You are on page 1of 14

Review Article

Cells Tissues Organs Received: September 5, 2020


Accepted: October 27, 2020
DOI: 10.1159/000512792 Published online: March 5, 2021

3D Bioprinted Cardiac Tissues and


Devices for Tissue Maturation
Veronika Sedlakova a, b Christopher McTiernan a David Cortes a Erik
J. Suuronen a Emilio I. Alarcon a, c
aBEaTS
Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada;
b Department
of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic;
cDepartment of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa,

Ottawa, ON, Canada

Keywords that match those from the native heart. Finally, we also dis-
3D printing · Cardiac tissue · Valve · Maturation · Bioreactor cuss the suitability of this technology in the design and fab-
rication of custom-made devices intended for the matura-
tion of the cardiac tissue, a process that has been shown to
Abstract increase the viability of implants. Altogether this review
Cardiovascular diseases are the leading cause of mortality shows that 3D printing and bioprinting are versatile and
worldwide. Given the limited endogenous regenerative ca- highly modulative technologies with wide applications in
pabilities of cardiac tissue, patient-specific anatomy, chal- cardiac regeneration and beyond. © 2021 S. Karger AG, Basel
lenges in treatment options, and shortage of donor tissues
for transplantation, there is an urgent need for novel ap-
proaches in cardiac tissue repair. 3D bioprinting is a technol-
ogy based on additive manufacturing which allows for the Introduction
design of precisely controlled and spatially organized struc-
tures, which could possibly lead to solutions in cardiac tissue Accounting for over 17 million deaths in 2016 [World
repair. In this review, we describe the basic morphological Health Organization, 2018], cardiovascular diseases
and physiological specifics of the heart and cardiac tissues (CVDs) are the number one cause of mortality world-
and introduce the readers to the fundamental principles un- wide. At the top of the list is ischemic heart disease, fol-
derlying 3D printing technology and some of the materials/ lowed by other conditions such as rheumatic heart dis-
approaches which have been used to date for cardiac repair. ease and various cardiomyopathies. Current treatments
By summarizing recent progress in 3D printing of cardiac tis- for CVD encompass a wide variety of pharmacological
sue and valves with respect to the key features of cardiovas- and lifestyle interventions [Metra and Teerlink, 2017;
cular tissue (such as contractility, conductivity, and vascular- Waller and Sampson, 2018; Watkins et al., 2018]. How-
ization), we highlight how 3D printing can facilitate surgical ever, if the damage to the heart is too severe, surgical in-
planning and provide custom-fit implants and properties tervention may be the only option, including coronary
130.209.6.61 - 8/12/2021 3:25:54 PM

karger@karger.com © 2021 S. Karger AG, Basel Emilio I. Alarcon


www.karger.com/cto Department of Biochemistry, Microbiology, and Immunology
Faculty of Medicine, University of Ottawa
40 Ruskin St, Rm H5229, Ottawa, ON K1Y 4W7 (Canada)
Glasgow Univ.Lib.
Downloaded by:

ealarcon @ ottawaheart.ca
Biomaterial/additives

Cells
In vitro maturation

Bioink u
sit
Ex
In vivo implantation
In
sit
u

3D bioprinter 3D printed tissue

Fig. 1. Possible strategies for the fabrication


of functional 3D bioprinted cardiac tissue.
Created with BioRender.com.

artery bypass grafting, valve replacement and, in the case 3D printing has allowed for the building of complex
of highly advanced disease, heart transplant. The urgency 3-dimensional structures [Birla and Williams, 2020].
for developing therapeutic approaches for cardiac tissue While 3D printing has taken on many forms over the
is multifactorial and includes (1) limited endogenous re- years, bioprinting, which is a specific subdiscipline of 3D
generative potential of cardiac tissue, (2) patient-specific printing that employs bioinks for the printing of artificial
anatomical requirements, and (3) shortages of donor tis- tissues and tissue-engineered constructs, has rapidly
sue for transplantation [Prabhu and Frangogiannis, 2016; gained momentum in recent years. Bioinks are typically
Taylor et al., 2018]. either cellular (containing biomaterial and cells) or acel-
In order to address the needs in cardiac tissue repair, lular (with cells seeded post-printing) [Hospodiuk et al.,
the field of tissue engineering has pushed to develop a 2017; Deo et al., 2020], and there are examples of bioma-
number of cell- and biomaterial-based therapies [Nguyen terial-free 3D bioprinting, which have employed cells
et al., 2019]. Although there are only a few examples of only [Ong et al., 2017; Yeung et al., 2019].
stem cell transplant therapies which have been shown 3D printing has been employed in the bioprinting of
successful in regard to cardiac repair in clinical trials [Ba- models, tissues, and organs, as well as in the development
nerjee et al., 2018], recent advances in engineered tissues of custom devices for tissue maturation. The various
presenting improved maturity [Ronaldson-Bouchard et modes of bioprinting can be subdivided into 2 main
al., 2018], electrical integration [Tandon et al., 2009], and groups:
lesser immune response [Boehler et al., 2011] hold great (1) Ex situ bioprinting where the material is printed out-
promise that in the future functional tissue constructs, side the host, matured, and then implanted [Gaetani et
such as myocardial tissue, valves, and even whole hearts al., 2015; Gao et al., 2017]; and
will provide an alternative to the current donor organ (2) In vivo bioprinting, where the material is printed di-
transplant model. However, in order to fabricate implants rectly in situ [Di Bella et al., 2018; Cheng et al., 2020]
that best recapitulate the physical form, conductivity, and within the host.
contractility of native cardiac tissue, advanced and scal- A simple scheme showing the possible routes to 3D
able fabrication techniques are required. One fabrication bioprinted cardiac tissue is shown in Figure 1. As the ma-
technique that holds great promise in this regard is 3D jority of 3D bioprinting techniques are derived from the
bioprinting. With the field of bioprinting coming of age more classical polymer/plastic and resin-based printing
and the advent of several new modes of printing, better techniques, many of the principles and components un-
cell survival, multi-material printing, and increased reso- derlying the printing process can be similarly classified.
lution [Dey and Ozbolat, 2020], the bioprinting of car- In general, 3D bioprinting can be thought of as a combi-
diac tissue seems poised to make an impact on the way nation of the top-down and bottom-up routes to the bio-
cardiac disease is treated. fabrication of tissues [Shtein and Shoseyov, 2017]. Tradi-
tionally the approach to the fabrication of artificial tissues
130.209.6.61 - 8/12/2021 3:25:54 PM

2 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
has taken a top-down approach whereby cells and signal- cardiac tissue components. While the ultimate goal is the
ing molecules are introduced within a 3D biomaterial, fabrication of a whole functioning heart, advances must
which can act as a scaffold that mimics the structure of be made to improve the vascularization of the constructs.
the extracellular tissue environment. More recently, there When considering that the diffusion limit of oxygen is
has been an interest in bottom-up approaches, in which approximately 100–200 μm from the nearest blood vessel
larger cellular structures such as spheroids and cell-sheets [Carmeliet and Jain, 2000], there is a need to incorporate
are prepared through self-assembly and then used to as- micro- and smaller macro-vessels into 3D constructs to
semble larger and more complex structures [Lee et al., provide the cells with vessel-like networks within this dis-
2017]. In 3D printing of tissues and organs, both the top- tance. Without such a vascular network, cell survival and
down and bottom-up approaches are combined. Bioinks physiological function can be significantly impaired, thus
comprising synthetic, natural, and mimetic polymers limiting the ability to fabricate functional implantable
along with cells, and various growth factors are deposited structures that more closely recapitulate a full thickness
in such a way as to prepare 3D structures that are not only heart muscle.
able to structurally mimic the tissue and organs but are Cardiac tissue is composed of cardiomyocytes sur-
also able to recapitulate physiological functions post- rounded by cardiac interstitium [Ross and Pawlina, 2006;
maturation. Herein, we will describe some of the different Laflamme et al., 2012]. Cardiomyocytes are cells capable
approaches and techniques that have been employed in of contraction (contain precisely organized sarcomeres
the fabrication of 3D printed cardiac tissue. with actin and myosin) and are interconnected by spe-
cific structures called intercalated discs. These intercalat-
ed discs are comprised of desmosome junctions that pro-
Cardiac Tissue Biology vide strength, and gap junctions (composed of connex-
ins) for the conduction of electrical impulses between
Before jumping into the 3D printing of cardiac tissue, cells. As a result, correct development and maturation of
it is necessary to have an understanding of the morphol- sarcomeres and intercellular junctions within cardiomy-
ogy, key features, and characteristics of cardiovascular ocytes in vitro is crucial for any in vivo tissue engineering
tissue, as these properties are critical in the design and application [Feric and Radisic, 2016]. Cardiac intersti-
fabrication of functional biomimetic structures. tium, the material surrounding the cardiomyocytes, is
The cardiovascular system is comprised of 2 key com- comprised of cardiac extracellular matrix (ECM) and var-
ponents, the heart and blood vessels [Ross and Pawlina, ious other cells (e.g., macrophages, fibroblasts, endothe-
2006; Laflamme et al., 2012], where the blood vessels (ar- lial cells) [Ross and Pawlina, 2006; Laflamme et al., 2012].
teries, veins, and capillaries) serve as transport elements, In the native healthy ECM, glycoproteins and proteogly-
and the heart acts as a pump to circulate the blood cans surround collagen type I (∼80%), collagen type III
throughout the body. For the heart to function in that ca- (∼10%), elastin, and collagens type IV, V, and VI [Jour-
pacity, it requires certain morphological and functional dan-LeSaux et al., 2010; Shamhart and Meszaros, 2010;
traits. For example, it contains cardiac muscle capable of Horn and Trafford, 2016]. Knowledge of cardiac ECM
contraction, specific cardiomyocytes that provide intrin- composition is vital in selecting an appropriate biomate-
sic pacemaker capacity, along with a fibrous skeleton and rial for 3D printing, as the best biomaterial should pro-
valves (2 atrioventricular and 2 semilunar) that allow for vide bioactive cues to the cells, be biocompatible and bio-
unidirectional flow. Histologically, from inside out the degradable, and display physicochemical properties that
heart is composed of an endocardium, myocardium (car- match those found in vivo at the site of implantation in
diac muscle tissue), and epicardium, and is surrounded order to withstand the demanding pressure and volume
by the pericardial sac. The constant need for oxygenation, load present in the heart.
nutrient supply, and waste product removal from the Heart valves are composed of dense irregular connec-
muscle can become a challenge if the coronary vascula- tive tissue, allowing them to withstand the flow of blood,
ture is unhealthy, which can lead to a loss of metabolic and are covered with endocardium [Ross and Pawlina,
homeostasis. 2006; Laflamme et al., 2012]. While the avascular nature
If the balance is affected, cells may start failing in their of the valve leaflets would appear to simplify the design
function, which marks the onset for CVDs. The current and manufacture of biosynthetic implants, difficulties
focus of 3D bioprinting for the cardiovascular system has arise when trying to produce strong yet flexible valves that
been largely concentrated on the fabrication of valves and display a nonthrombogenic surface [Dangas et al., 2016].
130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 3


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
Given the limited capacity of cardiomyocyte regenera- other issue that must be addressed is cell survival and re-
tion in humans, damaged heart tissue is substituted with tention at the site of implantation, as well as immunoge-
a collagenous scar upon injury [Bergmann et al., 2009; nicity [Huang NF et al., 2018; He and Chen, 2020]. Fol-
Steenbergen and Frangogiannis, 2012]. While this fibrotic lowing implantation, improper electrical coupling
region significantly hinders normal cardiac function between engineered and native tissue can cause life-
[Steenbergen and Frangogiannis, 2012], several cell-based threatening arrhythmias [Huang NF et al., 2018]. Lastly,
tissue engineering approaches have been developed to regulatory policies on research and clinical application of
limit scar formation and allow the tissue to retain its elas- stem cells are different all over the world, and this may
ticity. These cell-based therapies have been applied either restrict their therapeutic application [Gopalan et al.,
alone (single cell suspensions, cell sheets, spheroids) or in 2018].
combination with biomaterials (injectable hydrogels, The cell types that have most commonly been used in
patches, 3D printed scaffolds) [Atala, 2004; He and Chen, the design of 3D printed cardiac and vascular tissue in-
2020]. Typically, cell-based therapies for cardiac applica- clude primary cardiomyocytes, CPCs, cardiac fibroblasts,
tion employ human embryonic stem cells (hESCs), hu- hiPSC-derived cardiomyocytes, smooth muscle cells, en-
man induced pluripotent stem cells (hiPSCs), mesenchy- dothelial cells, hESC-derived cardiomyocytes, valvular
mal stromal cells, hematopoietic stem cells, cardiac stem interstitial cells, and human umbilical vein endothelial
cells, cardiac progenitor cells (CPCs), primary cardiomy- cells (HUVECs) (Table 1) [Duan et al., 2013; Gao et al.,
ocytes, skeletal myoblasts, fibroblasts, or endothelial pro- 2017; Bejleri et al., 2018; Wang et al., 2018; Lee et al.,
genitor cells [Jover et al., 2018; Müller et al., 2018; Wang 2019]. As for the biomaterials, the most commonly used
et al., 2018; He and Chen, 2020]. Despite the seemingly include collagens, gelatin, alginate, hyaluronic acid, and
large number of available cell types, there are many limita- decellularized cardiac ECM [Pati et al., 2014; Gaetani et
tions to these cells, which represents a major bottleneck al., 2015; Zhang et al., 2016; Izadifar et al., 2017]. To in-
toward cardiac tissue engineering [Huang NF et al., 2018; crease electroconductivity of biomaterials, carbon nano-
Müller et al., 2018; He and Chen, 2020]. One of the first tubes or gold nanorods have been used [Ho et al., 2017;
issues to arise relates to difficulties obtaining a clinically Zhu et al., 2017; Hosoyama et al., 2018], as well as other
relevant number of cells [Huang NF et al., 2018]. In par- substances that have been shown to increase electrocon-
ticular, primary cells have limited proliferative potential ductivity, such as graphene oxide, aniline, or polypyrrole
in vitro before entering senescence [Kengla et al., 2017], [Sedlakova et al., 2019].
while differentiation protocols for hESCs and hiPSCs and
other stem/stromal cells struggle with yield and purity of
the differentiated target cell type [He and Chen, 2020]. Fundamental Concepts of 3D Printing
Moreover, undifferentiated cell populations may pose a
risk of forming teratomas or tumors [He and Chen, 2020]. 3D printing, also known as additive manufacturing, is
The second major obstacle that must be addressed relates the process through which 3D objects are created from
to difficulties in obtaining cells of sufficient maturity. For 3D models using computer-aided design (CAD) software
example, surrogate cells may have a more amorphous or from medical imaging devices (magnetic resonance
shape compared to the rectangular one found in mature imaging [MRI], computerized axial tomography scan,
cardiomyocytes. Further, these cells may lack proper sar- functional MRI, among others). Whether one is interest-
comere organization and alignment, as well as calcium ed in printing plastic models and devices or bioprinting
handling mechanisms, all key properties for efficient con- functional tissues and organs, one needs to begin with a
tractile function. Metabolism and transcriptional profile digital 3D model of the object to be printed, or a pattern
also change with maturation, and such immaturity could to be recapitulated, as it is this model or pattern that pro-
influence successful engraftment. Many approaches have vides the cartesian coordinates as to where the printed
been proposed to promote maturation; these include the material is to be deposited or crosslinked. While there are
use of hormones, mechanical strain stimulation, electrical a number of software suites available for CAD design
stimulation, patterned surfaces and substrate stiffness (6– (both free and proprietary), in order to generate biomi-
10 kPa), and interactions with other cells or ECM [Huang metic tissues and organs that can imitate the features and
NF et al., 2018; Ahmed et al., 2020; Guo and Pu, 2020; He functions of the desired targets, it is necessary to intro-
and Chen, 2020]. However, even with these methods, cell duce advanced imaging modalities within the 3D model-
maturation into adult cardiomyocytes is still poor. An- ing process [Squelch, 2018]. Through the use of image
130.209.6.61 - 8/12/2021 3:25:54 PM

4 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
Table 1. 3D bioprinted cardiac tissue, implants, and components of the heart

Target tissue Bioink Cells used In vivo model Achievement Reference


or implant
Cardiac Fibrin Primary rat cardiomyocytes N/A Drug testing Wang et al. 2018)
Cardiac Gelatin Human mesenchymal stem cells N/A Enzymatic crosslinking Tijore et al. 2018)
Cardiac patch Hyaluronic acid; Human cardiac-derived progenitor Implantation onto Reduced adverse remodeling Gaetani et al.
gelatin cells infarcted murine hearts 2015)
Cardiac patch Methacrylated gelatin Human induced pluripotent stem Implantation onto Improved cardiac function; Gao et al. 2017)
cell-derived cardiomyocytes, smooth infarcted murine hearts reduced scar size
muscle cells and endothelial cells
Cardiac Decellularized cardiac Rat myoblast cells N/A Decellularized cardiac ECM Pati et al. 2014)
ECM
Cardiac patch Decellularized cardiac Human cardiac progenitor cells Implantation onto rat Decellularized cardiac ECM Bejleri et al. 2018)
ECM; gelatin hearts
methacrylate
Cardiac Alginate; Human umbilical vein endothelial Murine subcutaneous Formation of vessel-like Maiullari et al.,
PEG-fibrinogen cells; induced pluripotent stem implantation structures 2018
cell-derived cardiomyocytes
Cardiac Gelatin; alginate; Human umbilical vein endothelial N/A Perfusable hollow vessels Jia et al., 2016
poly(ethylene glycol)- cells; human mesenchymal stem
tetra-acrylate cells
Cardiac Alginate; gelatin Human umbilical vein endothelial N/A Toxicity testing; bioreactor Zhang et al., 2016
methacryloyl cells; neonatal rat cardiomyocytes;
human induced pluripotent stem
cell-derived cardiomyocytes
Cardiac patch Decellularized cardiac Human c-kit+ cardiac progenitor Implantation onto Decellularized cardiac ECM Jang et al., 2017
ECM cells infarcted rat hearts
Cardiac Gelatin methacryloyl; Cardiomyocytes; cardiac fibroblasts N/A Gold nanorods Zhu et al., 2017
gold nanorods (electroconductivity)
Cardiac patch Methacrylated Human coronary artery endothelial N/A Carbon nanotubes (electro- Izadifar et al.,
collagen; carbon cells conductivity); UV-integrated 2017
nanotubes 3D bioprinting technique
Cardiac Polycaprolactone; Rat H9c2 cells N/A Carbon nanotubes Ho et al., 2017
carbon nanotubes (electroconductivity)
Cardiac patch No Human induced pluripotent stem Implantation onto rat Biomaterial-free 3D Ong et al., 2017
cell-derived cardiomyocytes; hearts bioprinting
fibroblasts; endothelial cells
Cardiac patch No Human induced pluripotent stem Implantation onto rat Biomaterial-free 3D Yeung et al., 2019
cell-derived cardiomyocytes; hearts bioprinting
fibroblasts; endothelial cells
Valve Alginate; gelatin Smooth muscle cells; aortic valvular N/A Heterogenous cell Duan et al., 2013
interstitial cells encapsulation
Valve Methacrylated Human aortic valvular interstitial N/A Deposition of collagen and Duan et al., 2014
hyaluronic acid; cells glycosaminoglycans
methacrylated gelatin
Valve Polyethylene glycol- Porcine aortic valvular interstitial N/A Photocrosslinking Hockaday et al.,
diacrylate; alginate cells 2012
Heart Alginate N/A N/A FRESH method (freeform Hinton et al., 2015
reversible embedding of
suspended hydrogels)
Coronary Collagen type I; C2C12 cells; human embryonic Murine subcutaneous FRESH method (freeform Lee et al., 2019
artery; alginate stem cell-derived cardiomyocytes; vascularization model reversible embedding of
ventricle; cardiac fibroblasts; human umbilical suspended hydrogels)
valve; heart vein endothelial cells

ECM, extracellular matrix.


130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 5


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
and geometry data obtained from computed tomography ers, with the main difference being that the printing fila-
and MRI scanning, one can generate patient-specific ment found in traditional FDM printers is replaced with
models that are of high quality and accuracy, which is cell-laden and cell-free biocompatible inks (bioinks). The
valuable considering the current interest and push to- largest driving factor making extrusion-based printers a
ward personalized health care. Once the 3D model has go-to in the bioprinting community is that the printing
been created, it is then processed in slicing software that technique can be easily achieved through the conversion/
generates a 3D map with coordinates and specifications modification of conventional extrusion-based 3D print-
that direct the 3D bioprinter on how and where to print ers, a process that reduces the cost and time spent devel-
the desired construct [Bücking et al., 2017]. oping the printing device [Pusch et al., 2018]. Having said
While there are many different types and principles that, there are a variety of commercially available, modi-
behind the currently available printers for bioprinting, fied and custom-made extrusion-based 3D printers being
there has been much interest in the development of hand- employed throughout the literature.
held devices and robotically-assisted devices aimed at Even though there still remain challenges to overcome
printing tissues, organs, and scaffolds in situ [Singh et al., in order to produce more physiologically relevant 3D
2020]. Although there has been some progress toward printed models via extrusion-based bioprinting, the re-
this goal [Keriquel et al., 2010; Skardal et al., 2012; Di search community has made, and continues to make,
Bella et al., 2018; Hakimi et al., 2018; Albanna et al., 2019; great advancements in the development of better equip-
Adib et al., 2020; Cheng et al., 2020; Urciuolo et al., 2020], ment, techniques, and printing material. The efforts to
it remains challenging due to the unavailability of bioinks reduce printing time, the development of equipment to
that precisely match mechanical and biological properties print with multiple materials to create more complex
needed to print tissues with intricate geometric features structures, and the use of sacrificial materials to create
and complex biomechanical properties, such as cardiac channels and vasculature within the printed constructs
tissue and blood vessels. For these reasons there have still make extrusion-based 3D bioprinting a promising
been no examples of in situ printed cardiac tissue to date. technique to advance the field of tissue engineering.
As such, in the next sections, we will focus on ex situ bio- One of the main limitations encountered with extru-
printing. Many of the current ex situ bioprinting devices sion-based bioprinting techniques is the inability to
and techniques are derived from traditional 3D printing print complex 3D structures due to the need for support
as they have been thoroughly studied and developed. materials and the low rigidity and stiffness of the bio-
During ex situ bioprinting, the scaffold, tissue, or organ inks. During FRESH bioprinting, a liquid hydrogel is
is printed outside of the body under sterile aseptic condi- extruded within a thermoreversible bath, which be-
tions and then transplanted to the target site (with or haves as a Bingham fluid [Hinton et al., 2015]. The sup-
without the inclusion of exogenous cells) or used in vitro port bath acts as a gelatin-like solid at rest and as a liquid
in the study of tissue development, disease, and drug when sufficient shear stress is generated within the flu-
treatments. Considering that the bioprinting of cell-laden id. During 3D printing, the motion of the needle that
and cell-free constructs is most frequently accomplished extrudes the bioink generates shear stress of enough
through the use of extrusion-based printers, we will focus magnitude such that the bath behaves like a liquid dur-
on them. However, there are a number of other tech- ing the motion of the needle, but it acts as a solid sup-
niques which have been developed such as freeform re- port to the extruded bioink. Once the printing process
versible embedding of suspended hydrogels (FRESH) is completed, the temperature of the support bath is
[Hinton et al., 2015; Lee et al., 2019], inkjet [Angelopou- raised such that it becomes a liquid, and the crosslinked
los et al., 2020], laser/light-induced [Guillotin et al., 2013], 3D printed structure can be easily removed. This behav-
and other less explored techniques such as ultrasonic ior of the support bath, coupled with cross-linking reac-
standing bulk acoustic waves (SBAW) [Chansoria and tions between the bath and the bioink, allow for the 3D
Shirwaiker, 2019], aspiration-assisted [Ayan et al., 2020], printing of constructs with more complex structures
3D sacrificial molding [Miller et al., 2012], optogenetic and higher fidelity to the employed CAD model. Even
manipulation [Huang Y et al., 2018], Kenzan method though it is still in its early stages, this technology has
[Aguilar et al., 2019], and pick-and-place [Blakely et al., shown potential to produce geometrically complex
2015] based bioprinting. structures using both cell-free and cell-laden bioinks,
Extrusion-based bioprinters work in a very similar solving some of the limitations of conventional extru-
fashion to fused-deposition modeling (FDM) 3D print- sion-based bioprinting.
130.209.6.61 - 8/12/2021 3:25:54 PM

6 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
Another printing technique that also made use of pre- that are washed away or degraded after the printing pro-
viously developed technologies and adapted them to cess, generating vascularized and/or more geometrically
serve the purpose of tissue engineering and bioprinting is complex structures.
inkjet bioprinting. In inkjet bioprinting, the same con- Yet another 3D printing technique that has gained
cept of droplet printing is applied, but instead of ink, the some interest among research groups is the application of
modified printers eject bioinks. The droplets are ejected laser-induced forward transfer techniques for bioprint-
using thermal [Cui et al., 2012a], microvalve (pressur- ing [Gruene et al., 2011; Kingsley et al., 2013; Koch et al.,
ized) [Ng et al., 2017], or piezoelectric components 2013]. During this process, also referred to as laser-assist-
[Hoehne et al., 2020]. This technique allows for a non- ed bioprinting, a cell-loaded hydrogel is propelled for-
contact dispersal of cell-loaded bioinks with high accu- ward by the pressure generated by a laser-induced vapor
racy and, in some cases, the possibility to determine how bubble. Usually, there is a laser-absorbing layer and a cell-
many cells are encapsulated and dispersed in every drop- loaded hydrogel layer. Upon irradiation with the laser, a
let at each location [Li et al., 2018]. In spite of these ad- droplet of hydrogel is ejected onto another surface. Using
vancements, this bioprinting method has not gained as this technique, 3D printed structures with fine patterning
much attention as other methods due to low cell viability can be generated [Koch et al., 2013].
after the printing process, and the inability to generate
geometrically complex structures, which hinders its ap-
plication in clinically relevant settings [Li et al., 2018]. 3D Printing of Cardiac Tissue and Its Applications
Laser/light-induced bioprinting has also gained sub-
stantial interest among researchers and companies in re- Cardiac Tissue
cent years. Although within the concept of utilizing light Contractility is the most fundamental function of car-
to bioprint 3D structures there are various different de- diac tissue. There are several examples of 3D printed con-
vices and techniques, they all use light and/or lasers to structs displaying this property. For example, Wang et al.
initiate the crosslinking of reactive functionalities, or to [2018] developed a spontaneously contractile cardiac tis-
eject with high accuracy droplets of bioink in specific pat- sue from a fibrin-based bioink and primary rat cardio-
terns. In the former, researchers apply concepts of photo- myocytes, which maturated in vitro and responded phys-
chemistry to induce the photo-crosslinking of bioinks. To iologically to drugs. Tijore et al. [2018] generated a pat-
accomplish this, various teams have applied concepts re- terned 3D bioprinted gelatin hydrogel with microchannels
lated to stereolithography and digital light processing, that promoted differentiation of human mesenchymal
where lasers and reflective surfaces are used to photo- stem cells and beating of cardiomyocytes. While Gaetani
crosslink a liquid into very complex shapes in a layer-by- et al. [2015] developed a patch from hyaluronic acid, gel-
layer manner [Connell et al., 2013; Cui et al., 2019; Grigo- atin and human cardiac-derived progenitor cells, which,
ryan et al., 2019; Hong et al., 2020]. This approach allows upon implantation to the infarcted murine heart, pre-
for the rapid printing of complex and vascularized con- served cardiac function and reduced adverse ventricular
structs reducing the need for support material and avoid- remodeling. Similarly, Gao et al. [2017] showed that a car-
ing the risk of cell death due to shear forces created as the diac patch derived from printed methacrylated gelatin,
bioink would typically pass through the nozzles of an ex- hiPSC-derived cardiomyocytes, smooth muscle cells, and
trusion-based system. However, one of the main con- endothelial cells improved cardiac function and reduced
cerns and limitations of this technique is the toxicity of scar formation in infarcted murine hearts.
some of the photoinitiator molecules themselves, or the The importance of cardiac-specific ECM was recog-
reactive intermediates and byproducts that are generated nized by Pati et al. [2014], who used decellularized por-
during the photo-crosslinking process [Zhu et al., 2015; cine ventricles to develop a cardiac-specific decellularized
Bagheri and Jin, 2019]. Other devices that combine extru- ECM bioink that resulted in printed constructs that al-
sion-based bioprinting and light-induced techniques to lowed for better maturation of rat myoblast cells as com-
create complex structures with different materials have pared to collagen printed counterparts. Another ap-
also been developed [Cui et al., 2012b; Hockaday et al., proach by Bejleri et al. [2018] involved the combination
2012; Shao et al., 2018]. This method allows for the use of of decellularized cardiac ECM, gelatin methacrylate, and
more readily accessible 3D bioprinters, as well as the gen- human CPCs to generate cardiac patches. Incorporation
eration of multi-material constructs, where some of these of cardiac ECM within these patches led to greater matu-
materials are intended as support or sacrificial materials ration of human CPCs as confirmed by increased expres-
130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 7


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
sion of cardiogenic genes, while patches implanted with- ECM bioink, hoping there would be synergistic effects.
in the rat model showed good vascularization. The resulting patches with human c-kit+ cells and human
Given the limited oxygen diffusion [Gershlak et al., mesenchymal stem cells were implanted onto infarcted rat
2017] and exceptionally high demand of cardiac tissue for hearts and were found to promote cardiac function, vas-
oxygen and nutrients, the incorporation of vessel-like cularization, and new muscle formation.
structures into a 3D bioprinted construct is a necessity. Another key feature of cardiac tissue is its ability to
Maiullari et al. [2018] exploited the endogenous ability of propagate electrical signals owing to its intrinsic electro-
HUVECs to form monolayers and tubular structures and conductivity. There are several examples of 3D bioprint-
combined this with the idea of 3D bioprinted constructs ed constructs that have been printed using bioinks con-
consisting of perpendicularly printed fibers. In particular, taining electroconductive components such as metallic
they used 2 cell-laden bioinks, the first one combined algi- nanoparticles, graphene-like materials, and synthetic
nate and poly(ethylene glycol)-fibrinogen (PEG-fibrino- conductive polymers. For example, Zhu et al. [2017] em-
gen) hydrogel with HUVECs, whereas the second com- bedded gold nanorods into a gelatin methacryloyl bioink.
bined induced pluripotent stem cell (iPSC)-derived car- The gold nanorods were found to promote cardiomyo-
diomyocytes with the hydrogel. A variety of 3D printed cyte coupling and synchronized contraction. Carbon
structures consisting of perpendicular fibers were pre- nanotubes have also found use in the design of conductive
pared, each with a different pattern of incorporated cell constructs. In one study, a UV-integrated 3D bioprinting
types. For example, there were Janus constructs, which technique was employed to create nanoreinforced and
consisted of HUVECs and iPSC-derived cardiomyocytes conductive cardiac patches [Izadifar et al., 2017]. Through
within each fiber layer, as well as 2:2:2:2:2 structures, which the combination of human coronary artery endothelial
consisted of alternating double layers of fibers comprising cells with methacrylated collagen and carbon nanotubes,
either HUVECS or iPSC-derived cardiomyocytes, and the authors demonstrated that one can significantly im-
4:2:4 structures where the pattern of cell deposition was prove electrical conductivity in these types of materials.
interrupted by a double layer of unique cells every fifth Similarly, Ho et al. [2017] used carbon nanotubes and
layer. Upon subcutaneous implantation in NOD-SCID combined them with polycaprolactone and showed bio-
mice, the Janus constructs showed the highest formation compatibility with rat H9c2 cells.
of vessel-like structures originating from HUVECs as well A biomaterial-free 3D bioprinting approach has also
as host vascularization, while the incorporated cardiomy- been developed by Ong et al. [2017]. Spheroids derived
ocytes displayed the best alignment. Jia et al. [2016] have from hiPSC-derived cardiomyocytes, fibroblasts, and en-
also developed a method for incorporating hollow vessels dothelial cells were bioprinted onto a needle array and
within 3D printed constructs through the use of a multi- allowed to fuse. The resulting fused cardiac tissue was re-
layer coaxial extrusion system employing gelatin methac- moved from the array and allowed to mature. In vivo im-
ryloyl, sodium alginate, and poly(ethylene glycol)-tetra- plantation of the cardiac patches for 1 week onto the
acrylate derived bioinks which are amenable to both ionic- hearts of rats resulted in their engraftment and vascular-
and photo-crosslinking. This method allowed for the ization. This work was then expanded by Yeung et al.
bioprinting of perfusable hollow tubes as well as the incor- [2019], who investigated the consequences of in vivo im-
poration of HUVECs and human mesenchymal stem cells plantation for 4 weeks. After 4 weeks, cardiac function
into the constructs. A more stepwise approach to this goal trended toward improvement, while scar size was re-
was developed by Zhang et al. [2016]. First, a microfibrous duced, and vascularization was promoted.
alginate/gelatin methacryloyl scaffold with HUVECs was
printed. Next, the HUVECs were allowed to form lumen- Heart Valves
like structures. Finally, neonatal rat cardiomyocytes or Several approaches have been taken toward the fabri-
hiPSC-derived cardiomyocytes were seeded into the inter- cation of 3D printed valves. Duan et al. [2013] prepared
stitial space to form a beating endothelialized cardiac tis- heterogeneously populated aortic valve conduits based
sue. These constructs were intended to be used as in vitro on a alginate/gelatin hydrogel. The root region was popu-
models to reveal drug side effects and potential cardiotox- lated by human aortic root smooth muscle cells, whereas
icity. As a proof-of-concept, constructs were exposed to the leaflets contained porcine aortic valve interstitial cells.
doxorubicin and showed dose-dependent toxicity. Lastly, The authors then went on to further improve their ap-
Jang et al. [2017] looked at improving the vascularization proach for printing tri-leaflet valves by utilizing methac-
of constructs by using a decellularized porcine cardiac rylated hyaluronic acid and gelatin along with human
130.209.6.61 - 8/12/2021 3:25:54 PM

8 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
aortic valvular interstitial cells [Duan et al., 2014]. The disease or as replacements of diseased or damaged tissue,
cells actively remodeled the initial scaffold through col- they need to be matured/trained in an environment which
lagen and glycosaminoglycan deposition. Hockaday et al. recapitulates the in vivo environment they are to mimic
[2012] printed an aortic valve via photocrosslinking of or be employed. Considering the complexity of cardio-
printed polyethylene glycol-diacrylate supplemented vascular tissue, there is a clear need to design universal
with alginate and seeded with porcine aortic valve inter- bioreactors which can accommodate a variety of geome-
stitial cells. While Lee et al. [2019] printed a collagen-de- tries and allow for the culture of cells, perfusion with me-
rived tri-leaflet heart valve at adult human scale using the dia and growth factors, stimulation (mechanical, electri-
FRESH method and showed mechanical functionality in cal, electromechanical), visualization, and real-time
a flow system as well as biocompatibility with HUVECs monitoring of tissue health, maturity, and physical prop-
that eventually formed a monolayer on the surface. erties, all while maintaining a sterile/aseptic environment
[Borovjagin et al., 2017]. While the advantages of 3D bio-
Heart reactors over 2D culture are well-known, and there exist
As previously mentioned, the FRESH method of 3D a number of different designs and approaches developed
printing, developed by Hinton et al. [2015], allows for the over the years, many of the devices developed to date are
3D printing of hydrated materials with elastic modulus quite simple, with the majority of them failing to incor-
<500 kPa using a secondary hydrogel support bath. Using porate features that would allow for accurate mimicry of
this method, the authors successfully printed a chick em- in vivo environments [Murphy et al., 2020]. Also, they
bryonic heart model from alginate with complex trabecu- typically involve multi-step, complex, and destructive
lar architecture. The FRESH method has also been used transfer of bioprinted constructs into and out of the bio-
to bioprint collagen type I under pH-driven control of reactor, and have unintuitive interfaces and limited con-
gelation into various scale model structures [Lee et al., trol over stimulation parameters or regimes [Murphy et
2019]. Starting from small vessels and microporous scaf- al., 2020]. As the availability of both autoclavable and bio-
folds, they proceeded to print a model ventricle seeded compatible filaments and resins has increased, and both
with human stem cell-derived cardiomyocytes and car- FDM and stereolithography printing technologies have
diac fibroblasts, which was capable of contraction. The matured to the point where printers are readily accessible
resulting ventricle was capable of spontaneous beating at by most research groups, there is now the possibility of
a rate of ∼0.5 Hz and could also be paced at 1 and 2 Hz. rapidly generating 3D printed prototypes of complex bio-
Additionally, they designed a human heart model with reactors in-house. Using CAD software and the direct in-
valves, trabeculae, and vessels and demonstrated vessel put from scientists, researchers, and clinicians that are
patency in a segment if the vasculature was down to ∼100 developing, studying, and implanting these constructs, it
μm. is relatively simple to develop improved prototypes with
3D printed models have also found use in helping to features that could possibly enhance the translation and
better understand patient-specific anatomical conditions efficacy of the fabricated tissues. In the design of 3D bio-
and in planning the best surgical approach, especially in reactors for cardiac tissues, there are a number of features
complex or borderline cases, such as patients with con- that the device must allow for control of such as cell-cell
genital heart defects, patients requiring valve replace- interactions, cell-ECM interactions, mechanical stimula-
ment, ventricular assist device implantation or heart tion, electrical stimulation, tissue stiffness, and vessel for-
transplant [Ripley et al., 2016; Valverde et al., 2017; An- mation [Borovjagin et al., 2017]. Considering these re-
war et al., 2018; Farooqi et al., 2019; Forte et al., 2019; quirements, there have been relatively few 3D printed re-
Tuncay and van Ooijen, 2019]. A summary of the afore- actors reported for cardiac tissue, with the majority of
mentioned achievements can be found in Table 1. reactors focused primarily on mechanical stimulation
alone, a property which has been used for the condition-
ing of cardiac tissue but should ideally be accompanied
3D Printing of Devices for Culture and Maturation of with electrochemical cues as these combinations have
Cardiac Tissue been demonstrated to improve maturation of cell and tis-
sue cultures toward more functional phenotypes [Lu et
While there have been significant advances in the bio- al., 2013; Miklas et al., 2014; Morgan and Black, 2014;
printing of tissue constructs, in order for these materials Ruan et al., 2016; Kroll et al., 2017; Besser et al., 2018].
to be used as accurate models for the study of drugs and Table 2 lists some of the 3D bioreactors which have been
130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 9


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
Table 2. 3D printed bioreactors for tissue maturation and testing

Device name Printer/material Tissue(s) Stimulation Reference

FABRICA SLA Liver Perfusion Smith et al., 2018


Dental SG resin Kidney
Bone
BEaTS-α FDM Cardiac Electromechanical Cortes et al., 2020
Nylon 680
– SLA Tendon Mechanical Banik and Brown, 2020
Not described
– FDM Cardiac Mechanical Putame et al., 2019
ABS plus-P430
SR30 soluble
– FDM Tendon Mechanical Raveling et al., 2018
ABS
– FDM Cardiac Mechanical Putame et al., 2019
ABS plus-P430
SpinΩ FDM Neural Mechanical Qian et al., 2016
ULTEM 9085
– PµSL Vascularized Vascularization Xia and Fang, 2009
Poly(ethylene glycol)

ABS, acrylonitrile butadiene styrene; FDM, fused deposition modeling; SLA, stereolithography; SG, surgical
guide; PµSL, projection micro-stereolithography.

fabricated using 3D printing technologies; while this list In addition to in-house fabricated bioreactors, a num-
is not meant to be all-encompassing, it does highlight dif- ber of companies have begun using 3D printing technol-
ferent types of bioreactors using these techniques and ogies to speed up development of new and custom biore-
gives an idea of the kind of stimulation regimes that can actors. Having the ability to share design files between
be incorporated in these devices. Recently, we have re- sites and print locally means that devices do not have to
ported on the development of the BEaTS-α bioreactor, an be shipped between manufacturing and testing sites, sav-
open-source system for the simultaneous electro and me- ing time and money, and allowing for quicker optimiza-
chanical stimulation of cells in vitro (hiPSC-derived car- tion as slight modifications to the design files can be made
diomyocytes) [Cortes et al., 2020]. The BEaTS-α device by those deploying the device or, if necessary, shared dig-
was designed with CAD software, and most components itally with the manufacturer who can quickly make the
were 3D printed in-house via FDM using autoclavable changes and send back the file for printing [Formlabs,
and FDA-approved materials. Combined with a com- 2019].
mercially available C-PACE EP system, our device is ca-
pable of electromechanically stimulating cells cultured on
flexible silicone membranes fitted to a standard 6-well Future Directions
plate. The compact and open-access system is capable of
partially recapitulating the natural contraction and signal Cardiac tissue capable of contraction and spontaneous
propagation found within the heart. Our experiments in- beating and functional valves have been successfully cre-
dicated that hiPSC-derived cardiomyocytes cultured un- ated using 3D printing technology. While this is promis-
der electromechanical stimulation with the device showed ing, in order to proceed to cell-containing large-scale
a more mature cardiomyocyte phenotype than non-stim- constructs or even to whole hearts of human size, solu-
ulated cells after only 7 days of stimulation [Cortes et al., tions to problems facing the vascularization of theses con-
2020]. structs and artificial tissues will need to be developed.
130.209.6.61 - 8/12/2021 3:25:54 PM

10 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
Given the diffusion limit of oxygen [Carmeliet and Jain, most frequently used. Contractile and electroconductive
2000], an intricate network of capillaries supplied by ar- tissue constructs/patches with vessel-like structures have
terioles is needed. However, the current 3D printing tech- been successfully tested both in vitro and in vivo with very
nology is not able to print such microvascular structures promising outcomes. Custom-made bioprinted and cell-
[Gershlak et al., 2017]. Increases in printing resolution containing valves, as well as 3D printed models of the
and advances in the use of sacrificial materials might en- heart hold great promise for treating cardiac disease.
able us to overcome this limitation of printing micro- Maturation of the printed tissues can be further enhanced
sized hollow structures. Concurrently, the use of biocom- by cultivation in custom-made bioreactors that allow for
patible and biomimetic materials and reduced use of cy- perfusion and/or electromechanical stimulation. Novel
totoxic photocrosslinkers and other initiators might advancements in the bioprinting of cardiac tissue are also
significantly increase cell viability. Furthermore, the de- expected with improvements to currently available print-
velopment of even more sophisticated printers may en- ing techniques as well as through the combination of pre-
able simultaneous printing of multiple bioinks, thus lead- viously developed strategies.
ing to more geometrically and physicochemically com-
plex constructs allowing for higher spatiotemporal
specificity of biomimetic cues and heterogenous cell colo- Conflict of Interest Statement
nization of the construct. Miniaturization would also en-
able in situ bioprinting of cardiac tissue, as so far only in The authors have no conflicts of interest to declare.
situ printing for skin or cartilage repair was achieved [Di
Bella et al., 2018; Cheng et al., 2020]. An improvement of
reactors to encompass multiple stimulation modalities Funding Sources
and regimes, automated transfer of bioprinted constructs
This work was made possible by funding from the Natural Sci-
in and out (to achieve the highest standard of aseptic con- ences and Engineering Research Council (NSERC) RG-
ditions), or more user-friendly interfaces will also likely PIN-2015-0632 to E.I.A. and Canadian Institutes of Health Sci-
lead to materials with greater translational success. ences (CIHR) to E.I.A. and E.J.S. E.I.A. also thanks the New Fron-
tiers Research Program Exploration fund and the Government of
Ontario for an Early Career Research Award.
Conclusion

3D bioprinting is a versatile technology enabling the Author Contributions


development of cell-laden and cell-free constructs. Cur- The manuscript was written through contributions from all au-
rently, in the field of cardiovascular tissue engineering, thors. All authors have given approval to the final version of the
extrusion-based printing and the FRESH method are manuscript.

References
Adib AA, Sheikhi A, Shahhosseini M, Simeunović Albanna M, Binder KW, Murphy SV, Kim J, Qa- Atala A. Tissue Engineering and Regenerative
A, Wu S, Castro CE, et al. Direct-write 3D sem SA, Zhao W, et al. In situ bioprinting of Medicine: Concepts for Clinical Application.
printing and characterization of a GelMA- autologous skin cells accelerates wound heal- Rejuvenation Res. 2004;7(1):15–31.
based biomaterial for intracorporeal tissue ing of extensive excisional full-thickness Ayan B, Heo DN, Zhang Z, Dey M, Povilianskas
engineering. Biofabrication. 2020; 12(4): wounds. Sci Rep. 2019;9(1):1856. A, Drapaca C, et al. Aspiration-assisted bio-
045006. Angelopoulos I, Allenby MC, Lim M, Zamorano printing for precise positioning of biologics.
Aguilar IN, Olivos DJ 3rd, Brinker A, Alvarez MB, M. Engineering inkjet bioprinting processes Sci Adv. 2020;6(10):eaaw5111.
Smith LJ, Chu TG, et al. Scaffold-free bio- toward translational therapies. Biotechnol Bagheri A, Jin J. Photopolymerization in 3D
printing of mesenchymal stem cells using the Bioeng. 2020;117(1):272–84. Printing. ACS Appl Polym Mater. 2019; 1(4):
Regenova printer: Spheroid characterization Anwar S, Singh GK, Miller J, Sharma M, Manning 593–611.
and osteogenic differentiation. Bioprinting. P, Billadello JJ, et al. 3D Printing is a Trans- Banerjee M N, Bolli R, Hare J M. Clinical Studies
2019;15:e00050. formative Technology in Congenital Heart of Cell Therapy in Cardiovascular Medicine:
Ahmed RE, Anzai T, Chanthra N, Uosaki H. A Disease. JACC Basic Transl Sci. 2018; 3(2): Recent Developments and Future Directions.
Brief Review of Current Maturation Methods 294–312. Circ Res. 2018;123(2):266–87.
for Human Induced Pluripotent Stem Cells- Banik BL, Brown JL. 3D-Printed Bioreactor En-
Derived Cardiomyocytes. Front Cell Dev hances Potential for Tendon Tissue Engineer-
Biol. 2020;8(178):178. ing. Regen Eng Transl Med. 2020;6:419–28.
130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 11


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
Bejleri D, Streeter BW, Nachlas ALY, Brown ME, Cui X, Breitenkamp K, Finn MG, Lotz M, D'Lima Grigoryan B, Paulsen SJ, Corbett DC, Sazer DW,
Gaetani R, Christman KL, et al. A Bioprinted DD. Direct Human Cartilage Repair Using Fortin CL, Zaita AJ, et al. Multivascular net-
Cardiac Patch Composed of Cardiac-Specific Three-Dimensional Bioprinting Technology. works and functional intravascular topologies
Extracellular Matrix and Progenitor Cells for Tissue Eng Part A. 2012b;18(11-12):1304–12. within biocompatible hydrogels. Science.
Heart Repair. Adv Healthcare Mater. 2018; Dangas GD, Weitz JI, Giustino G, Makkar R, 2019;364(6439):458.
7(23):1800672. Mehran R. Prosthetic Heart Valve Thrombo- Gruene M, Pflaum M, Deiwick A, Koch L, Schlie
Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, sis. J Am Coll Cardiol. 2016;68(24):2670–89. S, Unger C, et al. Adipogenic differentiation
Barnabé-Heider F, Walsh S, et al. Evidence for Deo KA, Singh KA, Peak CW, Alge DL, Gaharwar of laser-printed 3D tissue grafts consisting of
Cardiomyocyte Renewal in Humans. Science. AK. Bioprinting 101: Design, Fabrication, human adipose-derived stem cells. Biofabri-
2009;324(5923):98. and Evaluation of Cell-Laden 3D Bioprinted cation. 2011;3(1):015005.
Besser RR, Ishahak M, Mayo V, Carbonero D, Scaffolds. Tissue Eng Part A. 2020; 26(5-6): Guillotin B, Catros S, Guillemot F. Laser Assisted
Claure I, Agarwal A. Engineered Microenvi- 318–38. Bio-printing (LAB) of Cells and Bio-materials
ronments for Maturation of Stem Cell De- Dey M, Ozbolat IT. 3D bioprinting of cells, tissues Based on Laser Induced Forward Transfer
rived Cardiac Myocytes. Theranostics. 2018; and organs. Sci Rep. 2020;10(1):14023. (LIFT). In: Schmidt V, Belegratis MR, editors.
8(1):124–40. Di Bella C, Duchi S, O'Connell CD, Blanchard R, Laser Technology in Biomimetics: Basics and
Birla RK, Williams SK. 3D bioprinting and its po- Augustine C, Yue Z, et al. In situ handheld Applications. Berlin, Heidelberg: Springer;
tential impact on cardiac failure treatment: three-dimensional bioprinting for cartilage 2013. p. 193–209.
An industry perspective. APL Bioeng. 2020; regeneration. J Tissue Eng Regen Med. 2018; Guo Y, Pu WT. Cardiomyocyte Maturation: New
4(1):010903. 12(3):611–21. Phase in Development. Circ Res. 2020;126(8):
Blakely AM, Manning KL, Tripathi A, Morgan JR. Duan B, Hockaday LA, Kang KH, Butcher JT. 3D 1086–106.
Bio-Pick, Place, and Perfuse: A New Instru- Bioprinting of heterogeneous aortic valve Hakimi N, Cheng R, Leng L, Sotoudehfar M, Ba
ment for Three-Dimensional Tissue Engi- conduits with alginate/gelatin hydrogels. J PQ, Bakhtyar N, et al. Handheld skin printer:
neering. Tissue Eng Part C Methods. 2015; Biomed Mater Res A. 2013;101(5):1255–64. in situ formation of planar biomaterials and
21(7):737–46. Duan B, Kapetanovic E, Hockaday LA, Butcher tissues. Lab Chip. 2018;18(10):1440–51.
Boehler RM, Graham JG, Shea LD. Tissue engi- JT. Three-dimensional printed trileaflet valve He L, Chen X. Cardiomyocyte Induction and Re-
neering tools for modulation of the immune conduits using biological hydrogels and hu- generation for Myocardial Infarction Treat-
response. Biotechniques. 2011; 51(4): 239–40, man valve interstitial cells. Acta Biomater. ment: Cell Sources and Administration Strat-
242, 244 passim. 2014;10(5):1836–46. egies. Adv Healthc Mater. 2020; 9(22):
Borovjagin AV, Ogle BM, Berry JL, Zhang J. From Farooqi KM, Cooper C, Chelliah A, Saeed O, Chai 2001175.
Microscale Devices to 3D Printing: Advances PJ, Jambawalikar SR, et al. 3D Printing and Hinton TJ, Jallerat Q, Palchesko RN, Park JH,
in Fabrication of 3D Cardiovascular Tissues. Heart Failure: The Present and the Future. Grodzicki MS, Shue HJ, et al. Three-dimen-
Circ Res. 2017;120(1):150–65. JACC Heart Fail. 2019;7(2):132–42. sional printing of complex biological struc-
Bücking TM, Hill ER, Robertson JL, Maneas E, Feric NT, Radisic M. Maturing human pluripo- tures by freeform reversible embedding of
Plumb AA, Nikitichev DI. From medical im- tent stem cell-derived cardiomyocytes in hu- suspended hydrogels. Sci Adv. 2015; 1(9):
aging data to 3D printed anatomical models. man engineered cardiac tissues. Adv Drug e1500758.
PLoS One. 2017;12(5):e0178540. Deliv Rev. 2016;96:110–34. Ho CM, Mishra A, Lin PT, Ng SH, Yeong WY,
Carmeliet P, Jain RK. Angiogenesis in cancer and Formlabs . How 3D Printing is Accelerating Tis- Kim YJ, et al. 3D Printed Polycaprolactone
other diseases. Nature. 2000; 407(6801): 249– sue Engineering Research at the University of Carbon Nanotube Composite Scaffolds for
57. Sheffield. 2019. https: //3d.formlabs.com/ Cardiac Tissue Engineering. Macromol Bios-
Chansoria P, Shirwaiker R. Characterizing the white-paper-how-3d-printing-is-accelerat- ci. 2017;17(4):1600250.
Process Physics of Ultrasound-Assisted Bio- ing-tissue-engineering-research/ Hockaday LA, Kang KH, Colangelo NW, Cheung
printing. Sci Rep. 2019;9(1):13889. Forte MNV, Hussain T, Roest A, Gomez G, Jong- PY, Duan B, Malone E, et al. Rapid 3D print-
Cheng RY, Eylert G, Gariepy JM, He S, Ahmad H, bloed M, Simpson J, et al. Living the heart in ing of anatomically accurate and mechanical-
Gao Y, et al. Handheld instrument for wound- three dimensions: applications of 3D printing ly heterogeneous aortic valve hydrogel scaf-
conformal delivery of skin precursor sheets in CHD. Cardiol Young. 2019;29(6):733–43. folds. Biofabrication. 2012;4(3):035005.
improves healing in full-thickness burns. Bio- Gaetani R, Feyen DA, Verhage V, Slaats R, Mes- Hoehne JL, Carlstron R, Dernorwsek J, Cristovam
fabrication. 2020;12(2):025002. sina E, Christman KL, et al. Epicardial appli- PC, Bachiega HL, Abensur SI, et al. Piezoelec-
Connell JL, Ritschdorff ET, Whiteley M, Shear JB. cation of cardiac progenitor cells in a 3D- tric 3D bioprinting for ophthalmological ap-
3D printing of microscopic bacterial commu- printed gelatin/hyaluronic acid patch pre- plications: process development and viability
nities. Proc Natl Acad Sci USA. 2013;110(46): serves cardiac function after myocardial analysis of the technology. Biomed Phys Eng
18380. infarction. Biomaterials. 2015;61:339–48. Express. 2020;6(3):035021.
Cortes D, McTiernan CD, Ruel M, Franco W, Gao L, Kupfer ME, Jung JP, Yang L, Zhang P, Da Hong H, Seo YB, Kim DY, Lee JS, Lee YJ, Lee H,
Chu C, Liang W, et al. BEaTS-α an open ac- Sie Y, et al. Myocardial Tissue Engineering et al. Digital light processing 3D printed silk
cess 3D printed device for in vitro electrome- With Cells Derived From Human-Induced fibroin hydrogel for cartilage tissue engineer-
chanical stimulation of human induced plu- Pluripotent Stem Cells and a Native-Like, ing. Biomaterials. 2020;232:119679.
ripotent stem cells. Sci Rep. 2020;10(1):11274. High-Resolution, 3-Dimensionally Printed Horn MA, Trafford AW. Aging and the cardiac
Cui H, Zhu W, Huang Y, Liu C, Yu ZX, Nowicki Scaffold. Circ Res. 2017;120(8):1318–25. collagen matrix: Novel mediators of fibrotic
M, et al. In vitro and in vivo evaluation of 3D Gershlak JR, Hernandez S, Fontana G, Perreault remodelling. J Mol Cell Cardiol. 2016;93:175–
bioprinted small-diameter vasculature with LR, Hansen KJ, Larson SA, et al. Crossing 85.
smooth muscle and endothelium. Biofabrica- kingdoms: Using decellularized plants as per- Hosoyama K, Ahumada M, McTiernan CD, Da-
tion. 2019;12(1):015004. fusable tissue engineering scaffolds. Biomate- vis DR, Variola F, Ruel M, et al. Nanoengi-
Cui X, Boland T, D'Lima DD, Lotz MK. Thermal rials. 2017;125:13–22. neered Electroconductive Collagen-Based
inkjet printing in tissue engineering and re- Gopalan N, Nor SNM, Mohamed MS. Global Hu- Cardiac Patch for Infarcted Myocardium Re-
generative medicine. Recent Pat Drug Deliv man Embryonic Stem Cell Laws and Policies pair. ACS Appl Mater Interfaces. 2018;10(51):
Formul. 2012a;6(2):149–55. and Their Influence on Stem Cell Tourism. 44668–77.
Biotechnol Law Rep. 2018;37(5):255–69.
130.209.6.61 - 8/12/2021 3:25:54 PM

12 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:
Hospodiuk M, Dey M, Sosnoski D, Ozbolat IT. Lee JK, Link JM, Hu JCY, Athanasiou KA. The Pusch K, Hinton TJ, Feinberg AW. Large volume
The bioink: A comprehensive review on bio- Self-Assembling Process and Applications in syringe pump extruder for desktop 3D print-
printable materials. Biotechnol Adv. 2017; Tissue Engineering. Cold Spring Harb Per- ers. HardwareX. 2018;3:49–61.
35(2):217–39. spect Med. 2017;7(11):a025668. Putame G, Terzini M, Carbonaro D, Pisani G,
Huang NF, Serpooshan V, Morris VB, Sayed N, Li X, Chen J, Liu B, Wang X, Ren D, Xu T. Inkjet Serino G, Di Meglio F, et al. Application of 3D
Pardon G, Abilez OJ, et al. Big bottlenecks in Printing for Biofabrication. In: Ovsianikov A, Printing Technology for Design and Manu-
cardiovascular tissue engineering. Commun Yoo J, Mironov V, editors. 3D Printing and facturing of Customized Components for a
Biol. 2018;1(1):199. Biofabrication. Cham: Springer International Mechanical Stretching Bioreactor. J Healthc
Huang Y, Xia A, Yang G, Jin F. Bioprinting Living Publishing; 2018. p. 283–301. Eng. 2019;2019:3957931.
Biofilms through Optogenetic Manipulation. Lu L, Mende M, Yang X, Körber HF, Schnittler Qian X, Nguyen HN, Song MM, Hadiono C, Og-
ACS Synth Biol. 2018;7(5):1195–200. HJ, Weinert S, et al. Design and validation of den SC, Hammack C, et al. Brain-Region-
Izadifar M, Chapman D, Babyn P, Chen X, Kelly a bioreactor for simulating the cardiac niche: Specific Organoids Using Mini-bioreactors
ME. UV-assisted 3D bioprinting of nano-re- a system incorporating cyclic stretch, electri- for Modeling ZIKV Exposure. Cell. 2016;
inforced hybrid cardiac patch for myocardial cal stimulation, and constant perfusion. Tis- 165(5):1238–54.
tissue engineering. Tissue Eng Part C Meth- sue Eng Part A. 2013;19(3-4):403–14. Raveling AR, Theodossiou SK, Schiele NR. A 3D
ods. 2017;24(2):74–88. Maiullari F, Costantini M, Milan M, Pace V, printed mechanical bioreactor for investigat-
Jang J, Park HJ, Kim SW, Kim H, Park JY, Na SJ, Chirivì M, Maiullari S, et al. A multi-cellular ing mechanobiology and soft tissue mechan-
et al. 3D printed complex tissue construct us- 3D bioprinting approach for vascularized ics. MethodsX. 2018;5:924–32.
ing stem cell-laden decellularized extracellu- heart tissue engineering based on HUVECs Ripley B, Kelil T, Cheezum MK, Goncalves A, Di
lar matrix bioinks for cardiac repair. Bioma- and iPSC-derived cardiomyocytes. Sci Rep. Carli MF, Rybicki FJ, et al. 3D printing based
terials. 2017;112:264–74. 2018;8(1):13532. on cardiac CT assists anatomic visualization
Jia W, Gungor-Ozkerim PS, Zhang YS, Yue K, Metra M, Teerlink JR. Heart failure. Lancet. 2017; prior to transcatheter aortic valve replace-
Zhu K, Liu W, et al. Direct 3D bioprinting of 390(10106):1981–95. ment. J Cardiovasc Comput Tomogr. 2016;
perfusable vascular constructs using a blend Miklas JW, Nunes SS, Sofla A, Reis LA, Pahnke A, 10(1):28–36.
bioink. Biomaterials. 2016;106:58–68. Xiao Y, et al. Bioreactor for modulation of Ronaldson-Bouchard K, Ma SP, Yeager K, Chen
Jourdan-LeSaux C, Zhang J, Lindsey ML. Extra- cardiac microtissue phenotype by combined T, Song L, Sirabella D, et al. Advanced matu-
cellular matrix roles during cardiac repair. static stretch and electrical stimulation. Bio- ration of human cardiac tissue grown from
Life Sci. 2010;87(13-14):391–400. fabrication. 2014;6(2):024113. pluripotent stem cells. Nature. 2018;
Jover E, Fagnano M, Angelini G, Madeddu P. Cell Miller JS, Stevens KR, Yang MT, Baker BM, 556(7700):239–43.
Sources for Tissue Engineering Strategies to Nguyen DH, Cohen DM, et al. Rapid casting Ross MH, Pawlina W. Histology: A Text and At-
Treat Calcific Valve Disease. Front Cardio- of patterned vascular networks for perfusable las: With Correlated Cell and Molecular Biol-
vasc Med. 2018;5:155. engineered three-dimensional tissues. Nat ogy. 6th ed. Philadelphia: Lippincott Williams
Kengla C, Kidiyoor A, Murphy SV. Chapter 68. Mater. 2012;11(9):768–74. & Wilkins; 2006.
Bioprinting Complex 3D Tissue and Organs. Morgan KY, Black LD 3rd. Mimicking isovolu- Ruan JL, Tulloch NL, Razumova MV, Saiget M,
In: Orlando G, Remuzzi G, Williams DF, edi- mic contraction with combined electrome- Muskheli V, Pabon L, et al. Mechanical Stress
tors. Kidney Transplantation, Bioengineering chanical stimulation improves the develop- Conditioning and Electrical Stimulation Pro-
and Regeneration. London: Academic Press; ment of engineered cardiac constructs. Tissue mote Contractility and Force Maturation of
2017. p. 957–71. Eng Part A. 2014;20(11-12):1654–67. Induced Pluripotent Stem Cell-Derived Hu-
Keriquel V, Guillemot F, Arnault I, Guillotin B, Müller P, Lemcke H, David R. Stem Cell Therapy man Cardiac Tissue. Circulation. 2016;
Miraux S, Amédée J, et al. In vivo bioprinting in Heart Diseases - Cell Types, Mechanisms 134(20):1557–67.
for computer- and robotic-assisted medical and Improvement Strategies. Cell Physiol Sedlakova V, Ruel M, Suuronen EJ. Therapeutic
intervention: preliminary study in mice. Bio- Biochem. 2018;48(6):2607–55. Use of Bioengineered Materials for Myocar-
fabrication. 2010;2(1):014101. Murphy SV, De Coppi P, Atala A. Opportunities dial Infarction. In: Alarcon EI, Ahumada M,
Kingsley DM, Dias AD, Chrisey DB, Corr DT. and challenges of translational 3D bioprint- editors. Nanoengineering Materials for Bio-
Single-step laser-based fabrication and pat- ing. Nat Biomed Eng. 2020;4(4):370–80. medical Uses. Cham: Springer International
terning of cell-encapsulated alginate micro- Ng WL, Lee JM, Yeong WY, Win Naing M. Mi- Publishing; 2019. p. 161–93.
beads. Biofabrication. 2013;5(4):045006. crovalve-based bioprinting - process, bio-inks Shamhart PE, Meszaros JG. Non-fibrillar colla-
Koch L, Gruene M, Unger C, Chichkov B. Laser and applications. Biomater Sci. 2017; 5(4): gens: Key mediators of post-infarction cardi-
assisted cell printing. Curr Pharm Biotechnol. 632–47. ac remodeling?. J Mol Cell Cardiol. 2010;
2013;14(1):91–7. Nguyen AH, Marsh P, Schmiess-Heine L, Burke 48(3):530–7.
Kroll K, Chabria M, Wang K, Häusermann F, PJ, Lee A, Lee J, et al. Cardiac tissue engineer- Shao L, Gao Q, Zhao H, Xie C, Fu J, Liu Z, et al.
Schuler F, Polonchuk L. Electro-mechanical ing: state-of-the-art methods and outlook. J Fiber-Based Mini Tissue with Morphology-
conditioning of human iPSC-derived cardio- Biol Eng. 2019;13(1):57. Controllable GelMA Microfibers. Small.
myocytes for translational research. Prog Bio- Ong CS, Fukunishi T, Zhang H, Huang CY, 2018;14(44):e1802187.
phys Mol Biol. 2017;130:212–22. Nashed A, Blazeski A, et al. Biomaterial-Free Shtein Z, Shoseyov O. When bottom-up meets
Laflamme MA, Sebastian MM, Buetow BS. 10. Three-Dimensional Bioprinting of Cardiac top-down. Proc Natl Acad Sci USA. 2017;
Cardiovascular. In: Treuting PM, Dintzis SM, Tissue using Human Induced Pluripotent 114(3):428–9.
editors. Comparative Anatomy and Histolo- Stem Cell Derived Cardiomyocytes. Sci Rep. Singh S, Choudhury D, Yu F, Mironov V, Naing
gy. San Diego: Academic Press; 2012. p. 135– 2017;7(1):4566. MW. In situ bioprinting - Bioprinting from
53. Pati F, Jang J, Ha DH, Won Kim S, Rhie JW, Shim benchside to bedside? Acta Biomater. 2020;
Lee A, Hudson AR, Shiwarski DJ, Tashman JW, JH, et al. Printing three-dimensional tissue 101:14–25.
Hinton TJ, Yerneni S, et al. 3D bioprinting of analogues with decellularized extracellular Skardal A, Mack D, Kapetanovic E, Atala A, Jack-
collagen to rebuild components of the human matrix bioink. Nat Commun. 2014;5(1):3935. son JD, Yoo J, et al. Bioprinted amniotic fluid-
heart. Science. 2019;365(6452):482–7. Prabhu SD, Frangogiannis NG. The Biological derived stem cells accelerate healing of large
Basis for Cardiac Repair After Myocardial In- skin wounds. Stem Cells Transl Med. 2012;
farction: From Inflammation to Fibrosis. Circ 1(11):792–802.
Res. 2016;119(1):91–112.
130.209.6.61 - 8/12/2021 3:25:54 PM

3D Printing of Cardiac Tissues and Cells Tissues Organs 13


Devices for Their Maturation DOI: 10.1159/000512792
Glasgow Univ.Lib.
Downloaded by:
Smith LJ, Li P, Holland MR, Ekser B. FABRICA: Urciuolo A, Poli I, Brandolino L, Raffa P, Scatto- Xia C, Fang NX. 3D microfabricated bioreactor
A Bioreactor Platform for Printing, Perfusing, lini V, Laterza C, et al. Intravital three-dimen- with capillaries. Biomed Microdevices. 2009;
Observing, & Stimulating 3D Tissues. Sci Rep. sional bioprinting. Nat Biomed Eng. 2020; 11(6):1309–15.
2018;8(1):7561. 4(9):901–915. Yeung E, Fukunishi T, Bai Y, Bedja D, Pitaktong
Squelch A. 3D printing and medical imaging. J Valverde I, Gomez-Ciriza G, Hussain T, Suarez- I, Mattson G, et al. Cardiac regeneration using
Med Radiat Sci. 2018;65(3):171–2. Mejias C, Velasco-Forte MN, Byrne N, et al. human-induced pluripotent stem cell-de-
Steenbergen C, Frangogiannis NG. Chapter 36. Three-dimensional printed models for surgi- rived biomaterial-free 3D-bioprinted cardiac
Ischemic Heart Disease. In: Hill JA, Olson cal planning of complex congenital heart de- patch in vivo. J Tissue Eng Regen Med. 2019;
EN, editors. Muscle. Boston/Waltham: Aca- fects: an international multicentre study. Eur 13(11):2031–9.
demic Press; 2012. p. 495–521. J Cardiothorac Surg. 2017;52(6):1139–48. Zhang YS, Arneri A, Bersini S, Shin SR, Zhu K,
Tandon N, Cannizzaro C, Chao PH, Maidhof R, Waller DG, Sampson AP. 5. Ischaemic heart dis- Goli-Malekabadi Z, et al. Bioprinting 3D mi-
Marsano A, Au HT, et al. Electrical stimula- ease. In: Waller DG, Sampson AP, editors. crofibrous scaffolds for engineering endothe-
tion systems for cardiac tissue engineering. Medical Pharmacology and Therapeutics. 5th lialized myocardium and heart-on-a-chip.
Nat Protoc. 2009;4(2):155–73. ed. Elsevier; 2018. p. 93–110. Biomaterials. 2016;110:45–59.
Taylor DA, Elgalad A, Sampaio LC. What will it Wang Z, Lee SJ, Cheng HJ, Yoo JJ, Atala A. 3D Zhu F, Friedrich T, Nugegoda D, Kaslin J, Wlod-
take before a bioengineered heart will be im- bioprinted functional and contractile cardiac kowic D. Assessment of the biocompatibility
planted in patients?. Curr Opin Organ Trans- tissue constructs. Acta Biomater. 2018;70:48– of three-dimensional-printed polymers using
plant. 2018;23(6):664. 56. multispecies toxicity tests. Biomicrofluidics.
Tijore A, Irvine SA, Sarig U, Mhaisalkar P, Bai- Watkins DA, Beaton AZ, Carapetis JR, Karthikey- 2015;9(6):061103.
sane V, Venkatraman S. Contact guidance for an G, Mayosi BM, Wyber R, et al. Rheumatic Zhu K, Shin SR, van Kempen T, Li YC, Ponraj V,
cardiac tissue engineering using 3D bioprint- Heart Disease Worldwide: JACC Scientific Nasajpour A, et al. Gold Nanocomposite Bio-
ed gelatin patterned hydrogel. Biofabrication. Expert Panel. J Am Coll Cardiol. 2018;72(12): ink for Printing 3D Cardiac Constructs. Adv
2018;10(2):025003. 1397–416. Funct Mater. 2017;27(12):1605352.
Tuncay V, van Ooijen PMA. 3D printing for heart World Health Organization. Global Health Esti-
valve disease: a systematic review. Eur Radiol mates 2016: Deaths by Cause, Age, Sex, by
Exp. 2019;3(1):9. Country and by Region, 2000–2016. Geneva:
World Health Organization; 2018.

130.209.6.61 - 8/12/2021 3:25:54 PM

14 Cells Tissues Organs Sedlakova/McTiernan/Cortes/Suuronen/


DOI: 10.1159/000512792 Alarcon
Glasgow Univ.Lib.
Downloaded by:

You might also like