You are on page 1of 32

Home Search Collections Journals About Contact us My IOPscience

3D bioprinting of skin: a state-of-the-art review on modelling, materials, and processes

This content has been downloaded from IOPscience. Please scroll down to see the full text.

2016 Biofabrication 8 032001

(http://iopscience.iop.org/1758-5090/8/3/032001)

View the table of contents for this issue, or go to the journal homepage for more

Download details:

IP Address: 207.162.240.147
This content was downloaded on 10/09/2016 at 12:11

Please note that terms and conditions apply.

You may also be interested in:

Organ Printing: Natural, synthetic and semi-synthetic polymers


D-W Cho, J-S Lee, J Jang, J W Jung, J H Park and F Pati

Towards artificial tissue models: past, present, and future of 3D bioprinting


Ahu Arslan-Yildiz, Rami El Assal, Pu Chen et al.

Electrospun nanostructured chitosan--poly(vinyl alcohol) scaffolds: a biomimetic extracellular


matrix as dermal substitute
Dhakshinamoorthy Sundaramurthi, Kirthanashri Srinivasan Vasanthan, Purushothaman Kuppan et al.

Fabrication of a nanofibrous scaffold with improved bioactivity for culture of human dermal
fibroblasts for skin regeneration
Arun Richard Chandrasekaran, J Venugopal, S Sundarrajan et al.

Layer-by-layer assembly for biomedical applications in the last decade


P Gentile, I Carmagnola, T Nardo et al.

Viscoelastic, physical, and bio-degradable properties of dermal scaffolds and related cell
behaviour
Vaibhav Sharma, Nimesha Patel, Nupur Kohli et al.
Biofabrication 8 (2016) 032001 doi:10.1088/1758-5090/8/3/032001

TOPICAL REVIEW

3D bioprinting of skin: a state-of-the-art review on modelling,


RECEIVED
18 March 2016
materials, and processes
REVISED
21 July 2016
ACCEPTED FOR PUBLICATION
S Vijayavenkataraman1, W F Lu1 and J Y H Fuh1,2
8 August 2016 1
Department of Mechanical Engineering, National University of Singapore (NUS), Singapore
2
PUBLISHED NUS Research Institute, Suzhou Industry Park, People’s Republic of China
7 September 2016
E-mail: vijayavenkataraman@u.nus.edu

Keywords: 3D printing, skin, additive manufacturing, bioprinting, tissue engineering, skin printing

Abstract
The skin is the largest organ of the body, having a complex multi-layered structure and guards the
underlying muscles, bones, ligaments, and internal organs. It serves as the first line of defence to any
external stimuli, hence it is the most vulnerable to injury and warrants the need for rapid and reliable
regeneration methods. Tissue engineered skin substitutes help overcome the limitations of traditional
skin treatment methods, in terms of technology, time, and cost. While there is commendable progress
in the treating of superficial wounds and injuries with skin substitutes, treatment of full-thickness
injuries, especially with third or fourth degree burns, still looks murkier. Engineering multi-layer skin
architecture, conforming to the native skin structure is a tougher goal to achieve with the current
tissue engineering methods, if not impossible, restoring all the functions of the native skin. The testing
of drugs and cosmetics is another area, where engineered skins are very much needed, with bans being
imposed on product testing on animals. Given this greater need, 3D bioprinting is a promising
technology that can achieve rapid and reliable production of biomimetic cellular skin substitutes,
satisfying both clinical and industrial needs. This paper reviews all aspects related to the 3D bioprinting
of skin, right from imaging the injury site, 3D model creation, biomaterials that are used and their
suitability, types of cells and their functions, actual bioprinting technologies, along with the challenges
and future prospects.

1. Skin: anatomy and functions these will cause undesirable skin conditions like rash,
dermatitis, cellulitis, and even melanoma.
The skin, also called as integument is the largest organ Skin, mostly seen just as a fleshly covering, does a lot
of the body, accounting for about 15% of the total body more vital functions than we could possibly imagine,
weight in adults. Skin, along with its derivative structure including (i) Protective (barrier, UV light absorption,
constitutes the integumentary system [1]. The skin has a immune surveillance, mechanical), (ii) Perceptive
very complex multi-layered structure, which in turn (touch, temperature, pain), and (iii) Regulatory (ther-
consists of several other components such as cells, mal, hydration, excretory) functions. With all the above
fibres, extra-cellular matrix (ECM), veins, capillaries, functions, skin thus aids in the maintenance of
nerves, and the hair follicles. The skin predominantly homeostasis [4].
consists of three layers namely epidermis, dermis, and Each and every component of the skin performs
hypodermis (also called as subcutaneous tissue). The specialised functions, in addition to the general func-
epidermis is the outer most layer, consisting of Kerati- tions stated above. There are many reported works on
nocytes (KCs), the middle layer is the dermis, consisting the functions and mechanism of functions of kerati-
of collagen and fibroblasts (FCs) and the inner most nocytes [5–9], fibroblasts [10–13], melanocytes
layer is hypodermis, consisting of lipocytes and collagen [14–19], merkel cells [20–24], Langerhans cells
(figure 1). The constituents of three layers [2], in detail, [25–30], mast cells [31–37], macrophages [28, 38–41],
are summarized in table 1. Malfunction of any one of and other components of skin as well.

© 2016 IOP Publishing Ltd


Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 1. Anatomy of skin, showing the three layers (epidermis, dermis, hypodermis), the skin appendages (hair shaft, sweat gland)
and the cellular constituents (keratinocytes and, melanocytes in epidermis; fibroblasts in dermis; adipose tissue at the base of
hypodermis; collagen in all three layers). Reprinted by copyright permissions from [3].

Table 1. Constituents and functions of the layers of skin.

Layer Constituents Function

Epidermis Keratinocytes Guards from environmental damage by pathogens, heat, UV radiation and water loss
Melanocytes Responsible for the production of the pigment melanin, protection from UV-B light exposure
Merkel Cells Associated with tactility (sense of touch)
Langerhans Cells Antigen-presenting immune cells
Dermis Collagen Fibrous family of proteins responsible for stress-resistance and elasticity
Fibroblasts Synthesizes ECM and Collagen, plays a critical role in wound healing
Mast cells Allergy response, anaphylaxis, wound healing and angiogenesis
Hypodermis Fibroblasts Synthesizes ECM and Collagen, plays a critical role in wound healing
Lipocytes/Adipocytes Energy storage in the form of fat
Macrophages Phagocytosis, adaptive immunity, wound healing

2. Compelling needs for artificial bio- medical treatment per year are 486 000 and the num-
mimic skin ber of hospitalizations per year for acute burn injuries
are 40 000, per the 2016 report [44]. Fire, flames, elec-
Artificial skins are biological or synthetic substitutes tricity, scald and chemical products were the major
for human skin, produced in laboratories. There are causes of such acute burn injuries. The severity of the
two compelling reasons as to why the world is in need burn injury depends on the duration and intensity of
of artificial skins: (i) wound healing and skin regenera- exposure and can be classified accordingly [45, 46].
tion—especially for burn victims, and (ii) drug and Of the classification shown in figure 2, third degree
skin care products (cosmetics) testing. and fourth degree burns are considered as full-thick-
Injury or illness that causes loss of skin integrity ness injuries and are the most difficult to treat. This
will lead to substantial physiological imbalance and requires the regeneration of the whole skin structure,
ultimately to significant disability, sometimes even multi-layers with each having specialized cells, appen-
fatality. The 2016 global wound management marke t dages, and other components. Not only is the thick-
is expected to hit $15 billion and forecasted to be ness or volume of skin to be regenerated that matters,
worth over $22 billion in 2024 [42]. The global tissue but restoring the functionality of the original skin is a
engineered skin substitutes market was valued at USD necessity. Understanding the wound healing mechan-
958.8 million in 2014 and is projected to reach USD isms [46–50] will help achieve the objective of making
3873.5 million by 2023, expanding at a Compound functional skins by emerging advanced technologies.
Annual Growth Rate (CAGR) of 17.2% from 2015 to The second important reason that accelerates the
2023 [43]. American Burn Association recorded that, need for artificial skins is the drug and skin-care pro-
in the US, the number of burn injuries receiving ducts (cosmetics) testing. Not all drugs are mild

2
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 2. Classification of wounds based on four criteria namely open wounds, depth of the wound, burns, and closed wounds. Open
wounds are those where the skin is broken and the underlying tissues are exposed to the external environment; depth refers to the
depth of skin injury, where superficial wounds involves loss of epidermis only, partial thickness wounds involves loss of some part of
dermis also along with epidermis and dermal wounds involves loss of both epidermis and dermis; burns, based on the extent of injury
and severity can be classified into first up to fourth degree burns; closed wounds are those where the skin is intact and the underlying
tissues are not exposed to the environment.

enough to be tested directly on the humans. Research- —114th Congress (2015–2016))’) [59]; and (iv) Com-
ers in pharmaceutical and skin care industries were panies were classified as: Brands that DO NOT test on
and are using animals to test their products. However, animals, Brands that DO test on Animals, Brands
testing the products on animals is not always pre- whose animal testing status is unknown. And people
dictive of responses in humans. In other words, the are encouraged not to buy the products of those com-
response to the same drug or product may be slightly panies that do product tests on animals [60, 61]. These
or even drastically different from the response that is are the most important reasons behind the need for
observed in animals, making the whole testing process artificial skins, not just an alternate, but bio-mimetic,
unreliable, meaningless and absurd. Also, these testing fully-functional human skin, which is a big challenge
procedures may cause discomfort and pain to the ani- but not impossible.
mals [51–53]. Many organisations and associations
were formed since long, like Alternatives to Animal
3. Current skin substitutes
Testing (SCAAT) Steering Committee formed by The
European Cosmetics Association (COLIPA) in 1992, Skin substitutes are defined as a heterogeneous group
the workshop on use of non-invasive methods in cos- of substances that help in either temporary or perma-
metics testing organised by European Centre for the nent closure of many types of wounds, depending on
Validation of Alternative Methods (ECVAM) in 1994 wound coverage that vary based on wound and
[53–57], pressing the need to regulate the policies and product characteristics [62]. Skin substitutes help
procedures of industrial animal testing and if possible, when standard surgical therapies like flap coverage or
to ban them and find other alternatives. The voice is surgical debridement is not desirable, which is decided
louder now as ever before, after almost a decade. Due on a case to case basis, depending on the type and
to increasing concerns on the animal welfare and con- extend of burn, shallow or deep wounds, the degree of
stant pressure by newer and stronger organisations burn, and the area of the body that is injured. There is
like PETA (People for the Ethical Treatment of Ani- no perfect or ideal substitute for the original skin per se
mals), urgency is created for alternative viable technol- but something is better than nothing always. Not to
ogies. In summary, (i) People are concerned about mention, researchers around the world, in interdisci-
using products which are tested on animals and many plinary teams, comprising of scientists in mechanical,
avoid such products; (ii) Many countries (including chemical, biomedical engineering, biological, and
EU, Israel and India) had already banned industries medical sciences are progressing towards the greater
from ‘testing their products’ on animals [58]; (iii) goal of bio-mimicking the original skin, a fully
Other countries also are considering deploying the ban functional skin substitute, with all its intricacies in it.
(like the USA ‘The Humane Cosmetics Act (H.R.2858 We are not there yet, but good news is we are moving

3
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 2. Characteristics of an ideal skin substitute. reasons as to why we did not succeed, such as the
technological limitations to have multi-layered bio-
Able to resist infection
Able to prevent water loss or dehydration mimicking structure, with each layer made of different
Able to withstand the shear forces and wound hypoxia cells and appendages. However, the most important
Cost-efficient and affordable challenges are vascularization and innervation. Boyce
Easy to prepare, store and use et al [70] give an insightful discussion on the limita-
Wide availability and Long Shelf life tions of the skin grafts based on the wound healing
Lack of antigenicity
physiology [71]. Lack of organogenesis is stated as one
Flexible in thickness
Durable with long-term wound stability
big short-coming of the current models. Other dis-
Can be conformed to irregular wound surfaces advantages include mechanical fragility, susceptibility
Provides permanent wound coverage to microbial contamination, lesser rates of engraft-
Recreates dermal and epidermal components ment, time delay in healing, and of course the cost.
Most of the skin substitutes are predominantly
acellular and even if they are cellular, they only use two
types of cells, namely fibroblasts and keratinocytes,
closer. There are a few important characteristics that a
most of the cases, only either of them. This prevents
perfect skin substitute is expected to have [62, 63] and
the substitute from being fully functional like the
they are summarized in table 2.
native skin [66]. Another limitation was pigmentation
Quantification of each of the properties listed in
[66], which can also be solved if the pigment secreting
table 2 is not straight forward and there is no catalogue
cells are made available in the substitutes. Vasculariza-
readily available to compare which is acceptable,
tion and innervation is a challenge, especially for drug
which is not and what is best. It is up to the clinicians
and cosmetics testing, where fully functional skin is a
to decide how these tenets affect the product selection
requirement and not a choice. In wound healing appli-
and availability, depending on the type of cases they
cations, fully functional skin is not needed because the
are dealing with and the practice environment. There
healthy skin can provide nourishment to the injured
are many commercially available products, each hav-
area where artificial skin is grafted.
ing its own advantages and disadvantages, strengths
and limitations, giving choices for the practitioners to
choose. Comparison of some of these products is dealt 4. Major approaches in tissue engineering
with, in the later part of this section. of skin

3.1. Classification of skin substitutes Having said that about the limitations of the current
Skin substitutes can be classified based on their skin substitutes, it is important to understand the
durability, single or multi-layer, the type of layer, and approaches in using advanced skin regeneration
composite substitutes. Skin substitutes may be broadly strategies such as 3D bioprinting. There are two
classified as temporary or permanent and biological or fundamental strategies for tissue engineering in gen-
synthetic, which is more practical. In response to the eral [72–77], namely (i) bottom-up approach and (ii)
classification proposed by Balasubramani et al [64], top-down approach.
which is based on the plasticity and composition, The bottom-up approaches aim to fabricate a lar-
Kumar et al [65] had added upon to give a comprehen- ger complex tissue by biological sintering of smaller
sive framework for classification of skin substitutes, as building blocks, at micrometric scale, mimicking the
given in table 3. natural tissue architecture. The building blocks may be
There are other ways of classification too. Dorothy cell aggregates, cell-laden hydrogel, polymer microbe-
et al [66] classifies the skin substitutes in to three clas- ads with a homogenous or heterogeneous composi-
ses namely Acellular, Cellular-Allogenic, and Cellular- tion. These smaller blocks are spatially arranged in 3D
Autologous; Auger et al [67] reports three classes to form a biomimetic complex architecture. Cell sheet
namely Epidermal substitutes, Dermal substitutes, technology, which aims at building up functional thick
and Bi-layer substitutes. Some of the current skin sub- tissues without the use of biodegradable scaffolds and
stitutes available commercially are tabulated in table 4. bioreactor by layering individual cell sheets of few
Many papers have reported the detailed information, microns thick and bottom-up assembly of cell-laden
application, clinical trial results of the listed skin sub- microgels are most common examples [78, 79]. Some
stitutes [63, 66–69]. of the other technologies used in this approach are
laser-assisted techniques like direct writing [80, 81],
3.2. Limitations of current skin substitutes laser-induced forward transfer [82, 83], inkjet printers
Skin substitutes help in the restoration and regenera- or Drop-On-Demand systems [84, 85], and micro-
tion of skin to a greater extent but they are not without dispensing techniques [86].
limitations. Mimicking the original skin in all aspects The top-down approaches aims at processing lar-
including functionality has not been done successfully ger bulk materials into smaller complex structures.
to date, to the best of our knowledge. There are many Top-down approaches generally involves the use of

4
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 3. Classification of Skin Substitutes.

Class Sub-classes Examples

Class I: Temporary impervious dres- (i) Single layer Materials - Biological dressing substitute (potato peel, amoniotic
sing materials membrane)
(ii) Bi-layered Tissue Engineered - Synthetic dressing substitute (synthetic polymer sheet
Materials (Tegaderm®, Opsite®), polymer foam or spray
TransCyte®
Class II: Single layer durable skin (i) Epidermal Substitutes cultured epithelial autograft (CEA), Apligraft®
substitutes
(ii) Dermal Substitutes bovine collagen sheet, e.g. Kollagen®,
porcine collagen sheet, bovine dermal matrix, e.g. Matri-
derm®, human dermal matrix, e.g. Alloderm®
Class III: Composite skin substitutes (i) Skin graft Allograft, Xenograft
(ii) Tissue Engineered Skin Dermal regeneration template, e.g. Integra®, Biobrane®

biodegradable scaffolds. The cells are seeded on to the understanding the cell-material interaction becomes
scaffolds; growth factors are added to aid cell growth critical in both the approaches. Figure 3 dia-
and proliferation, usually preserved in a bioreactor grammatically explains both the approaches.
that provide appropriate mechanical and hydro- In order to successfully fabricate a bio-mimic,
dynamic stimuli for maturation in to a 3D tissue con- fully-functional skin substitute, it is important to have
struct. Gel casting [87, 88], sol-gel process, replication the advantages of both the approaches; thus, over-
of polymer sponge [89, 90], solvent casting and parti- coming the limitations in using either of them sepa-
culate leaching [91], phase separation technique [92], rately. Integration of both the bottom-up and top-
freeze drying, electro-spinning [93–95], selective laser down approaches [110] is an excellent idea, yet to be
sintering (SLS) and stereolithography apparatus (SLA) experimented for skin tissue engineering. In bone tis-
[96–104] are some of the processes used in the fabrica- sue engineering, Ouyang et al [111] used an integrated
tion of biodegradable scaffolds. approach for engineering ligament analogues. They
Both of the approaches have their own pros and used bottom-up approach to fabricate a bone marrow
cons. Bottom-up approach is said to have the potential stromal cells (bMSCs) sheet in the presence of ascorbic
to improvise the vascularization in 3D constructs, by acid and top-down approach to fabricate a scaffold
perfusing them with intraorgan vascular trees [3] and made of poly (L-lactide) (PLLA) and assembled them
the feasibility of depositing multiple-cell types with together using wrapping technique and it is reported
desired spatial orientation in 3D. However, one major to be a promising method for fabricating tissue-like
drawback in this approach is the inability of certain and functional ligament analogues. Sargeant et al [112]
cell types to secrete sufficient amounts of the extra-cel- also reported an integrated approach for hybrid bone
lular matrix (ECM), migration of cells and inability to implants, where self-assembly of peptide amphiphile
form cell-cell junctions [75]. Top-down approach has (PA) nanofibers (bottom-up approach) takes place
many advantages such as the structural stability, ease within the pores of metallic Ti–6Al–4 V foams (top-
of cell migration on the scaffold structure thus form- down approach). Though the integrated approach
ing cell-cell junctions and better cell-cell interactions, possesses many advantages, technical advancements
and availability of a range of biodegradable, bio- are necessary to make the approach feasible for tissue
compatible materials for fabrication of scaffolding engineering [110]. With 3D bioprinting, this integra-
structure. The main disadvantage is the lack of under- tion is possible. As explained earlier, both bottom-up
standing on the cell-scaffold interaction in detail. The (inkjet printers/Drop-On-Demand systems) and top-
mechanical and surface properties of the scaffold will down approaches (SLS, SLA) are possible with 3D bio-
affect the cell viability, growth and proliferation of dif- printing technologies and hence, the integration is also
ferent types of cells. Also, if stem cells are seeded on to not impossible. A hybrid system, consisting of various
the scaffolds, it may differentiate in to different types tenets of 3D bioprinting can achieve this integration.
of cells based on the scaffold properties [105–109]. Especially for 3D bioprinting of skin, bottom-up
Though bottom-up approach is credited with the approach may be used for constructing each layer of
advantage of being scaffold-free, some of the approa- the skin, the epidermis or dermis and then these layers
can be put together using top-down approaches.
ches like extrusion bioprinting involves fabrication of
3D structures with cell laden hydrogels. For reasons
like increasing the cell density, cells can be further 5. 3D Bioprinting of skin
added to the bioprinted constructs (printed from cell-
laden hydrogels by extrusion bioprinting), where the 3D Bioprinting of skin is a popular research area now
properties of the bioprinted construct influences the and big players in the market are investing to further
behaviour of cells that are being added. Hence, research in this area. L’Oreal USA, the largest

5
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 4. Engineered Skin substitutes (Data taken from [66–68]).

Product Company Description Application

Alloderm LifeCell Corporation, Branch- Acellular (freeze-dried) allogeneic dermis Treat full- and partial-thick-
burg, NJ ness wounds
Apligraf Organogenesis Inc., Canton, MA Allogeneic keratinocytes seeded over dermal Treat venous and diabetic foot
scaffold (bovine collagen sponge) contain- ulcers
ing allogeneic fibroblasts
Biobrane UDL Laboratories Inc., Rock- Porcine collagen chemically bound to sili- Temporary covering of partial-
ford, IL cone/nylon membrane thickness burns and wounds
CellSpray Avita Medical. Perth, Australia Preconfluent autologous keratinocytes deliv- Treat partial-thickness burns
ered into a suspension for spray and chronic ulcers
Cymetra LifeCell Corporation, Branch- Micronized particulate acellular cadaveric Wound filler in plastic surgery
burg, NJ dermal matrix
Dermagraft Advanced Biohealing Inc., La Cryopreserved (viable cells) allogeneic fibro- Treat full-thickness diabetic
Jolla, CA blast-derived dermal matrix foot ulcers
Epicel Genzyme Tissue Repair Cor- Confluent autologous keratinocytes from Treat full- and partial-thick-
poration, Cambridge, MA skin on petrolatum gauze backing ness burns and chronic
ulcers
Epidex PharmaTec Confluent autologous keratinocytes from Treat full- and partial-thick-
hair follicles outer root sheath on silicone ness burns and chronic
membrane ulcers
EZ-Derm Molnlycke Health Care, Sweden Aldehyde-crosslinked porcine dermal Treat full- and partial-thick-
collagen ness wounds
FortaFlex Organogenesis Inc., Canton, MA Acellular porcine small intestine submucosa Treat full- and partial-thick-
ness burns, venous and dia-
betic ulcers
Hyalograft 3D Fidia Advanced Biopolymers, Esterified hyaluronic acid matrix seeded with Treat full- and partial-thick-
Italy autologous fibroblasts ness wounds
Hyalomatrix Medline Industries Inc., Munde- bi-layered, sterile and flexible layer made of a Treat pressure ulcers, diabetic
lien, IL derivative of hyaluronic acid (HA) in foot ulcers and deep second-
fibrous form with an outer layer com- degree burns
prised of a semipermeable silicone
membrane
ICX-RHY Intercytex, Ltd, Manchester, UK suspension of human dermal fibroblasts treat a variety of skin related
(HDFs) in cell storage medium problems including Epi-
dermolysis Bullosa and scar
contractures
Integra Integra Life Science Corpora- Acellular. Temporary silicone epidermal Treat partial- or full-thickness
tion, Plainsboro, NJ substitute over dermal scaffold made of burns
collagen and chondroitin-6 sulfate
LaserSkin Fidia Advanced Biopolymers, Subconfluent autologous keratinocytes see- Treat full- and partial-thick-
(Vivoderm) Italy ded on esterified laser-perfor- ness burns and chronic
atedhyaluronic acid matrix ulcers
MatriDerm MedSkin Solutions Dr Suwelack Acellular scaffold made with bovine collagen Treat partial- or full-thickness
AG, Germany types I, III, V and elastin burns
Myskin CellTran Ltd, Sheffield, UK Subconfluent autologous keratinocytes see- Treat partial-thickness burns
ded on specially treated silicone sheet and chronic ulcers
OASIS Smith&nephew plc, London, UK Acellular porcine small intestine submucosa Treat full- and partial-thick-
ness burns, venous and dia-
betic ulcers
OrCel Ortec International, Inc., New Allogeneic keratinocytes seeded over dermal Treat skin graft donor sites and
York, NY scaffold (bovine collagen sponge) contain- mitten-hand surgery for epi-
ing allogeneic fibroblasts dermolysis bullosa
Permacol Tissue ScienceLaboratories, Processed dermal xenograft Temporary burn coverage &
Andover, MA clean partial thickness
wounds
Permaderm Regenicin Inc., Little Falls, NJ Autologous keratinocytes seeded onto der- Treat chronic wounds
mal substitute made with autologous
fibroblasts in bovine collagen matrix
Repliform Acellular human dermal allograft Urological plastic surgery
applications
Suprathel Institute of Textile and Process Synthetic Epidermal substitute made of DL- Partial-thickness burns and
Engg, Denkendorf, Germany Lactatide monolayer skin graft donor sites
TissueTech Fidia Advanced Biopolymers, Combination of Hyalograft 3D and Laserskin Treat partial- or full-thickness
Italy burns and chronic ulcers

6
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 4. (Continued.)
Product Company Description Application

VCT-01 Organogenesis Inc., Canton, MA Keratinocytes seeded on top of dermal sub- Treat partial thickness wounds
stitute made with fibroblasts secreting
their own ECM

subsidiary of the world’s leading cosmetics company, the patients to know their needs and wishes on the
recently (May 2015) signed a Research Collaboration facial modification. Though creation of 3D facial
Agreement with a 3D Bioprinting company, Organovo shapes is possible with CT-scans [119, 120], the radia-
Holdings, Inc. to 3D bioprint skin models using the tion effect and the costs are impractical for aesthetic
NoveGen Bioprinting platform of the latter, for testing procedures. Other hardware dependent techniques
their cosmetic products [113]. Procter & Gamble like laser or stereophotogrammetric scanners [121–
(P&G), another big player in consumer goods, 123] are too expensive and much complex to handle.
launched a research project in Singapore (in May To overcome the complexity of hardware dependent
2015), seeking research collaborations from academia methods, many hardware independent software tech-
on 3D bioprinting applications, especially for work- niques were proposed such as shape from shading
able skin models [114]. Rokit, a market leader in 3D [124], structure from motion [125, 126], shape from
printing technology in Korea, announced recently silhouette [127, 128], and statistical facial models
(July 2015) that it is participating in a government [129], but all have limitations [130]. Oliveira-Santos
funded project for the development of bioprinting et al [130] proposed a hardware independent web-
technology and the first focus will be on bioprinting of based application, which used 2D-digital pictures to
human skin tissue [115]. Therefore, there is a huge create 3D representation of a patient’s face for plan-
demand for this technology and potential for immedi- ning the aesthetic procedures. The three important
ate commercialization, if successfully developed. steps in this application are (i) 2D feature and contour
Broadly speaking, there are three important steps points detection (from the 2D-digital image); (ii) 2D to
in 3D Bioprinting process, namely, Pre-processing 3D face reconstruction; and (iii) Texture mapping.
(3D Model Generation, Bio-ink preparation, etc), 3D The greatest advantage of this application is that it is
bioprinting, and Post-processing (using a bioreactor completely internet-based and no skilled manpower is
for tissue maturation). In general, the process flow required to use, unlike the CT or MRI. The same
given by Murphy et al [116] holds good for most of the method can be used for scanning the injured skin, to
bioprinting processes (as given in figure 4). analyse the requirements for skin graft. Though the 3D
reconstruction is not as precise as other advanced
5.1. Imaging, 3D modelling and design approach methods, its simplicity and ease of use gives this tech-
The first and most important step in 3D Bioprinting is nique an edge over others and can be used in places
the process of imaging. This involves scanning of the where affordability is the main issue. Hani et al [131]
injured tissue or part of the body. Three scanning or used various algorithms for useful wound data acqui-
medical imaging technologies, x-ray, Computed sition from laser scanned images. They used two
Tomography, commonly called as CT scan, and methods for solid construction and volume computa-
Magnetic Resonance Imaging or MRI are widely tion namely mid-point projection and convex hull
available and used in hospitals. Though the images approximation, also called as Delaunay tetrahedraliza-
from these scanning methods can be then converted to tion. The main aim of this procedure is to determine
a 3D model using available software, in order to apply critical wound data namely wound top area, true sur-
it for tissue or organ bioprinting, specialized techni- face area, depth, and volume of the ulcer wounds in
ques are required. For instance, a clinician requires to patients, in order to continuously monitor the healing
replace part of injured skin in the visible areas of a burn progress throughout the treatment period. AutoCAD
victim, may be on the face or hands. If the imaging software is used to reconstruct the model, subject to
system does not have the capability to scan and the two methods earlier, from the input laser scan
differentiate the colour of the skin, then exact or at images; these reconstructed 3D models were then used
least a closer representation of the victim’s skin colour to 3D print wax models of ulcer wounds; wound para-
cannot be replicated in the printed skin and hence will meters were measured and compared against the sur-
not be aesthetically appealing. There are some technol- face scan measurements. This method can also be used
ogies that are being developed to solve this problem. to other wound causes like the burn injury, to assess
Most clinicians, especially plastic surgeons depend the wound parameters, thus aiding the clinicians to get
on either free-hand 2D sketches on picture printouts a customized skin graft for each injury site. Active
or computerized picture morphing for defining the dynamic thermography (ADT) or non-destructive
goals of facial aesthetic procedures [117, 118]. These thermography (NDT) is a non-contact imaging tech-
sketches and morphs are then used for discussion with nique used widely in quality inspection of material

7
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 3. Top-down versus Bottom-up approach in Tissue Engineering. (a) Top-down approach involves culturing of cells in a porous
scaffold, cells grow, proliferate, migrate on the scaffold structure and creates ECM while the scaffolds undergo biodegradation and
finally a matured tissue is obtained. (b) Bottom-up approach use cell aggregates, cell sheets or cell-laden hydrogels to produce modular
blocks or tissue building blocks, which are then assembled together to form engineered tissues.

Figure 4. A typical process for bioprinting 3D tissues involving six steps namely Imaging (imaging the injured site/tissue), design
approach (image converted to 3D models; approach can be either printing the biomimetic structure along with cells, self-assembly of
cells or constructing mini-tissues to be assembled later), material selection (based on the intended application, can be natural or
synthetic polymers, ECM or combinations of them), cell selection (based on the intended application, can be stem cells or
differentiated cells), bioprinting (using commercial or custom-made bioprinters) and application (post-processing in a bioreactor for
maturation, grafting or implantation at the injury site). Reprinted with permission from [116].

defects in many industries including aerospace and parameters including the healing time within three
automobile [132]. Recently, this technology has weeks are also assessed previously using this technique
gained much attention in medical applications, for [136, 138]. Pirindeze et al [139] examined the optical
characterizing cutaneous lesions [133, 134] and burns resolution of ADT, to define the resolution limits of
[135–138]. Non-homogeneity of burn wounds, which this system and to establish a set of parameters for
refers to demarcation of different zones based on the thermal simulation using this system. In fact, they
severity of the injury, scaling from first to fourth used human skin of varying thicknesses for this resolu-
degree can be done using ADT [135]. Many tion study and have successfully demonstrated the

8
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 5. Summary of imaging and 3D modelling systems.

Imaging/3D modelling methods Pros Cons Reference

CT-scan Accurate measurement of Radiation exposure, High cost [120, 121]


tissue depths
Stereophotogrammetric scanners Fast capture speeds Complex to handle Very expensive [121–123]
Hardware independent methods (Shape from Sophisticated hardware not Used in research environments and [124–129]
shading, Structure from motion, Shape from required, relatively not optimized for clinical use, lim-
silhouette, Statistical facial models) inexpensive itations on speed and accuracy
Web-based application Completely web-based, no Less reproduction accuracy, sub-opti- [130]
hardware required mal 3D reconstruction
Active dynamic thermography (ADT) Accurate measurement of Necessity of using proper thermal [135–139]
tissue/burn depths, High models of living tissues, controlled
resolution experimental conditions required

ability of ADT’s resolution to differentiate details of other colour reproduction techniques to recognize the
human skin as thin as 0.025″. ADT can be another skin colour of the patient and also replicate the same
potential technology for 3D imaging of burns or colour in 3D bioprinted skin grafts or prostheses.
wounds in the pre-processing stage of 3D bioprinting. Wang et al [144] developed a gradient-based multi-
The imaging and 3D modelling systems are sum- spectral method for face liveliness detection, which
marized in table 5. explored the reflectance of all the distinctive regions in
Colour recognition is another important aspect, a face and their spatial relationship, unlike the avail-
aiding to replicate the exact skin colour of the patient able methods, a complement to the previous pixel-
in engineered skin grafts. Xiao et al [140] developed a based methods. Application of this technology in 3D
colour reproduction system for processing of soft tis- imaging, especially for medical applications like skin
sue prostheses accurately and automatically using wound imaging, is yet to be explored.
advanced technologies like 3D Bioprinting. A Minolta The design approach should be based on the sever-
CM-2600d spectrophotometer was used for colour ity of the injury. If the wound or burn is a partial thick-
measurement, along with SpectraMagic NX Color ness injury, then epidermal layer skin graft should be
Data Software to decipher values in CIELAB values fine. When the severity of the injury is of higher degree,
[141]. The study used Zcorp Z510 3D colour printer. A multi-layered structure is required. If the wound area to
protocol was developed using many mathematical be treated is smaller, scaffold-free methods may be
models based on 240 training colours. Of all the math- acceptable as against the larger wound area, where scaf-
ematical models, the third-order polynomial regres- fold based approach is necessary to facilitate easy cell
sion based on least-square fitting provided the best migration, forming cell clusters and cell-cell junctions
model performance for colour profile. Based on the and finally tissue formation.
colour profile, the colour reproduction system was
established and parameters such as accuracy of colour 5.2. Materials
reproduction, performance of colour repeatability and Material selection is one of the key steps in 3D
colour gamut were evaluated using 14 known human Bioprinting. Biomaterials are a group of materials
skin shades. The printed prostheses demonstrated which are biocompatible and biodegradable. Since the
successful colour reproduction of all these 14 skin most common type of materials used is polymer, they
shades. Simulation of colour gamut for the proposed are also called as biopolymers. Based on their source of
bioprinting system was done and except the extreme production, they can be broadly divided in to two,
skin shades of dark and light, majority of skin colours namely natural polymers, and synthetic polymers.
are reproduced in the 3D bioprinted prostheses. The Natural biopolymers are those that are naturally
colour gamut is shown in figure 5. occurring, including (i) proteins (such as collagen,
The 3D colour image reproduction system devel- gelatin, albumin, thrombin, fibrinogen, etc), and (ii)
oped by Xiao et al [142] is represented in figure 6. polysaccharides (such as chitosan, chitin, cellulose,
Jang et al [143] proposed a spectrum-based colour etc) [145]. Biocompatibility, biodegradability, hydro-
reproduction algorithm for make-up simulation of 3D philicity, biological characteristics, resemblance to the
facial avatar. All the colour processing is based on RGB native ECM are strong reasons why natural polymers
data as of now and its inability to represent a real are preferred in tissue engineering applications.
make-up simulation on a 3D facial avatar encouraged Synthetic biopolymers are man-made polymers
them to work on a spectrum-based colour reproduc- which includes polylactic acid (PLA), polyglycolic acid
tion process, which takes into account the intrinsic (PGA), poly(ε-caprolactone) (PCL), poly(lactic-co-gly-
characteristics of the objects and also the illuminant. colic acid) (PLGA), poly(propylene fumarate), poly-
The performance evaluation of the system was found anhydrides, polycarbonates, polyorthoesters,
acceptable. This method can be coupled along with polyurethanes, and polyphosphazenes. Synthetic

9
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 5. Colour gamut of 3D printing system for 14 human skin colours: (a) top view. and (b) side view. Reprinted with permissions
from [140].

Figure 6. 3D colour image reproduction system for facial prostheses involving six steps namely 3D Image acquisition (facial photos
taken in different angles), 3D image design (processing of the acquired image and converting them to 3D models), colour management
(chosen based on the human skin shade), colour texture mapping (texture matched to the original facial skin), 3D colour printing and
post processing. Reprinted with permission from [142].

biopolymers are known for their excellent mechanical composite polymers, which has both natural and
properties, which are important for a scaffold material. synthetic biopolymer constituents, thus combining
Key advantage is the ability to tailor the mechanical the advantages of both and overcoming the limita-
properties and degradation kinetics, by altering the tions. This is extremely important with 3D bioprinting
polymer structure, to suit various biomedical applica- because, in addition to the basic requirements of a bio-
tions, for example, bone graft or skin graft [146]. polymer, there is another important factor to be con-
Both of the above types suffer from certain limita- sidered while preparing the bio-ink, which is
tions. Natural biopolymers, in spite of having the ‘printability’. The requirements of an optimal bioma-
greatest advantage of mimicking the native tissue terial [145] are biocompatibility with tissues, biode-
ECM, suffer from poor mechanical properties. On the gradability at the ideal rate corresponding to the rate of
other hand, synthetic biopolymers have the advantage new tissue formation, nontoxicity, non-
of better mechanical properties but very different from immunogenicity, optimal mechanical properties, ade-
the natural ECM. And hence, researchers use quate porosity and morphology for transporting of

10
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

cells, gases, metabolites, nutrients, and signal mole- available, which can be selected based on the type of
cules both within the biomaterial and between the bio- printing process used and bio-ink formulation can be
material and the local environment, and printability. done with or without cells, based on the tissue engi-
The most common natural biopolymers used in neering approach that is aimed at. With the develop-
skin tissue engineering are collagen, gelatin, chitosan, ment of smart hydrogels, 3D bioprinting of functional
and silk fibroin; the synthetic biopolymers being PCL, skin will no more be just a dream, but a realistic goal
PLA, PLGA, and PLLA. The composite biopolymers that is achievable in the near future.
are formed by combining one from each group. Elec-
trospinning is one of the most successful technologies 5.3. Cell selection
to produce nanofibers of the above biopolymers, One of the most important factors that decide the
which serve as the scaffold material for skin tissue success of tissue engineering is the choice of suitable
regeneration. A list of such electro-spun nanofibrous
cells, assuring persistence and function of the regener-
scaffolds is tabulated in table 6 (data taken from [147]).
ated tissue for the patient’s life time [198]. The most
This information is included in this review as most of
common type of cells used extensively in current skin
the biopolymers or composites that are electro-spun
substitutes is keratinocytes, which forms the outer
can also be 3D bioprinted using inkjet and Drop-on-
epidermal layer. There are many advantages in using
Demand systems. They are grouped into four main
keratinocytes in skin tissue engineering; they can be
types, viz. natural, synthetic, natural composite (two
cultured, maintained and propagated in the laboratory
or more natural biopolymers), and composites (at
with ease [199, 200] and their resistance to senescence
least one natural and one synthetic biopolymer).
is so great that they can be passaged for many hundreds
As mentioned earlier, in addition to the satisfac-
of generations [201]. However, there are overwhelm-
tion of basic requirements of a biomaterial, the bio-ink
ing limitations that prevent the sole use of keratino-
should be printable. Ideal properties of bioprinting
cytes to engineer a fully functional skin substitute. The
hydrogels are tabulated in table 7 [197]. Malda et al
first major drawback is the time required to culture
[194] summarizes the properties of bio-ink or hydro-
these cells. It takes about 3 weeks for a 2 sq.cm biopsy
gels, under two headings: (i) rheology, which includes
to expand on a surface, sufficient enough to cover the
viscosity, shear thinning, and yield stress and (ii) cross-
whole body (73 sq.m), which may take even longer for
linking mechanisms of hydrogels namely physical,
ionic, chemical, stereocomplex or thermal cross- elderly patients. Scarring is another problem, referred
linking. Nakamura et al [195] discussed the roles of to as hypertrophic scarring. The presence and degree
hydrogels for biofabrication, especially with inkjet and of scarring is directly proportional to the healing time,
3D bioprinting technologies. Murphy et al [196] eval- which is obviously longer when keratinocytes are used
uated the properties of twelve different hydrogels, [202]. Fibroblasts come next to keratinocytes, present
which are commercially available and commonly used in the dermal layer of natural skin and obviously used
in research. These evaluated properties are relevant to in the dermal skin substitutes. They represent a diverse
bioprinting of skin such as gelation time, swelling or population of cells, with the ability to differentiate
contraction, stability, biocompatibility, and print- along a lineage from highly proliferative progenitor
ability. The twelve hydrogels that are evaluated are fibroblasts. They are ubiquitous, static in nature and
Collagen Type I, Collagen/Fibrin, Fibrin, ExtracelTM provide support and maintenance to the tissues. The
Hydrogel, ExtracelTM UV, Tyramine substituted hya- inherent problem of long-term fragility of keratino-
luronic acid (TS-NaHy)-CorgelTM, Methylcellulose- cytes can be overcome by having a layer of dermal
Hyaluronan (MC-HA), Chitosan, Chitosan/Collagen, fibroblasts underneath it. It is reported that there are
Alginate, Alginate/Gelatin, and Polyethylene Glycol specialized subsets of fibroblasts and each respond to
Diacrylate (PEGDA). The results revealed that Extra- the different phases of wound healing and skin
cel-UV, the photocrosslinkable HA-based hydrogel regeneration. And also, the fact that fibroblasts from
was the best candidate for bioprinting, possessing the different sites of the body possesses different func-
desirable properties for bioprinting application. The tional properties, should be considered before selec-
regulatory, commercial and financial aspects of each tion for tissue engineering, as it may affect their
of the hydrogels were also discussed briefly. Wang et al suitability for dermal substitutes [203]. The bilayer or
[197] recently reported a state-of-the-art review of composite skin substitutes, normally referred to as
smart hydrogels for 3D bioprinting; ‘smart’ referring full-thickness skin grafts, consist of both keratinocytes
to the responsiveness of the hydrogel to various exter- and fibroblasts, representing the epidermal and der-
nal stimuli such as pH, temperature, light, electric, and mal layers respectively. The production process of
magnetic field. The potential of combining smart such a composite is complex, involving many phases
hydrogels with 3D bioprinting, their challenges and [204, 205] and there is no contention on the optimal
future prospects were also reviewed. product on long-term [206, 207]. Though the epider-
Material selection is a key for any bioprinting pro- mal keratinocyte and dermal fibroblast composite is
cess. For 3D bioprinting of skin, there is wide range of better than being individually used, there are so many
natural, synthetic, and composite biopolymers other cells and appendages that are required to be

11
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 6. Electro-spun nanofibrous scaffolds for skin tissue engineering (data from [147]).

Polymer Type Special feature Reference

Collagen Natural Reduced wound contraction compared to [148]


freeze-dried scaffolds
Higher cell proliferation in aligned fibres compared to [149]
random scaffold
Gelatin Natural Well-stratified dermal and epidermal layers including [150]
a continuous basal keratinocyte layer
Cross-linking gelatin nanofibers with genipin [151]
improves cell proliferation but cell viability is
reduced
Promotes cell adhesion and spreading of type I [152, 153]
collagen, suitable for wound dressing [152, 153]
Silk fibroin Natural Oxygen plasma-treated SF nanofibers showed higher [154, 155]
cellular activities than the untreated ones
Chitosan Natural Sonication of chitosan nanofibers resulted in higher [156]
porosity and increased cell proliferation

Fibrinogen Natural Suitable for wound repair or hemostatic products [157]


Good mechanical properties, suitable for wound [158]
dressing
Significantly increased peak stress and modulus [159]
values for the fibrinogen cross-linked with
1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
hydrochloride (EDC) and fibrinogen-genipin
cross-linked scaffolds
Collagen/Silk Natural Better cellular responses in hybrid nanofibrous [160]
fibroin composite scaffolds than that of blend nanofibrous scaffolds
Chitosan/Silk Natural Addition of silk fibroin enhanced the mechanical [161]
fibroin composite properties of nanofibers
Chitosan/ Natural Showed keratinocyte migration and wound [162]
Collagen composite re-epithelization-a prerequisite for healing and
regeneration
Chitosan/Chitin Natural Addition of chitin nanocrystals as well as crosslinking [163]
nanocrystals composite had a positive impact on the mechanical properties

PCL Synthetic Enhanced cell proliferation and wound healing due to [164]
the surface degradation of the polymer under
physiological conditions and the formation of
functional groups like hydroxyl and carboxyl
groups that promoted cell proliferation
A micro-machined human skin pattern mould was [165]
used as a collector in an electrospinning setup to
replicate the pattern onto the surface of the electro-
spun mat; exhibits guidance of cells along the skin
pattern without significant deterioration of pattern
geometry
PLLA Synthetic Nanofiber scaffolds enhanced epidermal skin cell [166]
migration across the wound when compared to a
control group without scaffold. Aligned nanofibers
promoted the infiltration of endothelial cells into
the scaffolds

PDLLA-LL Synthetic Hybrid microfibers possessed a unique porous high [167]


surface area mimicking native extracellular
matrix (ECM)
PLGA Synthetic Cells acquired a well spread morphology and collagen [168]
type III gene expression was significantly up-
regulated
Good cellular penetration, with no adverse inflamma- [169]
tory response outside the scaffold margin and with
no capsule formation around the periphery
The cell attachment and spreading were several times [170]
higher in the nano-/microfiber composite

12
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 6. (Continued.)
Polymer Type Special feature Reference

scaffolds than in the microfibrous scaffolds without


nanofibers
Highly porous scaffold provided mechanical support [171]
for cells to maintain uniform distribution; better
cell migration and collagen secretion
PCL/Chitosan Composite The presence of chitosan aided a significant improve- [172]
ment in the hydrophilicity of the scaffold, bioactiv-
ity and protein adsorption
Amino-remained chitosan-graft-poly (ε-capro- [173]
lactone) (2/8) mats with a moderate surface
zeta-potential (ζ=3 mV) were the best in promot-
ing the cell attachment and proliferation
Novel polysaccharide-coated PCL fibre mats remark- [174]
ably combine the mechanical resistance typical of
PCL with the surface properties of chitosan
PCL/Collagen Composite Anisotropic nanofibers significantly triggered integrin [175]
β1 signalling pathway
Collagen was immobilized on aminolysed PCL nano- [176]
fibers using glutaraldehyde as cross linker, enhan-
cing cell proliferation
PCL and collagen nanofiber matrices support the [177]
attachment and proliferation of human dermal
fibroblasts; suitable for dermal substitutes
Coaxial electrospinning of collagen-PCL showed [178]
superior cell proliferation than roughly collagen-
coated PCL
Acellular scaffold strength increased with increasing [179]
PCL content but the mechanical strength of the
Engineered Skin was not enhanced by the use of
collagen–PCL blended scaffolds, after a minimum
amount of PCL (10%)
Collagen gel coated PCL nanofibrous scaffolds [180]
showed better cell responses than PCL/collagen
nanofibrous matrices
PCL/Gelatin Composite Coaxially electrospun PCL/gelatin scaffolds exhibited [181]
increased cellular adhesion and metabolism versus
PCL alone or gelatin-PCL blended fibre scaffold
Acetic acid-doped TFE solvent system was used to [182]
prevent phase separation during electrospinning
PCL/Gelatin/ Composite Collagen type I-modified PCL/gelatin scaffold was [183]
Collagen successful in maintaining characteristic shape of
fibroblasts, besides good cell proliferation than the
PCL/gelatin scaffold
PLA/Chitosan Composite Core–shell structure nanofibers of PLA/chitosan [184]
easier to fabricate than double-needle
electrospinning
Aligned PLA/chitosan fibrous scaffold fabricated [185]
using a novel collector made of parallel blades;
PLA/chitosan nanofiber has enhanced degradation
than the pure PLA fibres
PLA/Gelatin Composite Modified PLA/gelatin (7/3) scaffold is more suitable [186]
for fibroblasts attachment and viability than the
PLA or gelatin nanofiber alone
PDLLA/ Composite Dual-layer chitosan/PDLLA structure result in [187]
Chitosan aligned migration and directional penetration of
skin fibroblasts into 3D domain
PLLACL/ Composite PLLACL/collagen scaffolds showed better cell differ- [188]
Collagen entiation of MSCs than PLLACL scaffolds
PLLACL/ Composite Fibroblasts proliferation, morphology, CMFDA dye [189]
Gelatin expression and secretion of collagen were sig-
nificantly increased in plasma-treated PLACL/
gelatin scaffolds compared to PLACL nanofibrous
scaffolds

13
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 6. (Continued.)
Polymer Type Special feature Reference

PLGA/Chitosan Composite Hybrid PLGA/chitosan and core/shell structured [190]


PLGA/chitosan membranes showed better
cytocompatibility than the PLGA membrane in
adhesion, viability assays as well as morphology
observation
PLGA/Collagen Composite Highly porous fibrous mats of PLGA and Type [191]
I collagen were produced by blend electrospinning;
inoculation of collagen enhanced the cell
attachment, proliferation and extracellular matrix
secretion, which were found to be dependent on
the amount of collagen in the composite scaffold
PLGA/collagen nanofibrous membranes were func- [192]
tionally active in responses in human fibroblasts,
and were very effective as wound-healing
accelerators in early-stage wound healing
PCL/PLGA/ Composite The upper layer of PCL/PLGA nanofibers provided [193]
Chitosan/ mechanical support and reduced degradation rate
Gelatin of the hydrogel layer. The lower layer of porous
chitosan/gelatin hydrogel could retain moisture.

present in order to make it fully functional per se. regeneration and development should be understood
Irregular pigmentation, for example, is a limitation, well, when undifferentiated stem cells are used as the
which can be overcome by co-culturing melanocytes source from which differentiated cell types arise,
along with keratinocytes, which ensures normal pig- thereby creating or regenerating functional tissues
mentation by secreting melanin [208]. Vascularization [210]. Also, there are many ethical and technical pro-
is another problem, which can be managed to some blems, controversies surrounding stem cells to be
extent by incorporating endothelial cells to facilitate overcome before we can attempt bioprinting stem
angiogenesis. Genetically modified keratinocytes, cells, utilising their full potential [216–218].
which overexpresses vascular endothelial growth fac- Having reviewed the types of cells that can be used
tor (VEGF) were reported to promote vascularization for skin substitutes, 3D bioprinting has the luxury of
[209]. Still, integration of pilosebaceous units, with using any kind of cells, or combination of cells, sus-
hair follicles and sebaceous glands to the skin sub- pended in the same solution or bio-ink or different
stitutes, remains an unsolved challenge [202]. cells in different bio-inks, that can be fed through dif-
Most of the current tissue engineering strategies ferent nozzles in a multi-nozzle bioprinting system.
depend on a sample of autologous cells from the Based on the type of cells chosen, the bio-ink composi-
damaged host tissue or organ. It becomes increasingly tion varies in order to satisfy the basic requirements of
difficult to obtain the cells from the damaged host, a printable bio-ink.
especially when the injury or damage is extensive.
Alternative cell sources become a necessity in such 5.4. 3D bioprinting processes
cases, which is debated in the field of regenerative Bioprinting can be defined as a ‘computer-aided transfer
medicine. In order for the tissues and organs to main- processes for patterning and assembling living and non-
tain a balance between cell loss and replacement, the living materials with a prescribed layer-by-layer stacking
tissues must possess cells that are capable of self- organization in order to produce bio-engineered struc-
renewal and differentiation and hence, stem cells are tures serving in regenerative medicine and other biologi-
looked upon as a promising source for future skin cal studies’ [219]. The bioprinting techniques can be
regeneration and skin tissue engineering [210]. Many classified under three main topics namely, Laser-based
types of stem cells that inhabit the skin have been iden- bioprinting or laser-induced forward transfer [80, 220–
tified and located which includes epidermal stem cells 225], Inkjet bioprinting/droplet-based bioprinting
(interfollicular stem cells and bulge stem cells), that are [226–231], and robotic dispensing/extrusion/deposi-
widely used recently, dermal stem cells [211], sebac- tion bioprinting [232–239] (refer to figure 7).
eous stem cells, melanocyte stem cells, hair follicle Laser-induced forward transfer utilizes laser energy
stem cells [212, 213], sweat gland stem cells, endothe- to deposit cell-encapsulated droplets of required
lial stem cells, mesenchymal stem cells (MSCs), hema- volume, in any defined 3D spatial arrangement. The
topoietic stem cells (HSCs), and neural stem cells main components of such a system are the donor slide,
[214, 215]. When using stem cells for bioprinting or the collector slide and laser generation system. The
tissue engineering in general, the relationship between donor slide has a conductive coating for laser energy

14
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Table 7. Ideal bioprinting hydrogel properties [197]. suspensions or bioink, down to a few hundred femto-
litres, with high resolution [220, 245] and ability to print
Printability Viscosity
Shear-thinning property high density cell suspensions [246] or hydrogel pre-
Response and transition time cursors or bioink with any desired viscosity, which is
Sol-gel transition stimulus the main limitation with ink-jet bioprinters, which can
Biocompatibility Degradability only handle lower viscosity ranges. Schiele et al [247]
Cell binding motifs comprehensively reviewed the laser-based direct write
Non-toxic cell printing technologies, giving an overview of differ-
Non-immunogenic
ent realisations and denominations of this technique.
Mechanical properties Stiffness
Elasticity
Research group from Hannover Medical School, Ger-
Strength many along with Laser Zentrum Hannover, had used
Shape and structure Pore size this technique to print skin tissue [248, 249].
Micro/Nano structure Koch et al [248] used LaBP to arrange the vital cells
of skin tissue, namely the keratinocytes and fibroblasts
embedded in collagen, in a 3D arrangement, repre-
absorption and a layer of the cell suspension or bioink senting multicellular skin grafts that are analogous to
that is to be bioprinted. The collector slide acts as the the native archetype. The bio-ink used in this study is
substrate, which can be biopaper or scaffolds or culture made of (a) murine cell line NIH-3T3 fibroblasts,
plates. When laser pulses are focussed on the donor representing the dermal layer, (b) keratinocytes cell
slide, the conducting layer absorbs the laser energy, cre- line HaCaT from adult human skin, representing the
ates a high gas pressure on absorption of laser energy epidermal layer, (c) suspended in a hydrogel, collagen,
and as a result, droplets of cell suspension are propelled which is the main component of ECM in native skin,
towards the collector slide. Required spatial arrange- and (d) a sheet of MatridermTM, which is a commer-
ment can be obtained by controlling the path of the cial dermal skin substitute, to serve as a scaffold or
laser beam. Inkjet printing is the second bioprinting substrate. The main focus of this study was the
technique. They are also called Drop-on-Demand sys- arrangement of these skin cells under a set of different
tems as it propels drops of cell suspension or bioink on conditions or parameters, namely 3D cell pattern, cell
to the substrate. Based on the mechanism of how dro- density and matrix material, in a way that the micro-
plets are generated, they are classified as thermal and environment is comparable to the native skin tissue
piezoelectric based inkjet printing. In thermal inkjet and to study the tissue formation process. The exper-
printing, a heater or a heating filament produces a imental set-up [250] is shown in figure 8. It consists of
vapour bubble in the nozzle, which propels the droplet two co-planar glass slides. The upper one is coated
out of the nozzle onto the substrate. In piezoelectric underneath with an energy absorbing layer or laser
based systems, the changes in the volume of piezo- absorbing layer, to be specific; a 60 nm thin gold layer
coating is used in this study and a layer of the cell sus-
electric crystal on application of electric pulses, expels
pension that is to be transferred or bioprinted, which
the droplet through the nozzle. Third comes the robotic
is keratinocytes or fibroblasts suspended in collagen
dispensing systems. In this type, the bioink is extruded
hydrogel. The lower glass slide acts as the substrate;
out of the nozzle by application of mechanical force.
scaffold or hydrogel layer can be positioned on this
The mechanical force can be pneumatic, piston-based
plate on which the cells will be printed. For better
or screw type. An extensive review of these technologies
understanding, the upper glass slide is the ‘donor-
has been done by Murphy et al [116], Dababneh et al
slide’ and the lower glass slide is the ‘collector-slide’.
[219], Malda et al [194] and Ozbolat et al [240]. Arslan-
The cell suspension is printed on to the collector-slide
Yildiz et al [241] had reviewed the recent advances in
from the donor-slide using laser induced forward
bioprinting technologies, current markets, approaches,
transfer. Laser pulses are focussed on the donor slide,
and biomedical applications of bioprinting.
which causes localized evaporation in the gold layer
beneath, the absorption of laser energy generates a
5.4.1. Laser assisted Bioprinting (LaBP) of skin tissue high gas pressure, thereby propelling the subjacent cell
Current 2D cell culture and 2D tissue engineering do suspension towards the lower glass slide, which is the
not well mimic the native tissue as the original natural collector-slide. The droplet size of the cell suspension
tissue is in a 3D environment. Studies have already transferred depends on a number of parameters
proved that cells behave differently in 2D and 3D namely the laser pulse energy, the gap between the
environment and it is better with in 3D space as it upper and lower glass slide, the laser spot size, thick-
mimics the complex 3D-microenvironment architec- ness of the laser absorbing layer and the thickness of
ture in vivo [242–244]. Laser cell printing or Laser the cell suspension layer.
assisted Bioprinting (LaBP) or Laser induced forward A bilayer skin construct, consisting of keratino-
transfer, is one technology which can precisely place cytes dominated outer epidermal layer and fibroblasts
cells in 3D spatial arrangement. Advantages of LaBP dominated the inner dermal layer was printed; 20 cell-
includes capability of positioning small volumes of cell collagen sublayers of NIH-3T3 Fibroblasts being

15
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 7. Three main bioprinting technologies (a) Laser-assisted bioprinting focuses laser pulses on to the donor slide, thus creating
high pressure to propel droplets of cell-laden hydrogel on to the collector slide (b) Inkjet printing ejects droplets of biopolymer or cell-
laden hydrogels through a nozzle by either thermal energy application (electrically heating to produce vapour bubbles that forces
droplets to come out through the nozzle) or piezoelectric actuator (actuation of piezoelectric crystals by applying electrical energy at
high frequencies). (c) Extrusion or robotic dispensing bioprinters extrude biopolymers or cell-laden hydrogels through the nozzle by
applying air pressure (pneumatic) or mechanical systems (piston or screw).

printed first on a sheet of MatridermTM, followed by of each printing process served as in vitro controls.
20 cell-collagen sublayers of HaCaT keratinocytes Two of them were directly fixed on Day 0 to depict the
over it. situation at the beginning of the experiments, whereas
Important factors that indicate a successful skin tis- the remaining four pieces were raised to the air-liquid-
sue construct were studied on the printed bilayer con- interface. In vitro controls were then fixed on Days 5
struct including immunohistochemistry, analysis of and 11 (duplicates per time point) and in vivo speci-
gap junction coupling, post-printing cell proliferation, men on Day 11.
development of a basement membrane between epi- The results were promising. The printed skin con-
dermis and dermis like the natural skin, analysis of structs placed in to the full-thickness wounds in the
adherens and gap junctions (which is important for dorsal skin chamber of nude mice, in vivo, were fully
mechanical connection between cell-cell and cell- connected to the surrounding tissue when explanted
ECM). The cells remained in the printed pattern, i.e. as after 11 days. Multi-layered epidermis was formed by
bilayer, and did not intermix even after 10 days in cul- the printed keratinocytes, with initial differentiation
ture; proliferation post-printing was good; develop- and stratum corneum. Adherens junctions, which are
ment of the basal lamina serves as a sign of skin tissue the main indicators of tissue formation, detected by
formation; better adherence of the cells to neighbouring the E-cadherin, could be found in the epidermis, both
cells and the ECM shown by the extensive formation of in vivo and in vitro. The printed fibroblasts produced
adherens junctions all go to say that this was a unique collagen, which is their main function, in both in vivo
successful first demonstration of 3D arrangement of and in vitro conditions, staying partly on the top of the
vital cells using LaBP method as multicellular grafts underlying matrix and partly migrated in to the
analogous to the native skin. matrix. Above all, some blood vessels were found
Michael et al [249], from the same group, used the growing from the wound bed and the wound edges
same technique to print the bilayer skin construct and towards the printed cells.
tested it in vivo in nude mice. In this work, same kind
of cells used in the previous study namely NIH3T3 5.4.2. Robotic dispensing based 3D-bioprinter
Fibroblasts and HaCaT keratinocytes suspended in Lee et al [251] reported a novel method to construct
collagen were used. A bilayer skin construct, having 20 stratified skin construct in 3D, layer by layer, using
layers of fibroblast-containing collagen and 20 layers robotic cell dispensing technology. The schematic of
of keratinocyte-containing collagen were printed onto the device is given in figure 9. The printer mainly
a sheet of MatridermTM (2.3 cm×2.3 cm, followed consists of an array of four micro valves as dispensers
by incubation under submerged conditions overnight and a three-axis Cartesian robotic stage. The dispen-
in an incubator. The next day (defined as Day 0), nine sers, each with a pneumatically driven control mech-
round pieces (diameter 6.0 mm) were removed from anism, were mounted to the horizontal (x–y) robotic
the large construct with a biopsy punch, three of which stage, which controlled the timing and location of
were implanted into the skin fold chambers in vivo dispensing of printable materials including hydrogel
(one per mouse, four independent printing processes, precursors and cell suspensions. The entire device was
tested in 12 animals in total). The remaining six pieces housed in a laminar flow hood. The MATLAB

16
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 8. Top left: sketch of the laser printing setup. The cell–hydrogel compound is propelled forward as a jet by the pressure of a
laser‐induced vapour bubble. Layer‐by‐layer, a 3D cell pattern is generated. Top right: A printed grid structure (top view) of fibroblasts
(green) and keratinocytes (red) demonstrates micropatterning capabilities of the laser printing technique. Bottom: Seven alternating
colour‐layers of red and green keratinocytes (left; detail right). Each colour‐layer consists of four printed sublayers. Scale bars are
500 μm. Reprinted with permission from [248].

computing environment was used to generate the reported to be the cap for thickness of printed tissues
robot control codes dictating the dispensing spatial at 100 um, as the resolution achieved was in the order
coordinates. Primary adult human dermal FBs and of 700 to 1000 um. Improvements in the dispensing
primary adult human epidermal KCs were used along system and printing mechanism should be made for
with Type I collagen as hydrogel precursor for a increasing the resolution. With thicker tissues comes
scaffold material. A total of 10 collagen layers were the problem of providing adequate perfusion, which
sequentially printed on a planar square in a 60 mm can be overcome by introducing fluidic channels
tissue culture dish. FB and KC layers were located in inside the hydrogel.
second and eighth layer of the collagen layers, counted Lee et al [252] utilised a similar system but with
from bottom, respectively. The five collagen layers eight independently controlled cell-dispensing chan-
sandwiched between the FB and KC layers demon- nels, instead of four, which can be utilised to place cells,
strate the ability to print spatially distinctive cell layers. ECM, scaffold materials, and other growth factors in
Post-printing, the printed tissue construct was cul- any user-defined 3D arrangement. Each dispenser is
tured in 37 °C, 5% CO2 in KC media. There are two independently operated by electromechanical valves, as
important and unique advantages of this device. The in the previous system [251] and mounted on a three-
first is the new crosslinking mechanism introduced axis, high precision, xyz robotic stage, with the option
and the second is printing on non-planar surfaces. to precisely control the volume of dispensed droplets,
Instead of the traditional method of dipping the with range as low as 15 nL, with high precision. FBs
printed construct into the crosslinking solution bath, (HFF-1) and KCs (HaCaT) were used to construct the
here, the nebulized mists of crosslinking agents were 3D skin tissue in this study. The schematic is shown in
coated on the surface before and after printing each figure 10. A total of six collagen layers for the dermis
layer. This new mechanism engenders the second and two collagen layers for the epidermis were printed
advantage of printing on non-planar surfaces, which to obtain the desired dermal and epidermal thicknesses.
was successfully demonstrated by printing on 3D non- FBs proliferated within the dermal layer and main-
planar Polydimethylsiloxane (PDMS) substrates. This tained a sparse distribution. KCs proliferated more
characteristic of this method makes it possible to print rapidly on the top of the collagen matrix and fully cov-
the tissues directly on to the wound surfaces in vivo, ered the surface in 4–7 days. No cell invasion across the
even on the most complex, non-planar, irregular dermal and epidermal layers was observed, which is a
wound area. One limitation of this system was notable advantage. A comparison of skin tissues

17
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 9. (Top) Picture of the modular tissue printing platform shown with: [1] 4 syringes as ‘cartridges’ to load cell suspensions and
hydrogel precursors; [2] an array of 4-channel dispensers; [3] target substrate; [4] horizontal stage; [5] vertical stage; [6] range finder;
[7] vertical stage heater/cooler; [8] optional independent heating/cooling unit for the dispenser. Inset: close-up view of the 4-channel
micro-dispensers. (Bottom) Schematic of layer-by-layer printing of the multi-layered skin cells and collagen. Reprinted with
permission from [251].

fabricated via 3D bioprinting and manual deposition enable the printed collagen strands to freeze almost
was made, which showed that printed skin samples instantly as it touches the substrate. The nozzle was
retain their form, dimensions and shape, whereas coated with silicone rubber to prevent clogging. The
manually deposited structures shrink, buckle and form plotted scaffold was immediately placed in a freeze-
concave shapes under submerged culture condition dryer at −76 °C for 3 days, followed by immersion in
after 7 days. In vitro tests namely the viability tests and 50 mM 1-ethyl-(3-3-dimethylaminopropyl) hydro-
immunohistochemistry tests indicated the develop- chloride (EDC) solution in 95% ethanol for 24 h at
ment of the printed construct in to a skin tissue, room temperature for cross-linking. Keratinocytes and
mimicking the epidermal and dermal layers of fibroblasts are co-cultured on the scaffolds then and
native skin. in vitro studies performed. The well designed pore
structure of the scaffold enabled the cells to proliferate,
5.4.3. Cryogenic extrusion based direct-plotting system readily migrate in to and differentiate in the scaffold.
Kim et al [253, 254] studied an extrusion based However, the structure suffered low shape stability due
cryogenic direct-plotting system for skin tissue engi- to highly porous structure and poor mechanical proper-
neering. The device set-up is shown in figure 11 ties of collagen.
[253]. The cryogenic 3D plotting system composed of In order to overcome the limitation of poor
a 3D robot system, a dispensing system, and a mechanical properties, Kim et al [254] plotted a core/
cryogenic manufacturing stage. The cryogenic refrig- shell structure scaffold using the same device set-up.
eration system consisted of two compressors, circu- Alginate and collagen were printed coaxially with
lating silicone oil to cool the stage, and a circulation alginate providing required mechanical strength to
pump. A collagen scaffold of required thickness and the scaffold. In vitro tests proved that the core/shell
pore size were processed by computer-driven direct scaffold had completely similar cellular behaviour to
deposition, layer by layer with a plotting needle under that of the pure collagen scaffold, with the added
controlled pneumatic pressure on a cryogenic stage at advantage of enhanced mechanical stability. In addi-
−40 °C and the ambient temperature being 10 °C, to tion to in vitro, in vivo studies had been conducted on

18
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 10. Construction of three-dimensional (3D) skin tissue. (a) Layer-by-layer printing of collagen matrix, KCs, and FBs to
construct the dermal and epidermal compartments in a single structure. (b) Schematic of the 3D printed skin tissue showing the cross-
section (left) and top view (right). Reprinted with permission from [252].

adult male BALB/c/Bkl-nu/nu mice. The plotted Additional layers located above can contain a different
scaffold with co-cultured keratinocytes/fibroblasts, cell suspension, like keratinocytes (though not used in
implanted as a dermal substitute on the wound area, this study), or focussing streams containing the cross
provided good granulation tissue formation and linker can be printed, indicated by red colour streams
rapid vascularization in the dermis and the scaffold. in figure 12. There was a rotating drum downstream
This technique can be used to co-extrude other nat- that collected the printed sheets, which in addition to
ural or synthetic biopolymers along with collagen or the shear stress exerted by the focusing fluid, aids in
even cell-laden bioink and expanded further with continuous formation of hydrogel or bioink sheets,
other growth factors and peptides in the core area to image based inspection and better collection. Human
make functional skin substitutes. However, one dis- fibroblast cells were incorporated within a continu-
advantage of this system is that the whole process
ously extruded biopolymer sheet with precise spatio-
takes place in a cryogenic environment unlike the
temporal control over the cell location and cell seeding
other systems where the processes take place at room
density (which is 1.94 million cells/mL in this study).
temperature.
Various shapes of biopolymer sheets like spots and
parallel strips were printed, with fibroblasts seeded in
5.4.4. Microfluidic approach based skin printer
to the structure. The patterns can also be hollow to
Leng et al [255] from University of Toronto, developed
promote vascularization while providing relevant
a skin printer based on a microfluidic approach, which
is widely accepted as a technique that will aid in thickness. In vivo testing was carried out on immuno-
vascularization of tissue engineered constructs. The deficient mice by performing excisional skin biopsy
experimental schematic is shown in figure 12. The and replacing the excised skin with printed biopoly-
device [256] consists of an annular gear pump, a mer sheets patterned with fibroblasts. Trichrome and
microfluidic flow focussing device or a micro-fabri- keratin 14 staining on the wounded site revealed that
cated cartridge and a collecting drum. The ECM used the printed skin construct led to keratinization and
in this study is a mixture of alginate and collagen. The better wound healing. The greatest advantage of this
base layer, which is indicated in blue colour shown in device is the shortest lead time for printing skin, which
figure 12, carried to the device exit via a set of parallel is reported to be 10 mm s−1, i.e. it will take only
microchannels, a time-varying content of the bioink. 48 min to print a 1 m2 skin construct.

19
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 11. Fabrication procedure for 3D collagen scaffolds. (a), Schematic diagram of the desired scaffold (20X20X1.5 mm3); (b)
schematic diagram of the cryogenic plotting and processing system at −40 °C (left) and images of the fabricated scaffold (right); (c)
complete freeze-drying of the fabricated 3D scaffold at −76 °C over 3 days. (d) Electron micrograph of scaffold curing in 50 mM EDC
in 95% ethanol for 24 h at room temperature. Scale bar=1 mm. Reproduced from [253] with permission of The Royal Society of
Chemistry.

5.4.5. In situ bioprinting of skin gathered data is processed to obtain a functional


Binder et al [257] from Wake Forest University wound map and based on the wound map the software
developed a novel device for in situ bioprinting of skin. determines the exact cell composition and thickness of
The criteria for developing this system includes: (i) different areas of skin. The most important compo-
portability; the system must be capable of being nents of the device are cartridge-based delivery system,
quickly transported to patients with extensive burn consisting of a number of nozzles, laser scanner for
wounds, (ii) must accommodate a wide range of body wound scanning, the mobile frame, and XYZ axes. In
types, (iii) capable of tailoring cell therapy to indivi- vivo experiments were performed on the porcine
dual patient’s specific needs, (iv) ease of sterilization, wound model, as porcine skin is similar to human
(v) allows for repeated treatments, and (vi) inexpensive skin. The bioprinting procedure was divided in to two
and easy maintenance. The first two criteria were stages. In the first stage, skin from the shoulder area of
accomplished using a movable lightweight frame on the pig was removed using a dermatome for prep-
which an XYZ plotting system was mounted. The aration of fibroblasts and keratinocytes for bioprint-
dimensions of the system are large enough to cover the ing. These cells isolated were then cultured and mixed
torso of an average patient but small enough to easily with biopolymers namely fibrin and type I collagen to
pass through most doorframes. During the treatment, make the bioink. After expanding these cells in culture,
the system is positioned over the patient’s wound area four full-thickness 10 cm×10 cm wounds were
and locked at the optimal position. The powerful locks excised through the panniculus carnosus layer of the
prevent the frame movement while bioprinting, thus thoracic dorsa on the animals. Prior to surgery, the
preserving both the mobility of the system and its print head, cartridges, and tubing were sterilized. No
delivery precision. A belt-driven plotting system with part of the printer was in direct contact with the animal
100 μm precise movements aids the XYZ axes in at any point during the printing procedure. The
precision delivery of cell suspension on to the patient’s wound area was placed under a portable bioprinter
body. The movement of Z axis can be independently and dermal cells and matrices were directly bioprinted
controlled, thereby allowing the delivery system to onto the wound bed. One layer of 10 million
track with the curvature of patient’s body and thus fibroblasts (1.0×105 cells cm−2) was bioprinted at
capable of accommodating many different body types. 2 mm intervals between drops with thrombin sprayed
A laser based wound scanning system was coupled from the atomizing nozzles simultaneously to form a
with the cartridge-based delivery system which helps fibrin/type I collagen hydrogel, followed by a 15 min
in tailoring the cell therapy according to individual break to allow complete conversion of fibrinogen to
patient’s need. The laser scanning system gathers fibrin and finally another layer of 10 million keratino-
length, width, and depth data about the wound; cytes was bioprinted above the fibroblast layer. Results

20
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

Figure 12. Illustration of proposed skin printer for formation of cell-populated skin substitute in a microfabricated printer cartridge
and application in vivo. The base layer, which is indicated in blue colour is carried to the device exit via a set of parallel microchannels.
Additional layers located above can contain a different cell suspension or focussing streams containing the crosslinker can be printed,
indicated by red colour streams [255]. Reproduced with permission from Chemical and Biological Microsystems Society.

were very much promising. Complete re-epithelializa- encapsulated fibres and particles having sub-micro-
tion could be witnessed at 8 weeks. Keratinocytes metre to micrometre scale dimensions were success-
proliferated over the course of 8 weeks to completely fully generated with the spray gun at different angles
cover the wound and their epithelium was found relative to a defined reference angle and deposited in a
connected to the advancing wound edge. Pre-labelled marked area of interest, proving its flexibility and
autologous fibroblasts and keratinocytes as well as portability. Though no cell laden bioink have been
allogeneic fibroblasts were visible in the wound at 8 printed and tested, the spray gun technology can
weeks, demonstrating that bioprinted skin cells can evolve in to a portable handheld in situ bioprinter in
survive, proliferate, and form skin tissue in an the future, facilitating ease of use by the clinicians.
immuno-competent large wound model. One limiting The summary of all the skin bioprinting technolo-
factor reported is that the performance of the laser gies are given in table 8.
imaging system is not up to the mark in vivo because of
the breathing motion of the patient. This could be 6. Challenges and future prospects
overcome by replacing the laser scanner with imaging
system that can take instantaneous snapshots of the Having reviewed in detail the various processes of 3D
wound and create the wound map. Certain optimiza- bioprinting skin and its advantages, it is important to
tions are also required to be made before using it for look at the realistic deployment potential of these
treating human wounds. technologies clinically. No technology is perfect and all
Sofokleous et al [258] worked on developing a por- of them suffers from certain limitations and pose
table handheld electrohydrodynamic (EHD) multi- challenges. 3D bioprinting is not an exception too.
needle spray gun for biomedical applications. The Current skin grafts suffer from the greatest limitations,
main component of the EHD spray gun is a multi-nee- in terms of long healing time and cost, which is reported
dle device assembly with 1–2 brass needles, a dremel to be over $50 000 to cover 40% burn area on an average
glue gun used as the spray gun case heat-shrinkable male [66, 69, 259, 260]. Though 3D bioprinting has the
tubing with adhesive inner liner, high current wire, potential to reduce the healing time considerably, the
and silicone tubes. The number of brass needles can cost factor should be taken in to account. Affordability
range from one up to four, based on the application. is the first challenge that is to be overcome. The next
The working principle of an EHD spraying/spinning biggest challenge is to bioprint a fully functional skin
of liquids is application of an electric force to the sur- substitute. Though most of the functions of the native
face of a conducting liquid to produce fibers and such skin can be mimicked in the bioprinted skin by using
a system consists of a high voltage supply, which cre- more types and number of cells in addition to
ates the required electric field between a charged capil- keratinocytes and fibroblasts, like the melanocytes or
lary filled with the material to be processed in liquid Langerhans cells or even hair follicle cells, path to
state and a ground collector. When the electrostatic vascularization and innervation in engineered tissues
force overcomes the surface tension of the liquid at the still remains murky. Use of stem cells is one option but
capillary tip, a fine jet emanates from the nozzle tip. the ethical issues, cost and skill requirement hinders
PLGA and Polymethysilsesquioxane (PMSQ) solu- from utilising them to their full potential. Genetically
tions were tested using the device. Simple, multi- modified keratinocytes, which overexpresses vascular
component and multifunctional products such as endothelial growth factor (VEGF) were reported to

21
Biofabrication 8 (2016) 032001
Table 8. Summary of skin bioprinting technologies.

Cellular In vivo animal


Uniqueness components Size of the tissue construct Cell viability Resolution Speed model In vivo results Scalability References

Laser based First demonstra- FBs (Murine 10 mm×10 mm×2 mm Proved vitality of Droplet — — — Low [248]
bioprinting tion of 3D cell line the printed cells volumes
arrangement of NIH-3T3) 10 days after of
vital cells by and KCs printing 0.1–1 nL
LaBP as multi- (cell line
cellular grafts HaCaT
analogous to from adult
native arche- human
type; the indivi- skin)
dual layers of embedded
the skin con- in collagen
22

struct do not
intermix with
each other
Possibility to posi- NIH3T3 FBs — Normal cell — — Male BALB/c- After 11 days, the Low [249]
tion different and HaCaT viability Nude mice borders of the
cell types in an KCs sus- skin constructs
exact three- pended in and the sur-
dimensional collagen rounding mouse
(3D) spatial skin were tightly
pattern grown together;
neither inflam-
matory/necrotic
processes nor
contraction of
the wounds
could be detec-
ted; small blood

S Vijayavenkataraman et al
vessels could be
found.
FBs and KCs, NIH3T3 FBs 9.6 mm×9.6 mm FBs and KCs Droplet sizes 1200 printed — — Low [250]
along with and HaCaT showed expo- of 80 to cell dro-
hMSCs were KCs, Bone nential growth 140 μm plets per
printed using marrow– starting on day diameter minute
Biofabrication 8 (2016) 032001
Table 8. (Continued.)
Cellular In vivo animal
Uniqueness components Size of the tissue construct Cell viability Resolution Speed model In vivo results Scalability References

laser bioprint- derived 1 up to day 6;


ing due to their hMSC continuously
high potential (Bm- increasing pro-
in regeneration hMSCs), liferation of
of human skin Adipose- Bm-hMSC until
and new appli- derived day 4 after
cation possibi- hMSC printing
lities of stem
cell therapy
Robotic A method to cre- Primary adult — There was no sig- Droplet — — Medium [251, 252]
dispensing ate multi- human nificant differ- volumes
layered engi- dermal FBs ence in cell of
23

neered tissue and pri- viabilities 7.6–10 nL


composites; mary adult between printed
direct, on- human epi- cells and manu-
demand fabri- dermal ally plated cells
cation of the 3D KCs, Type I as demon-
tissue compo- collagen strated in the
sites on non- viability assay
planar surfaces
[250]; First
study of print-
ing both FB and
KC in a single
experiment
with the forma-
tion of dermal/
epidermal-like

S Vijayavenkataraman et al
distinctive lay-
ers in a 3D
hydrogel scaf-
fold [251]
Cryogenic A novel system for Normal 18.4 mm×18.3 mm×1.4 mm Good cell migra- — — — — Low—Medium [253]
plotting the cryogenic human tion and differ-
plotting of 3D KCs and entiation;
Biofabrication 8 (2016) 032001
Table 8. (Continued.)
Cellular In vivo animal
Uniqueness components Size of the tissue construct Cell viability Resolution Speed model In vivo results Scalability References

scaffolds devel- FBs; Type I stratum cor-


oped to over- collagen for neum was well
come the fabricating expressed simi-
problems in porous lar to the nor-
fabricating scaffolds mal skin tissue
highly hydro-
philic natural
polymers into a
scaffold.
Cryogenic The hybrid core/ Normal 10 mm×10 mm×2 mm Cell proliferation — — Adult male Provided good Low—Medium [254]
plotting shell (collagen/ human of core/shell BALB/c/Bkl- granulation der-
alginate) scaf- KCs and (collagen/algi- nu/nu mice mal tissue for-
24

folds fabricated FBs; Type I nate) scaffold mation and rapid


by cryogenic collagen was completely vascularisation;
plotting, exhib- and algi- similar to that the epidermis
ited good struc- nate for of the pure col- was completely
tural stability, fabricating lagen scaffold covered after
increased porous 14 days.
Young’s mod- scaffolds
ulus about
seven times
compared to
pure collagen
scaffold, and
good cell
viability.
Microfluidic A scalable Human FBs up to 35 mm wide sheets Normal cell — 10 mm s−1 Immunodeficient Better wound heal- High [255, 256]
Skin approach and viability equivalent mice ing and keratini-

S Vijayavenkataraman et al
printer uniquely to 1 sq.m zation, suggest-
addresses the in 48 min ing an improved
need to provide skin
skin substitutes regeneration
at affordable
prices that are
easy to handle
Biofabrication 8 (2016) 032001
Table 8. (Continued.)
Cellular In vivo animal
Uniqueness components Size of the tissue construct Cell viability Resolution Speed model In vivo results Scalability References

and apply,
reduced wound
recovery times;
enables the
continuous for-
mation of cell-
populated sin-
gle-layer,
bilayer, and
vascularized
sheets
In situ skin Fist demonstra- FBs and KCs 10 cm×10 cm — — — Porcine wound Complete re-epi- Medium [257]
25

printer tion of in situ harvested model thelialization


skin printing on from Por- could be wit-
porcine wound cine model nessed at 8
model suspended weeks; KCs pro-
in fibrin liferated over the
and Type I course of 8 weeks
collagen to completely
cover the wound
and their epithe-
lium was found
connected to the
advancing
wound edge.

S Vijayavenkataraman et al
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

promote vascularization [209] and it is worth trying to [4] Boyce S T 2001 Design principles for composition and
use them to see how well it can help the vascularization performance of cultured skin substitutes Burns 27 523–33
[5] Nickoloff B J, Mitra R S, Green J, Zheng X G, Shimizu Y,
in bioprinted tissues, provided with all the other growth
Thompson C and Turka L A 1993 Accessory cell function of
factors and tissue environment in a bioreactor. The keratinocytes for superantigens. Dependence on lymphocyte
neural crest stem cells are a multipotent cell population function-associated antigen-1/intercellular adhesion
that can give rise to a diversity of neural and non-neural molecule-1 interaction J. Immunol. 150 2148–59
[6] Albanesi C, De Pità O and Girolomoni G 2007 Resident skin
cell types in the adult, such as melanocytes, neurons,
cells in psoriasis: a special look at the pathogenetic functions
and glial cells of the sensory and autonomic ganglia of of keratinocytes Clin. Dermatol. 25 581–8
the peripheral nervous system, and some endocrine [7] Schröder J M 2010 The role of keratinocytes in defense
cells [215, 261]. All these enhancements will benefit the against infection Curr. Opin. Infect. Dis. 23 106–10
[8] Tsuruta D 2009 NF-κB links keratinocytes and lymphocytes
wound healing application, where the bioprinted skin
in the pathogenesis of psoriasis Recent Pat. Inflamm. Allergy
substitute will be implanted on a patient’s body or Drug Discov. 3 40–8
printed in situ, either way, the healthy part of the skin [9] Ranzato E 2013 Keratinocytes: Structure, Molecular
can provide nourishment to the grafted part. But, the Mechanisms and Role in Immunity (New York: Nova Science)
[10] Nolte S V, Xu W, Rennekampff H O and Rodemann H P 2008
drug testing and cosmetics testing remains a challenge
Diversity of fibroblasts–a review on implications for skin
as the skin substitutes will not be grafted on to humans tissue engineering Cells Tissues Organs. 187 165–76
but it requires a biomimetic skin substitute ex vivo, with [11] Millioni R, Puricelli L, Iori E, Trevisan R and Tessari P 2012
all functionality of the native skin so that the drugs or Skin fibroblasts as a tool for identifying the risk of
nephropathy in the type 1 diabetic population Diabetes
cosmetics can be tested accurately. Application in
Metab. Res. 28 62–70
personalized or precision medicine is another prospect [12] Jordana M, Sarnstrand B, Sime P J and Ramis I 1994
which can be benefitted much with 3D bioprinting of Immune-inflammatory functions of fibroblasts Eur. Respir. J.
skin. As a matter of fact, all the steps in bioprinting skin 7 2212–22
[13] Kalluri R and Zeisberg M 2006 Fibroblasts in cancer Nat. Rev.
explained in this paper, right from imaging of wound
Cancer 6 392–401
site to the application, makes it a natural choice for [14] Takeda K, Takahashi N H and Shibahara S 2007
personalized or precision medicine care. Another Neuroendocrine functions of melanocytes: beyond the skin-
potential application is the study of melanoma and deep melanin maker Tohoku J. Exp. Med. 211 201–21
[15] Le Poole I C, Mutis T, Van Den Wijngaard R M,
other types of skin cancer. In vitro 3D tumour models
Westerhof W, Ottenhoff T, De Vries R R and Das P K 1993 A
with engineered tumour microenvironment (TME) novel, antigen-presenting function of melanocytes and its
[262] are very helpful for studying the modus operandi possible relationship to hypopigmentary disorders
of cancer cell proliferation, migration and metastasis. J. Immunol. 151 7284–92
[16] Yamaguchi Y, Brenner M and Hearing V J 2007 The
Different melanoma cell types or numbers could be
regulation of skin pigmentation J. Biol. Chem. 282
printed in designated skin areas or layers and monitored 27557–61
for proliferation, motility, survival, and drug response [17] Borovansky J and Riley P A 2011 Melanins and Melanosomes:
[263]. Even before initiating the treatment process for a Biosynthesis, Structure, Physiological and Pathological
Functions (Weinheim: Wiley)
cancer patient, clinicians can bioprint cancer models to
[18] Plonka P M et al 2009 What are melanocytes really doing all
see how each patient’s actual tumour responds to the day long? Exp. Dermatol. 18 799–819
drug and is there a possibility of metastasis. Such [19] Lu Y, Zhu W Y, Tan C, Yu G H and Gu J X 2002 Melanocytes
sophisticated 3D skin cancer models can be engineered are potential immunocompetent cells: evidence from
recognition of immunological characteristics of cultured
with 3D bioprinting technology, for example, the recent
human melanocytes Pigm. Cell. Res. 15 454–60
‘metastasis-on-a-chip’ platform [264, 265]. From a [20] Moll I et al 2005 Human Merkel cells–aspects of cell biology,
policy standpoint, standardization and government distribution and functions Eur. J. Cell. Biol. 84 259–71
support for clinical implementation is the need of the [21] Boulais N and Misery L 2007 Merkel cells J. Am. Acad.
Dermatol. 57 147–65
hour. 3D bioprinting is thus far a promising technology
[22] Lucarz A and Brand G 2007 Current considerations about
but lacks coordinated efforts from multi-disciplinary Merkel cells Eur. J. Cell. Biol. 86 243–51
teams. A collaborative approach, including experts [23] Ogawa H 1996 The Merkel cell as a possible mechanoreceptor
from various domains including engineering, medicine, cell Prog. Neurobiol. 49 317–34
[24] Gottschaldt K M and Vahle-Hinz C 1981 Merkel cell
biomedical, and biochemistry will further strengthen its
receptors: structure and transducer function Science 214
prospects and bolster its potential to be available quickly 183–6
in serving the people and saving their lives. [25] Stingl G, Katz S I, Clement L, Green I and Shevach E M 1978
Immunologic functions of Ia-bearing epidermal Langerhans
cells J. Immunol. 121 2005–13
References [26] Kissenpfennig A et al 2005 Dynamics and function of
langerhans cells in vivo: dermal dendritic cells colonize
[1] Kanitakis J 2001 Anatomy, histology and lymph node areasdistinct from slower migrating langerhans
immunohistochemistry of normal human skin Eur. J. cells Immunity 22 643–54
Dermatol. 12 390–9 [27] Stingl G, Tamaki K and Katz S I 1980 Origin and function of
[2] Kolarsick P A, Kolarsick M A and Goodwin C 2011 Anatomy epidermal Langerhans cells Immunol. Rev. 53 149–74
and physiology of the skin J. Dermatol. Nurse. Assoc. 3 203–13 [28] Stingl G, Katz S I, Shevach E M, Rosenthal A S and Green I
[3] Pereira R F, Barrias C C, Granja P L and Bartolo P J 2013 1978 Analogous functions of macrophages and Langerhans
Advanced biofabrication strategies for skin regeneration and cells in the initiation of the immune response J. Invest.
repair Nanomedicine 8 603–21 Dermatol. 71 59–64

26
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

[29] Braathen L R and Thorsby E 1980 Studies on human [52] Ponec M 2002 Skin constructs for replacement of skin tissues
epidermal Langerhans cells Scand J. Immunol. 11 401–8 for in vitro testing Adv. Drug. Deliv. Rev. 54 S19–30
[30] Klechevsky E et al 2008 Functional specializations of human [53] Welss T, Basketter D A and Schroder K R 2004 In vitro skin
epidermal Langerhans cells and CD14+ dermal dendritic irritation: facts and future. State of the art review of
cells Immunity 29 497–510 mechanisms and models Toxicol. In. Vitro. 18 231–43
[31] Galli S J, Nakae S and Tsai M 2005 Mast cells in the [54] de Silva O 2002 The contributions of the European cosmetics
development of adaptive immune responses Nature industry to the development of alternatives to animal testing:
Immunol. 6 135–42 dialogue with ECVAM and future challenges Altern. Lab.
[32] Wedemeyer J, Tsai M and Galli S J 2000 Roles of mast cells Anim. 30 189–94
and basophils in innate and acquired immunity Curr. Opin. [55] Boelsma E, Gibbs S, Faller C and Ponec M 2000
Immunol. 12 624–31 Characterization and comparison of reconstructed skin
[33] Schwartz L B 1994 Mast cells: function and contents Curr. models: morphological and immunohistochemical
Opin. Immunol. 6 91–7 evaluation Acta Derm. Venereol. 80 82–8
[34] Benyon R C, Lowman M A and Church M K 1987 Human [56] Fentem J H et al 2001 A prevalidation study on in vitro tests
skin mast cells: their dispersion, purification, and secretory for acute skin irritation Results and evaluation by the
characterization J. Immunol. 138 861–7 Management Team Toxicol. In Vitro. 15 57–93
[35] Dawicki W and Marshall J S 2007 New and emerging roles for [57] Fentem J H and Botham P A 2002 ECVAM’s activities in
mast cells in host defence Curr. Opin. Immunol. 19 31–8 validating alternative tests for skin corrosion and irritation
[36] Weller K, Foitzik K, Paus R, Syska W and Maurer M 2006 Altern. Lab. Anim. 30 61–8
Mast cells are required for normal healing of skin wounds in [58] https://secure.peta.org/site/Advocacy?cmd=display&
mice FASEB J. 20 2366–8 page=UserAction&id=4965
[37] Metz M, Siebenhaar F and Maurer M 2008 Mast cell [59] https://congress.gov/bill/114th-congress/house-bill/2858
functions in the innate skin immune system Immunobiol. 213 [60] http://mediapeta.com/peta/PDF/companiesdotest.pdf
251–60 [61] http://peta.org/action/easy-ways-help-animals-used-
[38] Lucas T et al 2010 Differential roles of macrophages in diverse killed-experiments/
phases of skin repair J. Immunol. 184 3964–77 [62] Shores J T, Gabriel A and Gupta S 2007 Skin substitutes and
[39] Delavary B M, van der Veer W M, van Egmond M, alternatives: a review Adv. Skin Wound Care. 20 493–508
Niessen F B and Beelen R H 2011 Macrophages in skin injury [63] Halim A S, Khoo T L and Yussof S J M 2010 Biologic and
and repair Immunobiol. 216 753–62 synthetic skin substitutes: an overview Indian J. Plast. Surg.
[40] Malissen B, Tamoutounour S and Henri S 2014 The origins 43 S23
and functions of dendritic cells and macrophages in the skin [64] Balasubramani M, Kumar T R and Babu M 2001 Skin
Nature Rev. Immunol. 14 417–28 substitutes: a review Burns 27 534–44
[41] Brancato S K and Albina J E 2011 Wound macrophages as key [65] Kumar P 2008 Classification of skin substitutes Burns 34 148–9
regulators of repair: origin, phenotype, and function Am. J. [66] Supp D M and Boyce S T 2005 Engineered skin substitutes:
Pathol. 178 19–25 practices and potentials Clin. Dermatol. 23 403–12
[42] MedMarket Diligence, LLC Worldwide Wound [67] Auger F A, Lacroix D and Germain L 2009 Skin substitutes
Management, Forecast to 2024: Established and Emerging and wound healing Skin Pharmacol. Physiol. 22 94–102
Products, Technologies and Markets in the Americas, [68] Shores J T, Gabriel A and Gupta S 2007 Skin substitutes and
Europe, Asia/Pacific and Rest of World (http://mediligence. alternatives: a review Adv. Skin Wound Care 20 493–508
com/rpt/rpt-s251.htm) [69] Jones I, Currie L and Martin R 2002 A guide to biological skin
[43] Transparent Market Research Report 2015, Tissue substitutes Brit. J. Plast. Surg. 55 185–93
Engineered Skin Substitutes Market (by Type—Acellular, [70] Boyce S T and Warden G D 2002 Principles and practices for
Cellular Allogeneic, Cellular Autologous, and Others; by treatment of cutaneous wounds with cultured skin substitutes
Application—Burn Injury, Diabetic/Vascular Ulcer, and Am. J. Surg. 183 445–56
Others)—Global Industry Analysis, Size, Share, Growth, [71] Adzick N S and Lorenz H P 1994 Cells, matrix, growth
Trends, and Forecast 2015–2023 http:// factors, and the surgeon. The biology of scarless fetal wound
transparencymarketresearch.com/tissue-engineered-skin- repair Ann. Surg. 220 10
substitute.html [accessed 04th Jun 2016] [72] Bártolo P J, Domingos M, Patrício T, Cometa S and
[44] American Burn Association, Burn Incidence and Treatment Mironov V 2011 Biofabrication strategies for tissue
in the United States 2016 http://ameriburn.org/resources_ engineering Advances on Modeling in Tissue Engineering
factsheet.php [accessed 04th Jun 2016] (Amsterdam: Springer) pp 137–76
[45] Blais M, Parenteau-Bareil R, Cadau S and Berthod F 2013 [73] Bartolo P et al 2012 Biomedical production of implants by
Concise review: tissue-engineered skin and nerve additive electro-chemical and physical processes CIRP Ann.
regeneration in burn treatment Stem Cells Transl. Med. 2 Manuf. Technol. 61 635–55
545–51 [74] Liao H T, Chang K H, Jiang Y, Chen J P and Lee M Y 2012
[46] Mayet N et al 2014 A comprehensive review of advanced Fabrication of tissue engineered PCL scaffold by selective
biopolymeric wound healing systems J. Pharmaceut. Sci. 103 laser-sintered machine for osteogeneisis of adipose-derived
2211–30 stem cells: the research has proven that a bone tissue-
[47] Li J, Chen J and Kirsner R 2007 Pathophysiology of acute engineered scaffold can be made using the selective laser
wound healing Clinic. Dermatol. 25 9–18 sintering method Virt. Phys. Prototyp. 6 57–60
[48] Ferguson M W and O’Kane S 2004 Scar–free healing: from [75] Nichol J W and Khademhosseini A 2009 Modular tissue
embryonic mechanisms to adult therapeutic intervention engineering: engineering biological tissues from the bottom
Philos. T. Roy. Soc. B 359 839–50 up Soft Matter 5 1312–9
[49] Diegelmann R F and Evans M C 2004 Wound healing: an [76] Du Y, Lo E, Ali S and Khademhosseini A 2008 Directed
overview of acute, fibrotic and delayed healing Front Biosci. 9 assembly of cell-laden microgels for fabrication of 3D tissue
283–9 constructs P. Nat. Acad. Sci. 105 9522–7
[50] Velnar T, Bailey T and Smrkolj V 2009 The wound healing [77] Khademhosseini A, Langer R, Borenstein J and Vacanti J P
process: an overview of the cellular and molecular 2006 Microscale technologies for tissue engineering and
mechanisms J. Int. Med. Res. 37 1528–42 biology Proc. Natl Acad. Sci. USA 103 2480–7
[51] Boelsma E, Verhoeven M C and Ponec M 1999 [78] Urciuolo F, Imparato G, Guaccio A, Mele B and Netti P A
Reconstruction of a human skin equivalent using a 2012 Novel strategies to engineering biological tissue in vitro
spontaneously transformed keratinocyte cell line (HaCaT) Nanotechnology in Regenerative Medicine (New York:
J. Invest. Dermatol. 112 489–98 Humana)

27
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

[79] Yang J, Yamato M, Shimizu T, Sekine H, Ohashi K, [101] Tan K H et al 2003 Scaffold development using selective laser
Kanzaki M, Ohki T, Nishida K and Okano T 2007 sintering of polyetheretherketone–hydroxyapatite
Reconstruction of functional tissues with cell sheet biocomposite blends Biomaterials 24 3115–23
engineering Biomaterials 28 5033–43 [102] Williams J M et al 2005 Bone tissue engineering using
[80] Nahmias Y, Schwartz R E, Verfaillie C and Odde D J 2005 polycaprolactone scaffolds fabricated via selective laser
Laser-guided direct writing for three-dimensional tissue sintering Biomaterials 26 4817–27
engineering Biotechnol. Bioeng. 92 129–36 [103] Dhariwala B, Hunt E and Boland T 2004 Rapid prototyping
[81] Raof N A, Schiele N R, Xie Y, Chrisey D B and Corr D T 2011 of tissue-engineering constructs, using photopolymerizable
The maintenance of pluripotency following laser direct-write hydrogels and stereolithography Tissue Eng. 10 1316–22
of mouse embryonic stem cells Biomaterials 32 1802–8 [104] Seck T M, Melchels F P, Feijen J and Grijpma D W 2010
[82] Lin Y, Huang G, Huang Y, Jeremy Tzeng T R and Chrisey D Designed biodegradable hydrogel structures prepared by
2010 Effect of laser fluence in laser-assisted direct writing of stereolithography using poly (ethylene glycol)/poly (d,
human colon cancer cell Rapid Prototyp. J. 16 202–8 l-lactide)-based resins J. Control. Release 148 34–41
[83] Catros S, Guillotin B, Bačáková M, Fricain J C and [105] Engler A J, Sen S, Sweeney H L and Discher D E 2006 Matrix
Guillemot F 2011 Effect of laser energy, substrate film elasticity directs stem cell lineage specification Cell 126
thickness and bioink viscosity on viability of endothelial cells 677–89
printed by laser-assisted bioprinting Appl. Surf. Sci. 257 [106] Yim E K and Sheetz M P 2012 Force-dependent cell signaling
5142–7 in stem cell differentiation Stem Cell Res. Ther. 3 41
[84] Saunders R E, Gough J E and Derby B 2008 Delivery of human [107] Iskratsch T, Wolfenson H and Sheetz M P 2014 Appreciating
fibroblast cells by piezoelectric drop-on-demand inkjet force and shape [mdash] the rise of mechanotransduction in
printing Biomaterials 29 193–203 cell biology Nature Rev. Mol. Cell Biol. 15 825–33
[85] Tirella A et al 2011 Substrate stiffness influences high [108] Ghassemi S et al 2012 Cells test substrate rigidity by local
resolution printing of living cells with an ink-jet system contractions on submicrometer pillars Proc. Natl Acad. Sci.
J. Biosci. Bioeng. 112 79–85 USA 109 5328–33
[86] Khalil S, Nam J and Sun W 2005 Multi-nozzle deposition for [109] Tiruvannamalai-Annamalai R, Armant D R and
construction of 3D biopolymer tissue scaffolds Rapid Matthew H W 2014 A glycosaminoglycan based, modular
Prototyp. J. 11 9–17 tissue scaffold system for rapid assembly of perfusable, high
[87] Sepulveda P, Binner J G P, Rogero S O, Higa O Z and cell density, engineered tissues PLoS one 9 e84287
Bressiani J C 2000 Production of porous hydroxyapatite by [110] Ramalingam M, Jabbari E and Ramakrishna S 2013
the gel-casting of foams and cytotoxic evaluation J. Biomed. Integrating top-down and bottom-up scaffolding tissue
Mat. Res. 50 27–34 engineering approach for bone regeneration (New Jesery, NY:
[88] Chu T M, Halloran J W, Hollister S J and Feinberg S E 2001 Wiley)
Hydroxyapatite implants with designed internal architecture [111] Ouyang H W, Toh S L, Goh J, Tay T E and Moe K 2005
J. Mat. Sci.: Mat. Med. 12 471–8 Assembly of bone marrow stromal cell sheets with knitted
[89] Zhang Y and Zhang M 2002 Three‐dimensional poly (L‐lactide) scaffold for engineering ligament analogs
macroporous calcium phosphate bioceramics with nested J. Biomed. Mat. Res. B: Appl. Biomat. 75 264–71
chitosan sponges for load‐bearing bone implants J. Biomed. [112] Sargeant T D et al 2008 Hybrid bone implants: self-assembly
Mat. Res. 61 1–8 of peptide amphiphile nanofibers within porous titanium
[90] Ni S, Chang J and Chou L 2006 A novel bioactive porous Biomaterials 29 161–71
CaSiO3 scaffold for bone tissue engineering J. Biomed. Mat. [113] http://ir.organovo.com/news/press-releases/press-
Res. A 76 196–205 releases-details/2015/LOreal-USA-Announces-Research-
[91] Agrawal C M, Parr J E and Lin S T 2000 Synthetic Partnership-with-Organovo-to-Develop-3D-Bioprinted-
Bioabsorbable Polymers for Implants vol 1396 (West Skin-Tissue/
Conshohocken, PA: ASTM International) [114] http://ft.com/intl/cms/s/0/02809474-ffe8-11e4-abd5-
[92] Hua F J, Kim G E, Lee J D, Son Y K and Lee D S 2002 00144feabdc0.html#axzz3mMp5pa1N
Macroporous poly (L‐lactide) scaffold 1. Preparation of a [115] http://3dprint.com/83334/rokit-bioprinting/
macroporous scaffold by liquid–liquid phase separation of a [116] Murphy S V and Atala A 2014 3D bioprinting of tissues and
PLLA–dioxane–water system J. Biomed. Mat. Res. 63 161–7 organs Nature Biotechnol. 32 773–85
[93] Lannutti J, Reneker D, Ma T, Tomasko D and Farson D 2007 [117] Ozkul T and Ozkul M H 2004 Computer simulation tool for
Electrospinning for tissue engineering scaffolds Mat. Sci. Eng. rhinoplasty planning Comput. Biol. Med. 34 697–718
C 27 504–9 [118] Rabi S A and Aarabi P 2006 Face Fusion: an automatic
[94] Agarwal S, Wendorff J H and Greiner A 2009 Progress in the method for virtual plastic surgery InInformation Fusion
field of electrospinning for tissue engineering applications 1 1–7
Adv. Mat. 21 3343–51 [119] Kim H, Jürgens P, Weber S, Nolte L P and Reyes M 2010 A
[95] Li W J, Laurencin C T, Caterson E J, Tuan R S and Ko F K new soft-tissue simulation strategy for cranio-maxillofacial
2002 Electrospun nanofibrous structure: a novel scaffold for surgery using facial muscle template model Prog. Biophys.
tissue engineering J. Biomed. Mat. Res. 60 613–21 Mol. Biol. 103 284–91
[96] Lam C X F, Mo X M, Teoh S H and Hutmacher D W 2002 [120] Koch R M et al 1996 Simulating facial surgery using
Scaffold development using 3D printing with a starch-based finite element models Proc. of the 23rd Annual Conf. on
polymer Mat. Sci. Eng. C 20 49–56 Computer Graphics and Interactive Techniques (ACM) 1,
[97] Bergmann C et al 2010 3D printing of bone substitute 421–428
implants using calcium phosphate and bioactive glasses [121] Heike C L, Upson K, Stuhaug E and Weinberg S M 2010 3D
J. Eur. Ceram. Soc. 30 2563–7 digital stereophotogrammetry: a practical guide to facial
[98] Fierz F C et al 2008 The morphology of anisotropic 3D- image acquisition Head Face Med. 6 18
printed hydroxyapatite scaffolds Biomaterials 29 3799–806 [122] Lin S J, Patel N, O’Shaughnessy K and Fine N 2008 A new
[99] Eshraghi S and Das S 2010 Mechanical and microstructural three-dimensional imaging device in facial aesthetic and
properties of polycaprolactone scaffolds with one- reconstructive surgery Otolaryngology–Head and Neck Surg.
dimensional, two-dimensional, and three-dimensional 139 313–5
orthogonally oriented porous architectures produced by [123] See M S, Foxton M R, Miedzianowski-Sinclair N A,
selective laser sintering Acta biomater. 6 2467–76 Roberts C E and Nduka C 2007 Stereophotogrammetric
[100] Yeong W Y et al 2010 Porous polycaprolactone scaffold for measurement of the nasolabial fold in repose: a study
cardiac tissue engineering fabricated by selective laser of age and posture-related changes Eur. J. Plast. Surg. 29
sintering Acta Biomater. 6 2028–34 387–93

28
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

[124] Zhang R, Tsai P S, Cryer J E and Shah M 1999 Shape-from- [147] Norouzi M, Boroujeni S M, Omidvarkordshouli N and
shading: a survey Pattern Analysis and Machine Intelligence, Soleimani M 2015 Advances in skin regeneration: application
IEEE Transactions 21 690–706 of electrospun scaffolds Adv. Healthcare Mat. 4 1114–33
[125] Jebara T, Azarbayejani A and Pentland A 1999 3D structure [148] Powell H M, Supp D M and Boyce S T 2008 Influence of
from 2D motion Signal Processing Magazine IEEE 16 66–84 electrospun collagen on wound contraction of engineered
[126] Torresani L, Hertzmann A and Bregler C 2008 Nonrigid skin substitutes Biomaterials 29 834–43
structure-from-motion: estimating shape and motion with [149] Zhong S et al 2006 An aligned nanofibrous collagen scaffold
hierarchical priors Pattern Analysis and Machine Intelligence, by electrospinning and its effects on in vitro fibroblast culture
IEEE Transactions 30 878–92 J. Biomed. Mat. Res. A 79 456–63
[127] Moghaddam B, Lee J, Pfister H and Machiraju R 2003 Model- [150] Powell H M and Boyce S T 2008 Fiber density of electrospun
based 3D face capture with shape-from-silhouettes InAnalysis gelatin scaffolds regulates morphogenesis of dermal–
and Modeling of Faces and Gestures, 2003. AMFG 2003. IEEE epidermal skin substitutes J. Biomed. Mat. Res. A 84 1078–86
International Workshop 1 20–7 [151] Ko J H et al 2010 Characterization of cross-linked gelatin
[128] Salzmann M, Pilet J, Ilic S and Fua P 2007 Surface nanofibers through electrospinning Macromol. Res. 18
deformation models for nonrigid 3D shape recovery Pattern 137–43
Analysis and Machine Intelligence, IEEE Transactions 29 [152] Min B M et al 2004 Electrospinning of silk fibroin
1481–7 nanofibers and its effect on the adhesion and spreading of
[129] Blanz V and Vetter T 1999 A morphable model for the normal human keratinocytes and fibroblasts in vitro
synthesis of 3D faces Proc. of the 26th Annual Conf. on Biomaterials 25 1289–97
Computer Graphics and Interactive Techniques (New [153] Min B M et al 2004 Formation of silk fibroin matrices with
York: ACM) different texture and its cellular response to normal human
[130] Oliveira-Santos T et al 2013 3D face reconstruction from 2d keratinocytes Int. J. Biol. Macromol. 34 223–30
pictures: first results of a web-based computer aided system [154] Jeong L et al 2009 Plasma-treated silk fibroin nanofibers for
for aesthetic procedures Ann. Biomed. Eng. 41 952–66 skin regeneration Int. J. Biol. Macromol. 44 222–8
[131] Hani A F M, Eltegani N M, Hussein S H, Jamil A and Gill P [155] Kundu B, Rajkhowa R, Kundu S C and Wang X 2013 Silk
2009 Assessment of ulcer wounds size using 3D skin surface fibroin biomaterials for tissue regenerations Adv. Drug Del.
imaging In Visual Informatics: Bridging Research and Practice Rev. 65 457–70
vol 1 (Berlin: Springer) pp 243–53 [156] Gu B K et al 2013 Fabrication of sonicated chitosan nanofiber
[132] Zhu Y K, Tian G Y, Lu R S and Zhang H 2011 A review of mat with enlarged porosity for use as hemostatic materials
optical NDT technologies Sensors 11 7773–98 Carbohydrate Polymers 97 65–73
[133] Lee S L and Lu Y H 2014 Modeling of bioheat equation for [157] Wnek G E, Carr M E, Simpson D G and Bowlin G L 2003
skin and a preliminary study on a noninvasive diagnostic Electrospinning of nanofiber fibrinogen structures Nano Lett.
method for skin burn wounds Burns 40 930–9 3 213–6
[134] Liu W M et al 2012 Reconstruction of thermographic signals [158] McManus M C et al 2006 Mechanical properties of
to map perforator vessels in humans Quant. Infrared electrospun fibrinogen structures Acta Biomater. 2 19–28
Thermography J. 9 123–33 [159] Sell S A et al 2008 Cross-linking methods of electrospun
[135] Prindeze N J et al 2014 Examination of the early diagnostic fibrinogen scaffolds for tissue engineering applications
applicability of active dynamic thermography for burn Biomed. Mat. 3 045001
wound depth assessment and concept analysis J. Burn Care [160] Yeo I S et al 2008 Collagen-based biomimetic nanofibrous
Res. 36 626–35 scaffolds: preparation and characterization of collagen/silk
[136] Renkielska A et al 2014 Active dynamic infrared thermal fibroin bicomponent nanofibrous structures
imaging in burn depth evaluation J. Burn Care Res. 35 Biomacromolecules 9 1106–16
e294–303 [161] Cai Z X et al 2010 Fabrication of chitosan/silk fibroin
[137] Renkielska A, Nowakowski A, Kaczmarek M and Ruminski J composite nanofibers for wound-dressing applications Int. J.
2006 Burn depths evaluation based on active dynamic IR Mol. Sci. 11 3529–39
thermal imaging—a preliminary study Burns 32 867–75 [162] Sarkar S D, Farrugia B L, Dargaville T R and Dhara S 2013
[138] Rumiński J, Kaczmarek M, Renkielska A and Nowakowski A Chitosan–collagen scaffolds with nano/microfibrous
2007 Thermal parametric imaging in the evaluation of skin architecture for skin tissue engineering J. Biomed. Mat. Res. A
burn depth Biomed. Eng. IEEE Transactions 54 303–12 101 3482–92
[139] Prindeze N J et al 2015 Evaluation of the variable depth [163] Naseri N et al 2014 Electrospun chitosan-based
resolution of active dynamic thermography on human skin In nanocomposite mats reinforced with chitin nanocrystals for
SPIE Biophotonics South America International Society for wound dressing Carbohydrate Polymers 109 7–15
Optics and Photonics 1 95310P [164] Augustine R et al 2014 Electrospun poly (ε‐caprolactone)‐
[140] Xiao K, Zardawi F, van Noort R and Yates J M 2013 Color based skin substitutes: in vivo evaluation of wound healing
reproduction for advanced manufacture of soft tissue and the mechanism of cell proliferation J. Biomed. Mat. Res.
prostheses J. Dentistry 41 e15–23 B: Appl. Biomat. 103 1445–54
[141] Technical C I E Report, Colorimetry, 3rd ed. Publication 15. [165] Kim J H, Jang J, Jeong Y H, Ko T J and Cho D W 2014
2004 Vienna: CIE Central Bureau Fabrication of a nanofibrous mat with a human skin pattern
[142] Xiao K, Zardawi F, van Noort R and Yates J M 2014 Langmuir 31 424–31
Developing a 3D colour image reproduction system for [166] Kurpinski K T, Stephenson J T, Janairo R R R, Lee H and Li S
additive manufacturing of facial prostheses Int. J. Adv. Manuf. 2010 The effect of fiber alignment and heparin coating on cell
Technol. 70 2043–9 infiltration into nanofibrous PLLA scaffolds Biomaterials 31
[143] Jang I S, Kim J W, You J Y and Kim J S 2013 Spectrum-based 3536–42
color reproduction algorithm for makeup simulation of 3D [167] Kluger P J et al 2010 Electrospun poly (D/L-lactide-co-L-
facial avatar ETRI J. 35 969–79 lactide) hybrid matrix: a novel scaffold material for soft tissue
[144] Wang Y, Hao X, Hou Y and Guo C 2013 A new multispectral engineering J. Mat. Sci.: Mat. Med. 21 2665–71
method for face liveness detection In Pattern Recognition [168] Kumbar S G, Nukavarapu S P, James R, Nair L S and
(ACPR), 2013 2nd IAPR Asian Conference on 1 922–6 IEEE Laurencin C T 2008 Electrospun poly (lactic acid-co-glycolic
[145] Agrawal P, Soni S, Mittal G and Bhatnagar A 2014 Role of acid) scaffolds for skin tissue engineering Biomaterials 29
polymeric biomaterials as wound healing agents Int. J. Lower 4100–7
Extrem. Wounds 1 1534734614544523 [169] Blackwood K A et al 2008 Development of biodegradable
[146] Gunatillake P A and Adhikari R 2003 Biodegradable synthetic electrospun scaffolds for dermal replacement Biomaterials 29
polymers for tissue engineering Eur. Cell Mater. 5 1–16 3091–104

29
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

[170] Kim S J, Jang D H, Park W H and Min B M 2010 Fabrication [190] Wu L et al 2010 Composite fibrous membranes of PLGA and
and characterization of 3-dimensional PLGA nanofiber/ chitosan prepared by coelectrospinning and coaxial
microfiber composite scaffolds Polymer 51 1320–7 electrospinning J. Biomed. Mat. Res. A 92 563–74
[171] Zhu X, Cui W, Li X and Jin Y 2008 Electrospun fibrous mats [191] Yang Y, Zhu X, Cui W, Li X and Jin Y 2009 Electrospun
with high porosity as potential scaffolds for skin tissue composite mats of poly [(D, L‐lactide)‐co‐glycolide] and
engineering Biomacromolecules 9 1795–801 collagen with high porosity as potential scaffolds for skin
[172] Shalumon K T et al 2011 Fabrication of chitosan/poly tissue engineering Macromol. Mat. Eng. 294 611–9
(caprolactone) nanofibrous scaffold for bone and skin tissue [192] Liu S J et al 2010 Electrospun PLGA/collagen nanofibrous
engineering Int. J. Biol. Macromol. 48 571–6 membrane as early-stage wound dressing J. Membrane Sci.
[173] Chen H, Huang J, Yu J, Liu S and Gu P 2011 Electrospun 355 53–9
chitosan-graft-poly (ε-caprolactone)/poly (ε-caprolactone) [193] Franco R A, Nguyen T H and Lee B T 2011 Preparation and
cationic nanofibrous mats as potential scaffolds for skin tissue characterization of electrospun PCL/PLGA membranes and
engineering Int. J. Biol. Macromol. 48 13–9 chitosan/gelatin hydrogels for skin bioengineering
[174] Croisier F, Atanasova G, Poumay Y and Jérôme C 2014 applications J. Mat. Sci.: Mat. Med. 22 2207–18
Polysaccharide‐coated PCL nanofibers for wound dressing [194] Malda J et al 2013 25 th anniversary article: engineering
applications Adv. Healthcare Mat. 3 2032–9 hydrogels for biofabrication Adv. Mat. 25 5011–28
[175] Huang C et al 2012 The involvement of integrin β1 signaling [195] Nakamura M, Iwanaga S, Henmi C, Arai K and Nishiyama Y
in the migration and myofibroblastic differentiation of skin 2010 Biomatrices and biomaterials for future developments
fibroblasts on anisotropic collagen-containing nanofibers of bioprinting and biofabrication Biofabrication 2 014110
Biomaterials 33 1791–800 [196] Murphy S V, Skardal A and Atala A 2013 Evaluation of
[176] Krithica N, Natarajan V, Madhan B, Sehgal P K and hydrogels for bio‐printing applications J. Biomed. Mat. Res. A
Mandal A B 2012 Type I collagen immobilized poly 101 272–84
(caprolactone) nanofibers: characterization of surface [197] Wang S, Lee J M and Yeong W Y 2015 Smart hydrogels for 3D
modification and growth of fibroblasts Adv. Eng. Mat. 14 bioprinting Int. J. Bioprint. 1 3–14
B149–54 [198] Bianco P and Robey P G 2001 Stem cells in tissue engineering
[177] Venugopal J and Ramakrishna S 2005 Biocompatible Nature 414 118–21
nanofiber matrices for the engineering of a dermal substitute [199] Rheinwald J G and Green H 2003 Serial cultivation of strains
for skin regeneration Tissue Eng. 11 847–54 of human epidermal keratinocytes: the formation of
[178] Zhang Y Z, Venugopal J, Huang Z M, Lim C T and keratinizing colonies from single cells Cell 6 331–4
Ramakrishna S 2005 Characterization of the surface [200] Rheinwald J G and Green H 1977 Epidermal growth factor
biocompatibility of the electrospun PCL-collagen nanofibers and the multiplication of cultured human epidermal
using fibroblasts Biomacromolecules 6 2583–9 keratinocytes Nature 265 421–4
[179] Powell H M and Boyce S T 2009 Engineered human skin [201] Alonso L and Fuchs E 2003 Stem cells in the skin: waste not,
fabricated using electrospun collagen–PCL blends: Wnt not Genes & Dev. 17 1189–200
morphogenesis and mechanical properties Tissue Eng. A 15 [202] Lootens L, Brusselaers N, Beele H and Monstrey S 2013
2177–87 Keratinocytes in the treatment of severe burn injury: an
[180] Fu X et al 2014 Regulation of migratory activity of human update Int. Wound J. 10 6–12
keratinocytes by topography of multiscale collagen- [203] Nolte S V, Xu W, Rennekampff H O and Rodemann H P 2008
containing nanofibrous matrices Biomaterials 35 1496–506 Diversity of fibroblasts–a review on implications for skin
[181] Blackstone B N, Drexler J W and Powell H M 2014 Tunable tissue engineering Cells Tissues Organs 187 165–76
engineered skin mechanics via coaxial electrospun fiber core [204] Fang C H, Robb E C, Yu G S, Alexander J W and Warden G D
diameter Tissue Eng. A 20 2746–55 1990 Observations on stability and contraction of composite
[182] Feng B et al 2015 Effect of inhomogeneity of the electrospun skin grafts: xenodermis or allodermis with an isograft overlay
fibrous scaffolds of gelatin/polycaprolactone hybrid on cell J. Burn Care Res. 11 538–42
proliferation J. Biomed. Mat. Res. A 103 431–8 [205] Lineen E and Namias N 2008 Biologic dressing in burns
[183] Gautam S, Chou C F, Dinda A K, Potdar P D and Mishra N C J. Craniofacial Surg. 19 923–8
2014 Surface modification of nanofibrous polycaprolactone/ [206] Middelkoop E et al 2004 Porcine wound models for skin
gelatin composite scaffold by collagen type I grafting for skin substitution and burn treatment Biomaterials 25 1559–67
tissue engineering Mat. Sci. Eng. C 34 402–9 [207] Harrison C A and MacNeil S 2008 The mechanism of skin
[184] Li Y, Chen F, Nie J and Yang D 2012 Electrospun poly (lactic graft contraction: an update on current research and potential
acid)/chitosan core–shell structure nanofibers from future therapies Burns 34 153–63
homogeneous solution Carbohydrate Polymers 90 1445–51 [208] Boyce S T et al 1993 Pigmentation and inhibition of wound
[185] Shalumon K T et al 2012 Fabrication of aligned poly (lactic contraction by cultured skin substitutes with adult
acid)-chitosan nanofibers by novel parallel blade collector melanocytes after transplantation to athymic mice J. Invest.
method for skin tissue engineering J. Biomed. Nanotechnol. 8 Dermatol. 100 360–5
405–16 [209] Duncan C O et al 2005 In vitro transfer of keratinocytes:
[186] Hoveizi E, Nabiuni M, Parivar K, Rajabi‐Zeleti S and comparison of transfer from fibrin membrane and delivery by
Tavakol S 2014 Functionalisation and surface modification of aerosol spray J. Biomed. Mat. Res. Part B: Appl. Biomat. 73
electrospun polylactic acid scaffold for tissue engineering Cell 221–8
Biol. Int. 38 41–9 [210] Metcalfe A D and Ferguson M W 2008 Skin stem and
[187] Chen S H, Chang Y, Lee K R and Lai J Y 2014 A three- progenitor cells: using regeneration as a tissue-engineering
dimensional dual-layer nano/microfibrous structure of strategy Cell. Mol. Life Sci.: CMLS 65 24–32
electrospun chitosan/poly (d, l-lactide) membrane for the [211] Fernandes K J et al 2004 A dermal niche for multipotent adult
improvement of cytocompatibility J. Membrane Sci. 450 skin-derived precursor cells Nature Cell Biol. 6 1082–93
224–34 [212] Mahjour S B, Ghaffarpasand F and Wang H 2011 Hair follicle
[188] Jin G, Prabhakaran M P and Ramakrishna S 2011 Stem cell regeneration in skin grafts: current concepts and future
differentiation to epidermal lineages on electrospun perspectives Tissue Eng. Part B: Rev. 18 15–23
nanofibrous substrates for skin tissue engineering Acta [213] Mistriotis P and Andreadis S T 2013 Hair Follicle: a novel
Biomater. 7 3113–22 source of multipotent stem cells for tissue engineering and
[189] Chandrasekaran A R, Venugopal J, Sundarrajan S and regenerative medicine Tissue Eng. Part B: Rev. 19 265–78
Ramakrishna S 2011 Fabrication of a nanofibrous scaffold [214] Zhang C P and Fu X B 2008 Therapeutic potential of stem
with improved bioactivity for culture of human dermal cells in skin repair and regeneration Chinese J. Traumatol. 11
fibroblasts for skin regeneration Biomed. Mat. 6 015001 209–21

30
Biofabrication 8 (2016) 032001 S Vijayavenkataraman et al

[215] Charruyer A and Ghadially R 2009 Stem cells and tissue- [241] Arslan-Yildiz A et al 2016 Towards artificial tissue models: past,
engineered skin Skin Pharmacol. Physiol. 22 55–62 present, and future of 3D bioprinting Biofabrication 8 014103
[216] Ferrari G et al 1998 Muscle regeneration by bone marrow- [242] Cukierman E, Pankov R, Stevens D R and Yamada K M 2001
derived myogenic progenitors Science 279 1528–30 Taking cell-matrix adhesions to the third dimension Science
[217] Gussoni E et al 1999 Dystrophin expression in the mdx mouse 294 1708–12
restored by stem cell transplantation Nature 401 390–4 [243] Cukierman E, Pankov R and Yamada K M 2002 Cell
[218] Peterson D A 2002 Stem cells in brain plasticity and repair interactions with three-dimensional matrices Curr. Opin. Cell
Curr. Opin. Pharmacol. 2 34–42 Biol. 14 633–40
[219] Dababneh A B and Ozbolat I T 2014 Bioprinting technology: a [244] Edelman D B and Keefer E W 2005 A cultural renaissance:
current state-of-the-art review J. Manuf. Sci. Eng. 136 061016 in vitro cell biology embraces three-dimensional context Exp.
[220] Barron J A, Wu P, Ladouceur H D and Ringeisen B R 2004 Neurol. 192 1–6
Biological laser printing: a novel technique for creating [245] Gruene M, Unger C, Koch L, Deiwick A and Chichkov B 2011
heterogeneous 3-dimensional cell patterns Biomed. Dispensing pico to nanolitre of a natural hydrogel by laser-
Microdevice 6 139–47 assisted bioprinting Biomed. Eng. Online 10 19
[221] Guillemot F et al 2010 High-throughput laser printing of cells and [246] Ringeisen B R, Othon C M, Barron J A, Young D and
biomaterials for tissue engineering Acta Biomater. 6 2494–500 Spargo B J 2006 Jet‐based methods to print living cells
[222] Wang W, Huang Y, Grujicic M and Chrisey D B 2008 Study of Biotechnol. J. 1 930–48
impact-induced mechanical effects in cell direct writing using [247] Schiele N R et al 2010 Laser-based direct-write techniques for
smooth particle hydrodynamic method J. Manuf. Sci. Eng. cell printing Biofabrication 2 032001
130 021012 [248] Koch L et al 2012 Skin tissue generation by laser cell printing
[223] Barron J A, Ringeisen B R, Kim H, Spargo B J and Chrisey D B Biotechnol. Bioeng. 109 1855–63
2004 Application of laser printing to mammalian cells Thin [249] Michael S et al 2013 Tissue engineered skin substitutes
Solid Films 453 383–7 created by laser-assisted bioprinting form skin-like structures
[224] Guillotin B et al 2010 Laser assisted bioprinting of engineered in the dorsal skin fold chamber in mice PloS one 8 e57741
tissue with high cell density and microscale organization [250] Koch L et al 2009 Laser printing of skin cells and human stem
Biomaterials 31 7250–6 cells Tissue Eng. Part C: Methods 16 847–54
[225] Gaebel R et al 2011 Patterning human stem cells and [251] Lee W et al 2009 Multi-layered culture of human skin
endothelial cells with laser printing for cardiac regeneration fibroblasts and keratinocytes through three-dimensional
Biomaterials 32 9218–30 freeform fabrication Biomaterials 30 1587–95
[226] Nakamura M et al 2005 Biocompatible inkjet printing [252] Lee V et al 2013 Design and fabrication of human skin by three-
technique for designed seeding of individual living cells Tissue dimensional bioprinting Tissue Eng. Part C: Methods 20 473–84
Eng. 11 1658–66 [253] Kim G, Ahn S, Yoon H, Kim Y and Chun W 2009 A cryogenic
[227] Xu T et al 2006 Viability and electrophysiology of neural cell direct-plotting system for fabrication of 3D collagen scaffolds
structures generated by the inkjet printing method for tissue engineering J. Mat. Chem. 19 8817–23
Biomaterials 27 3580–8 [254] Kim G, Ahn S, Kim Y, Cho Y and Chun W 2011 Coaxial
[228] Geckil H, Xu F, Zhang X, moon S and Demirci U 2010 structured collagen–alginate scaffolds: fabrication, physical
Engineering hydrogels as extracellular matrix mimics properties, and biomedical application for skin tissue
Nanomedicine 5 469–84 regeneration J. Mat. Chem. 21 6165–72
[229] Campbell P G and Weiss L E 2007 Tissue engineering with the [255] Leng L, Amini-Nik S, Ba Q, Jeschke M and Günther A 2013
aid of inkjet printers Expert Opin. Biol. Therapy 7 1123–7 Skin printer: Microfluidic approach for skin regeneration and
[230] Xu T, Jin J, Gregory C, Hickman J J and Boland T 2005 Inkjet wound dressings 17th International Conference on
printing of viable mammalian cells Biomaterials 26 93–9 Miniaturized Systems for Chemistry and Life Sciences,
[231] Yamazoe H and Tanabe T 2009 Cell micropatterning on an MicroTAS 2013 3 1833–5
albumin‐based substrate using an inkjet printing technique [256] Leng L, McAllister A, Zhang B, Radisic M and Günther A
J. Biomed. Mat. Res. A 91 1202–9 2012 Mosaic hydrogels: one‐step formation of multiscale soft
[232] Norotte C, Marga F S, Niklason L E and Forgacs G 2009 materials Adv. Mat. 24 3650–8
Scaffold-free vascular tissue engineering using bioprinting [257] Binder K W 2011 In situ bioprinting of the Skin (Doctoral
Biomaterials 30 5910–7 dissertation). Retrieved from Wake Forest University’s
[233] Smith C M et al 2004 Three-dimensional bioassembly tool for institutional repository (https://wakespace.lib.wfu.edu)
generating viable tissue-engineered constructs Tissue Eng. 10 [258] Sofokleous P, Stride E, Bonfield W and Edirisinghe M 2013
1566–76 Design, construction and performance of a portable
[234] Skardal A et al 2010 Photocrosslinkable hyaluronan-gelatin handheld electrohydrodynamic multi-needle spray gun for
hydrogels for two-step bioprinting Tissue Eng. A 16 2675–85 biomedical applications Mat. Sci. Eng. C 33 213–23
[235] Skardal A, Zhang J and Prestwich G D 2010 Bioprinting [259] Amini‐Nik S et al 2011 Pax7 expressing cells contribute to
vessel-like constructs using hyaluronan hydrogels crosslinked dermal wound repair, regulating scar size through a β‐catenin
with tetrahedral polyethylene glycol tetracrylates Biomaterials mediated process Stem Cells 29 1371–9
31 6173–81 [260] Horch R E, Jeschke M G, Spilker G, Herndon D N and Kopp J
[236] Lee Y B et al 2010 Bio-printing of collagen and VEGF- 2005 Treatment of second degree facial burns with allografts
releasing fibrin gel scaffolds for neural stem cell culture Exp. —preliminary results Burns 31 597–602
Neurol. 223 645–52 [261] Dupin E, Calloni G, Real C, Gonçalves-Trentin A and
[237] moon S et al 2009 Layer by layer three-dimensional tissue Le Douarin N M 2007 Neural crest progenitors and stem cells
epitaxy by cell-laden hydrogel droplets Tissue Eng. Part C: C R Biol. 330 521–9
Methods 16 157–66 [262] Asghar W et al 2015 Engineering cancer microenvironments
[238] Jakab K et al 2008 Tissue engineering by self-assembly of cells for in vitro 3D tumor models Mater. Today. 18 539–53
printed into topologically defined structures Tissue Eng. A 14 [263] Vultur A, Schanstra T and Herlyn M 2016 The promise of 3D
413–21 skin and melanoma cell bioprinting Melanoma. Res. 26 205–6
[239] Smith C M, Christian J J, Warren W L and Williams S K 2007 [264] Skardal A, Devarasetty M, Forsythe S, Atala A and Soker S
Characterizing environmental factors that impact the 2016 A reductionist metastasis‐on‐a‐chip platform for in vitro
viability of tissue-engineered constructs fabricated by a tumor progression modeling and drug screening Biotechnol.
direct-write bioassembly tool Tissue Eng. 13 373–83 Bioeng. 113 2020–32
[240] Ozbolat I T and Yu Y 2013 Bioprinting toward organ [265] Lee E, Song H G and Chen C S 2016 Biomimetic on-a-chip
fabrication: challenges and future trends Biomedical platforms for studying cancer metastasis Curr. Opin Chem.
Engineering, IEEE Transactions 60 691–9 Eng. 11 20–7

31

You might also like