vascular networks in vivo (Fig 3). United States, and has been the No. 1 reason for death
Vascularized skin. The skin accounts for approxi- since 1900.85 Although cardiovascular disease has
mately 16% of the body’s weight and is the primary been recognized as the primary risk of decreasing the
component of the integumentary system of the human average life expectancy during the past century, the
body.79 The most common misunderstanding with clinical approaches incorporating organ transplantation
regard to the skin and its accessory structures is that still experience remaining challenges due to a lack of
they belong are a single type of tissue. In fact, the skin donor tissue.
consists of multiple layers of tissues and is classified as The primary organ of the cardiovascular system, the
the largest organ of the human body, and provides the heart consists of several types of cardiac tissues such
outmost barrier against the environment and patho- as myocardium, endocardium, and pericardium.86 The
gens.47,79 The human skin consists of 3 primary layers: myocardium is a thick muscle wall, composed of cardi-
the epidermis, dermis, and hypodermis, with each hav- omyocytes (or cardiac muscle cells) to perform con-
ing various subdivisions.79 traction and relaxation to supply blood.5 The
Fabrication of artificial skin has been actively pur- endocardium lies in the innermost layer and primarily
sued for cosmetic and therapeutic purposes, such as consists of ECs, and the pericardium is a double-walled
substitution of skin loss or defects. Before significant layer which covers the exterior of the heart.86 A high-
attention had been drawn toward the 3D printing density capillary network (3000/mm2) is located in the
approach, and even still in the present day, autografts myocardium a functions to regulate the metabolic
and allografts have been the most prevalent treatment activity of contraction, and works to confine the dis-
for large skin injuries.80 However, limited donor site tance between cardiomyocytes and ECs within a range
Translational Research
Volume 211 Hann et al 55

Fig 2. (a) Schematic illustration of the fabrication process of 3D bioprinted vascularized bone scaffolds with
bioactive growth factor nanocoatings. (b) A 3D confocal fluorescence image of hMSCs (green) and human
umbilical vein endothelial cells (HUVECs) (red) co-cultured for 5 days. (c) Immunofluorescence images of
hMSCs and HUVECs with CD31 antibody for 2 and 4 weeks on different scaffolds. A PLA based control scaf-
folds (left) and a genipin crosslinked bioactive nanocoating with growth factors (cBCG) based scaffold (right).
Scale bars, 100 mm. (d) Mineralization of osteogenic differentiation of hMSCs and hMSCs/HUVECs on PLA
and cBCG based scaffolds analyzed by Alizarin red staining. (e) Schematic illustration of a dual 3D bioprinting
platform. (f) Schematic representation of the microstructural design of a biomimetic biphasic vascularized bone
construct based on a matrix metalloprotease (MMP) sensitive GelMA hydrogel. The evolution of vascular lumen
and capillary network formation can be achieved in different regions during the culture period. (g) Computer-
aided design (CAD) model of the biphasic vascularized bone construct, including the bone region and vascular
channels (left). Microscopic photo image of manufactured vascularized bone construct (right). Red circles show
tubular vascular hydrogel regions. (h) A 3D immunofluorescence image of the vascular capillary network with
CD31 antibody in the designed different regions of the 10 wt% GelMA hydrogel printed by the dual 3D bioprint-
ing platform after 4 weeks. Scale bar, 50 mm. (i) A 3D immunofluorescence image of a vascular lumen in the 3D
bioprinted vascularized bone. Scale bar, 50 mm. Images are adapted from.2,3 (Color version of figure is available
online.)

of 2 3 mm. Additionally, the blood vessels in the car- Perfusable Vascular Networks. As mentioned above,
diovascular system have complex tree-shape structures, the absence of perfusable vascular networks in 3D
and exhibit vessel diameters in the range of printed scaffolds ultimately leads to necrosis of core
»5 10 mm for the smallest capillaries, to 30 mm for tissue, referred to as avascular necrosis, which results
the largest artery.1 Therefore, along with the develop- from over population of seeded cells and a lack of
ment of 3D vascularized network, the replacement of nutrition and oxygen supply.87 One of the reported
these cardiovascular tissues using 3D printing technol- studies to fabricate 3D printed perfusable vascular
ogy has been considered as an important issue in the tubes used a coaxial extrusion platform with a blended
clinical perspective. bioink of GelMA, sodium alginate, and 4-arm poly
Translational Research
56 Hann et al September 2019

Fig 3. (a) 3D printed scaffold before cell seeding. Scale bar, 1 mm. (b) Immunostaining images of vascular net-
work generation in vitro within skeletal muscle construct after one month. Skeletal myoblasts, ECs, and mouse
embryonic fibroblasts were tri-cultured. Scale bar, 50 mm. (c) Immunostaining images of in vivo study of the
vascularized skeletal model after two weeks of transplantation into immunodeficient mice. The constructs under-
went immunostaining with human-specific CD31 antibody. Scale bar, 50 mm. Images are adapted from.75

(ethylene glycol)-tetra-acrylate.88 In this study, 3D Moreover, our lab recently developed novel gelatin-
printed perfusable blood vessels were created in vari- based 3D printed vascular constructs with 2 distinctive
ous diameters and shape to prove physical and biolog- layers that are comprised of ECs and SMCs. As shown
ical stabilities. Miller et al have created 3D multiscale in Fig 4, the uniform distribution of ECs and SMCs
interconnected vascular networks with carbohydrate was demonstrated inside and outside the lumen, respec-
glass as a sacrificial material using extrusion-based tively. In addition to that, the 3D printed vascular con-
3D printing. They demonstrated the key components structs have shown outstanding abilities of myogenesis
of vascularized tissues including a vascular lumen, and vasculogenesis. In another recent study, we fabri-
HUVECs, and cells matrix in the interstitial zone.87 cated a 3D printed vascularized cardiac patch and suc-
Cui et al used a modified 3D thermal inkjet printer to cessfully implanted into the infarcted heart of mice
fabricate microvasculature. The authors utilized the (Fig 4). The experimental results prove that our inno-
simultaneous deposition of human microvascular vative techniques possess the feasibility to create bio-
endothelial cells, which are capable of forming mimetic structures for cardiac tissue regeneration.
capillaries, and fibrin, which has the capability of Most of the current 3D printed cardiovascular sys-
wound healing and promoting cell migration.89 This tems primarily lack mechanical robustness due to the
study demonstrated the integrated formation of perfus- inherent mechanical characteristics of their bioink
able microvasculature as cells proliferate. Thus, their materials, especially when hydrogels are used.91,92 The
approach can be considered for cardiovascular net- poor mechanical properties of the hydrogels in compar-
works formation based on 3D thermal inkjet printing. ison to the native blood vessels and valves of the car-
Vascularized Cardiac Tissues. Along with a notable diovascular system are considered to be the biggest
focus on the fabrication of perfusable tubular con- impedance to successful 3D cardiac printing. The lim-
structs, there have been a number of investigations on ited mechanical strength of hydrogels leads to poor
integrated cardiac tissues. For example, Zhang et al structural integrity and clinical failure after implanta-
performed 3D double crosslinked printing of vascular- tion. As such, prospective research with emphasis on
ized myocardial constructs.90 In order to generate the development of new suitable materials is ultimately
blood vessels, they utilized the combination of the 2 required. In the same context of structural integrity,
aforementioned approaches including vasculogenesis another considerable challenge to fabricate transla-
and angiogenesis using the self-promotion of ECs and tional 3D printed vascularized cardiac tissues is the
direct fabrication using the 3D printing technique.90 electrophysiological connection of cardiomyocytes to
Their strategy to encapsulate ECs in the 3D printed determine the proper activity of the sinoatrial node as a
microfiber structure offers a convincing approach to pacemaker.93 From the 3D printing techniques per-
develop hydrogel based vascular myocardial tissues. spective, 3D inkjet-based printed cardiovascular
Translational Research
Volume 211 Hann et al 57

Fig 4. (a) 3D printed vasculature with hollow and interconnected structure after 24 hours of perfusion culture.
(b) An immunofluorescence image of 3D printed vasculature (CD 31 antibody and a-smooth muscle actin
[a-SMA]) in a dynamic culture condition. (c) An optical microscopic image of dynamic flow in the 3D printed
vasculature. (d) An optical microscopic image of anisotropic honeycomb constructs of 3D bioprinted vascular-
ized cardiac patch. (e) A fluorescence image of 3D bioprinted vascularized cardiac patch (cardiomyocyte and
ECs). (f) in vivo implantation of 3D bioprinted vascularized cardiac patch into infarcted heart of mice.

systems are restricted to low viscosity bioinks to pre- Previously, Griffith et al fabricated a vascularized
vent nozzle clogging. On the contrary, bioinks with liver on a small scale with the 3D printing technique.6
high viscosity are only applicable for extrusion-based Up until now, many tissue engineers have attempted to
3D printing of cardiovascular systems, which results in fabricate biomimetic 3D printed vascularized liver
poor cell viability. Cell-laden cardiovascular systems constructs with the incorporation of the characteristics
that are 3D printed by the SLA method often result in listed above and its own unique properties such as
poor cell viability due to both the DNA damage rapid restoration ability even after considerable dam-
incurred by resident cells during the UV photocros- age.94 For example, 3D printing of hepatocytes using a
slinking process, and the cytotoxicity caused by vari- hydrogel has been introduced.95 Robbins et al from
ous photoinitiators.1 one of the 3D printing companies for human tissue,
Liver. The liver is considered one of the most signifi- Organovo, presented in vitro models of 3D vascular-
cant organs in the human body due to its special char- ized liver. The researchers demonstrated a multicellu-
acteristics. The liver is responsible for a number of lar liver structure involving hepatocytes, hepatic
important roles related to metabolism and metabolic stellates, and ECs.96 The liver transplantation into the
regulation, such as filtration of blood from the diges- patient’s body performed by Zein et al also proved
tive tract, detoxification of chemicals, accessory diges- the validity of the 3D printing techniques for the vas-
tion, and nutrient storage in the form of glycogen.5,79 cularized liver. Prior to the transplantation, the
The liver is primarily classified into 2 major lobes, left dimensions of the donor and recipient livers in detail
and right lobes. The right lobe, which is much bigger were recorded including the diameters of veins to
than the left lobe, involves 2 minor lobes such as quad- fabricate a vascularized liver using the 3D printing
rate and caudate lobes. Blood is supplied to the liver technique. To implement the external vasculariza-
through 2 different vessels. The hepatic artery supplies tion, the authors utilized a permanent adhesive to
arterial blood from the heart to the liver, and the attach to the liver lobe (Fig 5).7 These results demon-
hepatic portal vein carries blood consisting of nutrients strate the potential efficacy of 3D printed synthetic
and toxins from the intestines to the liver.79 liver with a vascular network in the human body as a
Translational Research
58 Hann et al September 2019

Fig 5. (a) Side view of 3D printed liver and extracted liver of a patient, where long, short, and double arrows
indicate hepatic artery, hepatic vein, and portal vein, respectively. (b) Right lobes of 3D printed and extracted
livers with indications of the hepatic artery (single arrows) and portal vein (double arrows). (c) Cross-sectional
views of 3D printed and extracted livers with indications of hepatic vein (single arrows) and portal vein (dotted
arrows). Images are adapted from.7

valuable tool for drug delivery and a substitute for carbon nanotubes and graphene are other viable candi-
treating partially or irreversibly damaged liver tissue. dates for 3D printing neural tissues due to their supe-
Brain. The brain, along with the spinal cords, is the rior capacity for neuronal cell-specific adhesion and
main component of the human central nervous system. outstanding electromechanical characteristics.100,101
Although the peripheral nervous system, which accounts Due to the inherent complexity of the blood-brain-
for the rest of the nervous system, possesses the capacity battier structure, the 3D printing incorporated vascular-
of innate regeneration, the central nervous system ization of the brain still remains challenging. As a
remains permanently damaged after severe injury due to result, the current progress of the associated studies is
the glial scar formation (gliosis).97 The brain consumes mostly limited at demonstrating an external shape of
approximately 25 percent of the total amount of oxygen the brain including skulls or focusing on the fabrication
in the human body to maintain its functionality as the of neural networking. For instance, Naftulin et al per-
command center. Therefore, sufficient blood supply is a formed a study in that manner to demonstrate an FDM
critical requirement, as the brain is comprised of a com- based 3D printed brain with synthetic polymers such as
plex vascularized system.79,98 Furthermore, the brain has PLA and acrylonitrile butadiene styrene.105 Lozano et
a distinctive mechanism called the blood-brain-battier, al designed and demonstrated a 3D printed brain shape
which is formed by brain ECs, to regulate the microen- construct using a novel peptide modified polymer, gel-
vironment.99 Considering these special characteristics lan gum-arginine-glycine-aspartic acid with primary
and importance, the 3D printing technique for a replica- cortical neurons. Utilizing a simplified hand-held extru-
ble brain has attracted many researchers. sion-based 3D printing system, they obtained successful
The blood is supplied into the brain via 2 major pairs results of neural projections, networks formation, and
of arteries, which are vertebral and internal carotid encapsulation of cortical tissues.106 In a recent study,
arteries. A pair of vertebral arteries merges into the Xu et al created a 3D printed segmental model of intra-
basilar artery, and divided further into the posterior cranial arteries from magnetic resonance angiography
cerebral and superior cerebellar arteries. The internal images as a feasible solution to the intracranial steno-
carotid arteries also split into the middle and anterior sis.107 As noted above, studies performed in 3D printing
cerebral arteries. Ultimately, the internal carotid and of the vascular brain are somewhat limited by now.
basilar arteries converge into the circle of Willis (cere- However, these approaches suggest the potential of 3D
bral arterial circle).79,98 In 3D printing of neural tis- printing as a solid platform with further optimization
sues, one of the most crucial requirements for bioink and investigation to construct a functional brain replace-
selection is the balance of biofunctionality with bio- ment containing vascularized channels (Fig 6).
compatibility.100 102 As neural cells possess compara-
bly high sensibility, bioinks must exhibit cell
recognition sequences to avoid potential immunologi- CONCLUSION AND FUTURE DIRECTION
cal rejection after implantation.101 In addition, bioma- 3D printing as integration of innovative techniques
terials with electrochemically conductive properties, offers remarkable benefits in terms of a vascular net-
such as polypyrrole and poly-3,4-ethyenedioxythio- work formation in tissue and organ. During the past
phene, are worthwhile to consider since interactions years, researchers involved in tissue engineering, mate-
between nerve cells are primarily electrochemical in rial science, and medicine have developed a variety of
nature.101,103,104 Carbon-based biomaterials including distinctive 3D printing techniques and biocompatible
Translational Research
Volume 211 Hann et al 59

Fig 6. (a) A 3D arterial construct of magnetic resonance imaging from a patient. (b) A magnified image of the
highlighted region. (c) A magnetic resonance angiography image and (d) 3D printed model of the indicated
region with arrows. Images are adapted from.107

materials with high-throughput, long-term sustainabil- such as the liver and brain are still multiple steps away
ity, and noncytotoxicity to make the 3D printed plat- from clinical application as of now. In other words,
form functional. 3D printing has especially attracted promotion of sufficient angiogenesis for tissues and
many researchers as a nonsubstitutable technique for organs in a shorter time, as well as enhancement of
the permanent vascular network formation due to its mechanical properties of 3D printing materials for vas-
feasibility, a variety of available printing methods, and cular networks, is the main obstacles. To overcome the
precise controllability. addressed limitations, prospective research on the
The current and prospective research in 3D printed development of new methods, materials, and cells with
vascularization aims to overcome several technical combinations of existing components will be actively
limitations, such as accounting for the dimensions of pursued. Examples of such research developments
blood vessels. For example, capillaries in the human include: the introduction of a novel dual 3D printing
body vary as low as 5 8 mm in diameter. The thick- method, a newly formed biocompatible hydrogel for
ness of the blood vessel walls varies immensely as structural integrity of vascular networks, and support-
well. Therefore, improvements in printing resolution ive novel cell sources to help ECs generate angiogene-
and precise controllability are necessary to generate sis more reliably. Surmounting the current constraints
refined features. The suitable biomaterial selection also listed above in the near future, the 3D printed vascular
enhances resolution, printability, biocompatibility, and network will be considered a future-oriented promising
cell viability as an inequivalent match of biomaterial technology based on its unlimited repeatability after
and cell may result in poor biological functionality. In optimization and punctuality in fabrication.
the particular case of 3D printing with multiple cell
types, the choice of print material is very important.9
In addition, rapid printing speed must be taken into
consideration for use in clinical applications. Despite ACKNOWLEDGMENTS
miniaturization and simplification of native tissues and The authors would like to thank the NIH Director’s
organs, the creation of 3D vasculature still takes a con- New Innovator Award 1DP2EB020549-01 for financial
siderable amount of time. support.
Currently, many researchers contribute to the We confirm that all authors have read the journal’s
improvement of 3D printed vascular networks on a authorship agreement and that the manuscript has been
best effort basis to make the technique more commer- reviewed by and approved by all named authors.
cially useful in the medical industry. As previously Conflicts of Interest: Furthermore, confirm that all
noted, complete 3D printing of some tissues or organs authors have read the journal’s policy on disclosure of
Translational Research
60 Hann et al September 2019

potential conflicts of interest and declare that they have 20. Golden AP, Tien J. Fabrication of microfluidic hydrogels using
no conflicts of interest. molded gelatin as a sacrificial element. Lab Chip 2007;7:
720–5.
21. Lee VK, Kim DY, Ngo H, Lee Y, Seo L, Yoo SS, et al. Creat-
REFERENCES ing perfused functional vascular channels using 3D bio-printing
technology. Biomaterials 2014;35:8092–102.
1. Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, et al. 3D 22. Zhao L, Lee VK, Yoo SS, Dai G, Intes X. The integration of 3-
bioprinting for cardiovascular regeneration and pharmacology. D cell printing and mesoscopic fluorescence molecular tomog-
Adv Drug Deliv Rev 2018;132:252–69. raphy of vascular constructs within thick hydrogel scaffolds.
2. Cui H, Zhu W, Holmes B, Zhang LG. Biologically Inspired Biomaterials 2012;33:5325–32.
smart release system based on 3D bioprinted perfused scaffold 23. Wu W, DeConinck A, Lewis JA. Omnidirectional printing of
for vascularized tissue regeneration. Adv Sci 2016;3:1600058. 3D microvascular networks. Adv Mater 2011;23:H178–83.
3. Cui H, Zhu W, Nowicki M, Zhou X, Khademhosseini A, Zhang 24. Zhang WJ, Liu W, Cui L, Cao Y. Tissue engineering of blood
LG. Hierarchical fabrication of engineered vascularized bone vessel. J Cell Mol Med 2007;11:945–57.
biphasic constructs via dual 3D Bioprinting: integrating 25. Malda J, Visser J, Melchels FP, Jungst T, Hennink WE, Dhert
regional bioactive factors into architectural design. Adv WJ, et al. 25th anniversary article: engineering hydrogels for
Healthc Mater 2016;5:2174–81. biofabrication. Adv Mater 2013;25:5011–28.
4. Zhou X, Castro NJ, Zhu W, Cui H, Aliabouzar M, Sarkar K, 26. Khalil S, Sun W. Bioprinting endothelial cells with alginate for
et al. Improved human bone marrow mesenchymal stem cell 3D tissue constructs. J Biomech Eng 2009;131:111002.
osteogenesis in 3D bioprinted tissue scaffolds with low inten- 27. Yamamoto K, Takahashi T, Asahara T, Ohura N, Sokabe T,
sity pulsed ultrasound stimulation. Sci Rep 2016;6:32876. Kamiya A, et al. Proliferation, differentiation, and tube forma-
5. Cui H, Nowicki M, Fisher JP, Zhang LG. 3D bioprinting for tion by endothelial progenitor cells in response to fluid shear
organ regeneration. Adv Healthc Mater 2017;6(1):1601118. stress. J Appl Physiol 2003;95:2081–8.
6. Griffith LG, Wu B, Cima MJ, Powers MJ, Chaignaud B, 28. Place ES, Evans ND, Stevens MM. Complexity in biomaterials
Vacanti JP. In vitro organogenesis of liver tissuea. Ann N Y for tissue engineering. Nat Mater 2009;8:457–70.
Acad Sci 1997;831:382–97. 29. Di Lullo GA, Sweeney SM, Korkko J, Ala-Kokko L, San Anto-
7. Zein NN, Hanouneh IA, Bishop PD, Samaan M, Eghtesad B, nio JD. Mapping the ligand-binding sites and disease-associated
Quintini C, et al. Three-dimensional print of a liver for preoper- mutations on the most abundant protein in the human, type I
ative planning in living donor liver transplantation. Liver collagen. J Biol Chem 2002;277:4223–31.
Transplant 2013;19:1304–10. 30. Hellio D, Djabourov M. Physically and chemically crosslinked
8. Novosel EC, Kleinhans C, Kluger PJ. Vascularization is the key gelatin gels. Macromol Symposia 2006;241:23–7.
challenge in tissue engineering. Adv Drug Deliv Rev 31. Kuijpers AJ, van Wachem PB, van Luyn MJ, Brouwer LA,
2011;63:300–11. Engbers GH, Krijgsveld J, et al. In vitro and in vivo evaluation
9. Paulsen S, Miller J. Tissue vascularization through 3D printing: of gelatin-chondroitin sulphate hydrogels for controlled release
will technology bring us flow? Dev Dyn 2015;244:629–40. of antibacterial proteins. Biomaterials 2000;21:1763–72.
10. Jose RR, Rodriguez MJ, Dixon TA, Omenetto F, Kaplan DL. 32. Rowe SL, Stegemann JP. Microstructure and mechanics of col-
Evolution of bioinks and additive manufacturing technologies lagen-fibrin matrices polymerized using ancrod snake venom
for 3D bioprinting. ACS Biomater Sci Eng 2016;2:1662–78. enzyme. J Biomech Eng 2009;131:061012.
11. Hull CW. Apparatus for production of three-dimensional 33. Rao RR, Peterson AW, Ceccarelli J, Putnam AJ, Stegemann JP.
objects by stereolithography. Google Patents; 1986. Matrix composition regulates three-dimensional network for-
12. Patra S, Young V. A review of 3D printing techniques and the mation by endothelial cells and mesenchymal stem cells in col-
future in biofabrication of bioprinted tissue. Cell Biochem Bio- lagen/fibrin materials. Angiogenesis 2012;15:253–64.
phys 2016;74:93–8. 34. Kleinman HK, Martin GR. Matrigel: basement membrane
13. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat matrix with biological activity. Semin Cancer Biol
Biotechnol 2014;32:773–85. 2005;15:378–86.
14. Ozbolat IT, Hospodiuk M. Current advances and future perspec- 35. Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex pro-
tives in extrusion-based bioprinting. Biomaterials 2016;76:321–43. tein mixture required for optimal growth of cell culture. Proteo-
15. Suri S, Han LH, Zhang W, Singh A, Chen S, Schmidt CE. Solid mics 2010;10:1886–90.
freeform fabrication of designer scaffolds of hyaluronic acid for 36. Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels
nerve tissue engineering. Biomed Microdevices 2011;13:983–93. as synthetic extracellular matrix materials. Biomaterials
16. Lin H, Zhang D, Alexander PG, Yang G, Tan J, Cheng AW, 1999;20:45–53.
et al. Application of visible light-based projection stereolithog- 37. Augst AD, Kong HJ, Mooney DJ. Alginate hydrogels as bioma-
raphy for live cell-scaffold fabrication with designed architec- terials. Macromol Biosci 2006;6:623–33.
ture. Biomaterials 2013;34:331–9. 38. Kogan G, Soltes L, Stern R, Gemeiner P. Hyaluronic acid: a
17. Laschke MW, Rucker M, Jensen G, Carvalho C, Mulhaupt R, natural biopolymer with a broad range of biomedical and indus-
Gellrich NC, et al. Incorporation of growth factor containing trial applications. Biotechnol Lett 2007;29:17–25.
Matrigel promotes vascularization of porous PLGA scaffolds. J 39. Ouyang L, Highley CB, Rodell CB, Sun W, Burdick JA. 3D
Biomed Mater Res A 2008;85:397–407. printing of shear-thinning hyaluronic acid hydrogels with sec-
18. Kinstlinger IS, Miller JS. 3D-printed fluidic networks as vascu- ondary cross-linking. ACS Biomater Sci Eng 2016;2:1743–51.
lature for engineered tissue. Lab Chip 2016;16:2025–43. 40. Johansson B. Agarose gel electrophoresis. Scand J Clin Lab
19. Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Invest 1972;29(sup124):7–19.
Shue H-J, et al. Three-dimensional printing of complex biologi- 41. Oliveira JT, Reis R. Polysaccharide based materials for carti-
cal structures by freeform reversible embedding of suspended lage tissue engineering applications. J Tissue Eng Regen Med
hydrogels. Sci Adv 2015;1:e1500758. 2011;5:421–36.
Translational Research
Volume 211 Hann et al 61

42. Sakai S, Hashimoto I, Kawakami K. Agarose-gelatin conjugate 62. Nemeno-Guanzon JG, Lee S, Berg JR, Jo YH, Yeo JE, Nam
for adherent cell-enclosing capsules. Biotechnol Lett 2007;29: BM, et al. Trends in tissue engineering for blood vessels. J
731–5. Biomed Biotechnol 2012;2012:956345.
43. Freier T, Koh HS, Kazazian K, Shoichet MS. Controlling cell 63. Bose S, Roy M, Bandyopadhyay A. Recent advances in bone tis-
adhesion and degradation of chitosan films by N-acetylation. sue engineering scaffolds. Trends Biotechnol 2012;30:546–54.
Biomaterials 2005;26:5872–8. 64. Wang J, Yang M, Zhu Y, Wang L, Tomsia AP, Mao C. Phage
44. da Silva RM, Lopez Perez PM, Elvira C, Mano JF, Roman JS, nanofibers induce vascularized osteogenesis in 3D printed bone
Reis RL. Poly (N isopropylacrylamide) surface grafted chito- scaffolds. Adv Mater 2014;26:4961–6.
san membranes as a new substrate for cell sheet engineering 65. Tsigkou O, Pomerantseva I, Spencer JA, Redondo PA, Hart
and manipulation. Biotechnol Bioeng 2008;101:1321–31. AR, O’Doherty E, et al. Engineered vascularized bone grafts.
45. Malafaya P, Pedro A, Peterbauer A, Gabriel C, Redl H, Reis R. Proc Natl Acad Sci 2010;107:3311–6.
Chitosan particles agglomerated scaffolds for cartilage and 66. Santos MI, Reis RL. Vascularization in bone tissue engineer-
osteochondral tissue engineering approaches with adipose tissue ing: physiology, current strategies, major hurdles and future
derived stem cells. J Mater Sci Mater Med 2005;16:1077–85. challenges. Macromol Biosci 2010;10:12–27.
46. Pati F, Jang J, Ha DH, Won Kim S, Rhie JW, Shim JH, et al. 67. Moore WR, Graves SE, Bain GI. Synthetic bone graft substi-
Printing three-dimensional tissue analogues with decellularized tutes. ANZ J Surg 2001;71:354–61.
extracellular matrix bioink. Nat Commun 2014;5:3935. 68. Holmes B, Bulusu K, Plesniak M, Zhang LG. A synergistic
47. Kim BS, Kwon YW, Kong JS, Park GT, Gao G, Han W, et al. approach to the design, fabrication and evaluation of 3D printed
3D cell printing of in vitro stabilized skin model and in vivo micro and nano featured scaffolds for vascularized bone tissue
pre-vascularized skin patch using tissue-specific extracellular repair. Nanotechnology 2016;27:064001.
matrix bioink: a step towards advanced skin tissue engineering. 69. Temple JP, Hutton DL, Hung BP, Huri PY, Cook CA, Kondra-
Biomaterials 2018;168:38–53. gunta R, et al. Engineering anatomically shaped vascularized
48. Kim BS, Kim H, Gao G, Jang J, Cho DW. Decellularized extra- bone grafts with hASCs and 3D-printed PCL scaffolds. J
cellular matrix: a step towards the next generation source for Biomed Mater ResA 2014;102:4317–25.
bioink manufacturing. Biofabrication 2017;9:034104. 70. Yan Y, Chen H, Zhang H, Guo C, Yang K, Chen K, et al. Vas-
49. Jang J, Kim TG, Kim BS, Kim S-W, Kwon S-M, Cho D-W. cularized 3D printed scaffolds for promoting bone regeneration.
Tailoring mechanical properties of decellularized extracellular Biomaterials 2019;190:97–110.
matrix bioink by vitamin B2-induced photo-crosslinking. Acta 71. Donneys A, Weiss DM, Deshpande SS, Ahsan S, Tchanque-
biomaterialia 2016;33:88–95. Fossuo CN, Sarhaddi D, et al. Localized deferoxamine injection
50. Gopinathan J, Noh I. Recent trends in bioinks for 3D printing. augments vascularity and improves bony union in pathologic
Biomater Res 2018;22:11. fracture healing after radiotherapy. Bone 2013;52:318–25.
51. Garlotta D. A literature review of poly (lactic acid). J Poly 72. Potier E, Ferreira E, Dennler S, Mauviel A, Oudina K,
Environ 2001;9:63–84. Logeart Avramoglou D, et al. Desferrioxamine driven upre-
52. Guvendiren M, Molde J, Soares RM, Kohn J. Designing bioma- gulation of angiogenic factor expression by human bone mar-
terials for 3D printing. ACS Biomater Sci Eng 2016;2:1679–93. row stromal cells. J Tissue Eng Regen Med 2008;2:272–8.
53. Serra T, Mateos-Timoneda MA, Planell JA, Navarro M. 3D 73. Liu X, Jakus AE, Kural M, Qian H, Engler A, Ghaedi M, et al.
printed PLA-based scaffolds: a versatile tool in regenerative Vascularization of natural and synthetic bone scaffolds. Cell
medicine. Organogenesis 2013;9:239–44. Transplant 2018;27:1269–80.
54. Lee H, Cho DW. One-step fabrication of an organ-on-a-chip 74. Merceron TK, Burt M, Seol YJ, Kang HW, Lee SJ, Yoo JJ,
with spatial heterogeneity using a 3D bioprinting technology. et al. A 3D bioprinted complex structure for engineering the
Lab Chip 2016;16:2618–25. muscle-tendon unit. Biofabrication 2015;7:035003.
55. Ouyang L, Highley CB, Sun W, Burdick JA. A generalizable 75. Levenberg S, Rouwkema J, Macdonald M, Garfein ES, Kohane
strategy for the 3D bioprinting of hydrogels from nonviscous DS, Darland DC, et al. Engineering vascularized skeletal mus-
photo-crosslinkable inks. Adv Mater 2017;29(8):1604983. cle tissue. Nat Biotechnol 2005;23:879–84.
56. Melchiorri AJ, Hibino N, Best CA, Yi T, Lee YU, Kraynak CA, 76. Zisch AH, Lutolf MP, Hubbell JA. Biopolymeric delivery
et al. 3D-printed biodegradable polymeric vascular grafts. Adv matrices for angiogenic growth factors. Cardiovasc Pathol
Healthc Mater 2016;5:319–25. 2003;12:295–310.
57. Bohorquez M, Koch C, Trygstad T, Pandit N. A study of the 77. Von Degenfeld G, Banfi A, Springer ML, Blau HM. Myo-
temperature-dependent micellization of pluronic F127. J Col- blast mediated gene transfer for therapeutic angiogenesis and
loid Interface Sci 1999;216:34–40. arteriogenesis. Br J Pharmacol 2003;140:620–6.
58. Mandrycky C, Wang Z, Kim K, Kim D-H. 3D bioprinting for 78. Lu Y, Shansky J, Del Tatto M, Ferland P, Wang X, Vanden-
engineering complex tissues. Biotechnol Sdv 2016;34:422–34. burgh H. Recombinant vascular endothelial growth factor
59. Higuchi A, Ling Q-D, Kumar SS, Chang Y, Alarfaj AA, secreted from tissue-engineered bioartificial muscles promotes
Munusamy MA, et al. Physical cues of cell culture materials localized angiogenesis. Circulation 2001;104:594–9.
lead the direction of differentiation lineages of pluripotent stem 79. Betts JG. Anatomy and physiology. Houston, Texas: OpenStax
cells. J Mater Chem B 2015;3:8032–58. College, Rice University; 2013.
60. Badorff C, Brandes RP, R Popp, Rupp S, Urbich C, Aicher A, 80. Kumbar SG, Nukavarapu SP, James R, Nair LS, Laurencin CT.
et al. Transdifferentiation of blood-derived human adult endo- Electrospun poly(lactic acid-co-glycolic acid) scaffolds for skin
thelial progenitor cells into functionally active cardiomyocytes. tissue engineering. Biomaterials 2008;29:4100–7.
Circulation 2003;107:1024–32. 81. Michael S, Sorg H, Peck C-T, Koch L, Deiwick A, Chichkov B,
61. Singh A, Singh A, Sen D. Mesenchymal stem cells in cardiac et al. Tissue engineered skin substitutes created by laser-
regeneration: a detailed progress report of the last 6 years assisted bioprinting form skin-like structures in the dorsal skin
(2010 2015). Stem Cell Res Ther 2016;7:82. fold chamber in mice. PloS One 2013;8:e57741.
Translational Research
62 Hann et al September 2019

82. Skardal A, Mack D, Kapetanovic E, Atala A, Jackson JD, Yoo J, 104. Heo DN, Acquah N, Kim J, Lee S-J, Castro NJ, Zhang LG.
et al. Bioprinted amniotic fluid-derived stem cells accelerate healing Directly induced neural differentiation of human adipose-
of large skin wounds. Stem Cells Transl Med 2012;1:792–802. derived stem cells using three-dimensional culture system of
83. Bibb R, Nottrodt N, Gillner A. Artificial vascularised scaffolds conductive microwell with electrical stimulation. Tissue Eng A
for 3D-tissue regeneration a perspective of the ArtiVasc 3D 2018;24:537–45.
Project. Int J Bioprint 2016;2(1):93–102. 105. Naftulin JS, Kimchi EY, Cash SS. Streamlined, inexpensive 3D
84. Ballaun C, Weninger W, Uthman A, Weich H, Tschachler E. printing of the brain and skull. PloS One. 2015;10:e0136198.
Human keratinocytes express the three major splice forms of vas- 106. Lozano R, Stevens L, Thompson BC, Gilmore KJ, Gorkin 3rd
cular endothelial growth factor. J Invest Dermatol 1995;104:7–10. R, Stewart EM, et al. 3D printing of layered brain-like struc-
85. Thom T, Haase N, Rosamond W, Howard VJ, Rumsfeld J, tures using peptide modified gellan gum substrates. Biomateri-
Manolio T, et al. Heart disease and stroke statistics 2006 als 2015;67:264–73.
update: a report from the American Heart Association Statistics 107. Xu WH, Liu J, Li ML, Sun ZY, Chen J, Wu JH. 3D printing of
Committee and Stroke Statistics Subcommittee. Circulation intracranial artery stenosis based on the source images of mag-
2006;113:e85–e151. netic resonance angiograph. Ann Transl Med 2014;2:74.
86. Tomanek RJ, Runyan RB. Formation of the heart and its regula- 108. Landers R, H€ubner U, Schmelzeisen R, M€ulhaupt R. Rapid pro-
tion. Springer Science & Business Media; 2012. totyping of scaffolds derived from thermoreversible hydrogels
87. Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen D-HT, and tailored for applications in tissue engineering. Biomaterials
Cohen DM, et al. Rapid casting of patterned vascular networks 2002;23:4437–47.
for perfusable engineered three-dimensional tissues. Nat Mater 109. Elvin C, Danon S, Brownlee A, White J, Hickey M, Liyou N,
2012;11:768. et al. Evaluation of photo crosslinked fibrinogen as a rapid
88. Jia W, Gungor-Ozkerim PS, Zhang YS, Yue K, Zhu K, Liu W, and strong tissue adhesive. J Biomed Mater Res A
et al. Direct 3D bioprinting of perfusable vascular constructs 2010;93:687–95.
using a blend bioink. Biomaterials 2016;106:58–68. 110. Khunmanee S, Jeong Y, Park H. Crosslinking method of hya-
89. Cui X, Boland T. Human microvasculature fabrication using ther- luronic-based hydrogel for biomedical applications. J Tissue
mal inkjet printing technology. Biomaterials 2009;30:6221–7. Eng 2017;8:2041731417726464.
90. Zhang YS, Arneri A, Bersini S, Shin SR, Zhu K, Goli-Maleka- 111. Childers EP, Wang MO, Becker ML, Fisher JP, Dean D. 3D
badi Z, et al. Bioprinting 3D microfibrous scaffolds for engi- printing of resorbable poly (propylene fumarate) tissue engi-
neering endothelialized myocardium and heart-on-a-chip. neering scaffolds. MRS Bulletin 2015;40:119–26.
Biomaterials 2016;110:45–59. 112. Chu CR, Coutts RD, Yoshioka M, Harwood FL, Monosov AZ,
91. Duan B. State-of-the-art review of 3D bioprinting for cardiovascular Amiel D. Articular cartilage repair using allogeneic perichondrocy-
tissue engineering. Ann Biomed Eng 2017;45:195–209. teseeded biodegradable porous polylactic acid (PLA): a tissue-
92. Seol YJ, Kang HW, Lee SJ, Atala A, Yoo JJ. Bioprinting tech- engineering study. J Biomed Mater Res 1995;29:1147–54.
nology and its applications. Eur J Cardiothorac Surg 113. Cutright DE, Hunsuck EE. Tissue reaction to the biodegradable
2014;46:342–8. polylactic acid suture. Oral Surg Oral Med Oral Pathol
93. Borovjagin AV, Ogle BM, Berry JL, Zhang J. From microscale 1971;31:134–9.
devices to 3D printing: advances in fabrication of 3D cardiovas- 114. Kweon H, Yoo MK, Park IK, Kim TH, Lee HC, Lee H-S, et al.
cular tissues. Circ Res 2017;120:150–65. A novel degradable polycaprolactone networks for tissue engi-
94. Taub R. Liver regeneration: from myth to mechanism. Nat Rev neering. Biomaterials 2003;24:801–8.
Mol Cell Biol 2004;5:836. 115. Lee K-W, Wang S, Lu L, Jabbari E, Currier BL, Yaszemski MJ.
95. Wang X, Yan Y, Pan Y, Xiong Z, Liu H, Cheng J, et al. Genera- Fabrication and characterization of poly (propylene fumarate)
tion of three-dimensional hepatocyte/gelatin structures with scaffolds with controlled pore structures using 3-dimensional
rapid prototyping system. Tissue Eng 2006;12:83–90. printing and injection molding. Tissue Eng 2006;12:2801–11.
96. Robbins JB, Gorgen V, Min P, Shepherd BR, Presnell SC. A 116. Tessmar JK, G€opferich AM. Customized PEG derived copoly-
novel in vitro three-dimensional bioprinted liver tissue system mers for tissue engineering applications. Macromol Biosci
for drug development. FASEB J 2013;27:812. 2007;7:23–39.
97. Gu X. Progress and perspectives of neural tissue engineering. 117. Wang Z, Abdulla R, Parker B, Samanipour R, Ghosh S, Kim K.
Front Med 2015;9:401–11. A simple and high-resolution stereolithography-based 3D bio-
98. Netter FH. Atlas of human anatomy E-book. Elsevier Health printing system using visible light crosslinkable bioinks. Bio-
Sciences; 2017. fabrication 2015;7:045009.
99. Abbott NJ. Astrocyte endothelial interactions and blood - 118. Weisel JW. Fibrinogen and fibrin. Adv Protein Chem
brain barrier permeability. J Anat 2002;200:629–38. 2005;70:247–99.
100. Lee SJ, Zhu W, Nowicki M, Lee G, Heo DN, Kim J, et al. 3D 119. Gao Q, Hu B, Ning Q, Ye C, Xie J, Ye J, et al. A primary study
printing nano conductive multi-walled carbon nanotube scaf- of poly (propylene fumarate) 2-hydroxyethyl methacrylate
folds for nerve regeneration. J Neural Eng 2018;15:016018. copolymer scaffolds for tarsal plate repair and reconstruction in
101. Lee S-J, Esworthy T, Stake S, Miao S, Zuo YY, Harris BT, rabbit eyelids. J Mater Chem B 2015;3:4052–62.
et al. Advances in 3D bioprinting for neural tissue engineering. 120. Lee Y, Chung HJ, Yeo S, Ahn C-H, Lee H, Messersmith PB,
Adv Biosys 2018;2:1700213. et al. Thermo-sensitive, injectable, and tissue adhesive sol gel
102. Lee SJ, Nowicki M, Harris B, Zhang LG. Fabrication of a highly transition hyaluronic acid/pluronic composite hydrogels pre-
aligned neural scaffold via a table top stereolithography 3D printing pared from bio-inspired catechol-thiol reaction. Soft Matter
and electrospinning. Tissue Eng A 2017;23:491–502. 2010;6:977–83.
103. Shi Z, Gao H, Feng J, Ding B, Cao X, Kuga S, et al. In situ syn- 121. Suggs LJ, Kao EY, Palombo LL, Krishnan RS, Widmer MS,
thesis of robust conductive cellulose/polypyrrole composite Mikos AG. Preparation and characterization of poly (propylene
aerogels and their potential application in nerve regeneration. fumarate-co-ethylene glycol) hydrogels. J Biomater Sci Polym
Angew Chem Int Ed Engl 2014;53:5380–4. Ed 1998;9:653–66.
Translational Research
Volume 211 Hann et al 63

122. Suggs LJ, Krishnan RS, Garcia CA, Peter SJ, Anderson JM, adipose stem cells in 3D printed chitosan scaffold. PloS One
Mikos AG. In vitro and in vivo degradation of poly (propylene 2014;9:e99410.
fumarate co ethylene glycol) hydrogels. J Biomed Mater Res 125. Daamen WF, Veerkamp J, Van Hest J, Van Kuppevelt T. Elastin as
1998;42:312–20. a biomaterial for tissue engineering. Biomaterials 2007;28:4378–98.
123. Bertassoni LE, Cecconi M, Manoharan V, Nikkhah M, Hjort- 126. Kirchmajer DM, Gorkin III R. An overview of the suitability of
naes J, Cristino AL, et al. Hydrogel bioprinted microchannel hydrogel-forming polymers for extrusion-based 3D-printing. J
networks for vascularization of tissue engineering constructs. Mater Chem B 2015;3:4105–17.
Lab Chip 2014;14:2202–11. 127. Fan R, Piou M, Darling E, Cormier D, Sun J, Wan J. Bio-print-
124. Ye K, Felimban R, Traianedes K, Moulton SE, Wallace GG, ing cell-laden Matrigel agarose constructs. J Biomater Appl
Chung J, et al. Chondrogenesis of infrapatellar fat pad derived 2016;31:684–92.

You might also like