You are on page 1of 11

PROGRESS REPORT

3D Skin Models www.advhealthmat.de

Toward Immunocompetent 3D Skin Models


Aleta Pupovac, Berna Senturk, Chiara Griffoni, Katharina Maniura-Weber,
Markus Rottmar,* and Sally L. McArthur*

models remains challenging due to its


3D human skin models provide a platform for toxicity testing, biomaterials structural and functional complexity as it
evaluation, and investigation of fundamental biological processes. However, encompasses a variety of specialized cells
the majority of current in vitro models lack an inflammatory system, and physiological subsystems, including
those responsible for controlling immune
vasculature, and other characteristics of native skin, indicating scope for responses.[2]
more physiologically complex models. Looking at the immune system, there Different animal models have been
are a variety of cells that could be integrated to create novel skin models, but used for skin research, but mouse models
to do this effectively it is also necessary to understand the interface between are most widely used because of easy han-
skin biology and tissue engineering as well as the different roles the immune dling and lower cost. Mouse models are
regularly employed to study skin physi-
system plays in specific health and disease states. Here, a progress report
ology, biochemistry, and immunology,
on skin immunity and current immunocompetent skin models with a focus and are often mandatory for in vivo trans-
on construction methods is presented; scaffold and cell choice as well as the lational research. While many facets of
requirements of physiologically relevant models are elaborated. The wide anatomical, physiological, and immuno-
range of technological and fundamental challenges that need to be addressed logical features are similar between mice
to successfully generate immunocompetent skin models and the steps and humans, many comparative studies
show large differences between mouse
currently being made globally by researchers as they develop new models are and human skin.[3] Human skin is struc-
explored. Induced pluripotent stem cells, microfluidic platforms to control turally and functionally different to mouse
the model environment, and new real-time monitoring techniques capable of skin; it is thicker and contains fewer hair
probing biochemical processes within the models are discussed. follicles. Further, the mouse epidermis
is composed of less keratinocyte layers,
which may contribute to decreased barrier
1. Introduction function and greater absorption observed in mouse skin com-
pared to human skin.[3] The skin immune system is composed
The in vitro development of skin models is an interdiscipli- of a complex network of cells and extracellular matrix (ECM)
nary effort with a wide range of applications. Skin models pro- molecules. Immune responses maintain homeostasis and pro-
vide 3D platforms to study various aspects of skin biology and vide the host with defense against pathogenic microorganisms
enable the testing of substances for cosmetic and pharmaceu- in the tissue.[4] There are several differences between mouse
tical industries (reviewed by Groeber et al.).[1] Engineering skin and human skin immune systems including the presence
and location of differing dominant T cells, a differing array of
chemokines expressed, and the presence or absence of antimi-
Dr. A. Pupovac, Prof. S. L. McArthur crobial peptides. These differences may contribute to the failure
Faculty of Science of translating mouse in vivo skin model outcomes to human
Engineering and Technology
Swinburne University of Technology skin disease.[3] 3D in vitro models that reconstitute the human
Hawthorn, Victoria 3122, Australia immune system would allow the development of new thera-
E-mail: smcarthur@swin.edu.au peutic materials for treatment of infectious diseases, delivery
Dr. A. Pupovac, Prof. S. L. McArthur agents, and also give insights into inflammatory pathologies.
Commonwealth Scientific and Industrial Research Organization (CSIRO) However, the majority of current 3D skin models do not take
Probing Biosystems Future Science Platform and Manufacturing
Clayton, Victoria 3168, Australia
immune components into account, a factor that has been high-
Dr. B. Senturk, C. Griffoni, Dr. K. Maniura-Weber, Dr. M. Rottmar
lighted in recent reviews on skin disease models.[2,5]
Laboratory for Biointerfaces Recent advances in tissue engineering and our under-
Empa standing of immune cell biology have seen a number of
Swiss Federal Laboratories for Materials Science and Technology researchers start to include immune cells within 3D skin
9014 St. Gallen, Switzerland models, making important steps toward recapitulating the func-
E-mail: markus.rottmar@empa.ch
tions of native human skin. The main challenge of developing
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adhm.201701405.
an immunocompetent 3D skin model remains the effective
inclusion of multiple immune components and the creation
DOI: 10.1002/adhm.201701405 of an environment that allows and regulates the cross talk

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (1 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

between those components.[6] It is only once these challenges


are addressed that it will be possible to properly mimic the Aleta Pupovac is currently a
pathogenesis of skin diseases in vitro. In this progress report, postdoctoral researcher at
the major components, challenges, and limitations of current the Commonwealth Scientific
skin models are discussed, with a focus on the immune com- and Industrial Research
ponent. Additionally, we discuss recent studies on applicability Organization (CSIRO) and
and limitations of endpoint and real-time monitoring tech- Swinburne University of
niques for inflammatory responses in skin models. Technology, Australia. She
received her Ph.D. in cell
biology and immunology at
the University of Wollongong,
2. Skin Immunity and Skin Models
Australia, and completed
Skin is a functionally heterogeneous and structurally complex 1.5 years of postdoctoral
organ. It is composed of three structural layers: the epidermis, research at Monash University, at the Biomedicine Discovery
dermis, and hypodermis. The epidermis is the avascular, out- Institute, Australia. Her current research interests include
ermost layer that consists of stratified epithelia exhibiting cru- infection and inflammatory pathways in 3D cell culture
cial regulatory and defensive functions including the regulation systems, tissue engineering, biomaterials, and biosensing.
of water loss,[7] defense against UV radiation[8] and infection.[9]
This layer predominantly consists of keratinocytes, which are Markus Rottmar is a group
organized into sublayers and exist at different stages of dif- leader in the Biointerfaces Lab
ferentiation. The dermis is the intermediate layer and is con- at Empa, the Swiss Federal
nected to the epidermis via the basement membrane. Blood Laboratories for Materials
and lymphatic vessels, nerves, sweat glands, and hair follicles Science and Technology,
reside in this extracellular-matrix-rich compartment which is Switzerland. He received his
secreted by resident fibroblasts. The deepest layer is the hypo- Ph.D. in the field of bioma-
dermis, which is mainly composed of loose connective tissue terials from ETH Zurich and
and adipose tissue that consists of fat-storing cells called adipo- with his team, he is interested
cytes. These inner layers provide nourishment, insulation, and in developing advanced,
energy reserves for the body.[10] A variety of cell types contribute predictive in vitro models
to immune regulation in human skin. The functions and ori- to understand and steer the
gins of these cells are listed in Table S1 and Figure S1 (Sup- interaction of human cells/tissues with materials relevant
porting Information). for skin wound healing and osseointegration.
Animal skin has a different structure and behavior compared
to human skin, making it difficult to unravel complex wound Sally L. McArthur is a
healing and disease processes in animal models. The develop- professor of biomedical
ment of functional in vitro skin models has also broadened the engineering at Swinburne
scope for the investigation of fundamental biological processes University of Technology
in human systems, which can in turn contribute to the devel- and a research science
opment of novel human therapies. Nonetheless, current 2D leader in biomaterials at the
and 3D in vitro models of healthy skin and skin disease do not Commonwealth Scientific
exhibit the complexity and heterogeneity of human skin. Sev- and Industrial Research
eral 2D models of skin disease have been described, including Organization (CSIRO) in
models of fibrosis, wounding, autoimmune disorders, cancer, Melbourne Australia. She
and infection.[2,11] However, it is well established that models has held academic roles in
utilizing 2D culture are not representative of in vivo conditions the United Kingdom and
and do not recapitulate the 3D environment of cells.[12] Australia for 15 years and leads research teams at both
More recent tissue engineered 3D skin models represent the CSIRO and Swinburne focusing on the development of 3D
most advanced constructs, and mimic the nature of skin by con- Cell Culture Systems and the development of coatings for
taining both epidermal and dermal layers. Current skin models, biointerface engineering, respectively.
with their respective components, applications, and limitations
are listed in Table 1. Applications of these models include basic,
pharmacological, and cosmetics research. The in vitro testing of
finished cosmetic products and cosmetic ingredients in Europe Epidermal 3D skin models[13,14] are actively used for in vitro
has been mandatory since 2013, since the European Commis- evaluation of ultra violet (UV) exposure,[15] bacterial adhesion,[16]
sion released a ban on animal research. The methods to eval- and penetration/permeation studies (Table 1).[17] However, these
uate irritation, corrosion, and sensitization of chemicals on skin models do not have a dermal layer or immune cells, which will
have been validated by the Organization for Economic Co-oper- likely affect data obtained in these studies. For example, the
ation and Development (OECD), who are responsible for the native epidermis contains melanocytes which are involved in
regulatory acceptance status of alternative methods. UV protection,[18] thus the inclusion of these cells are required

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (2 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Table 1.  Currently available skin equivalents and skin models, listed with their respective applications and limitations.

Model Components Application/study Limitations References


Reconstructed epidermis – Collagen, keratinocytes (EpiSkin) Skin irradiation, corrosion, penetration, No dermal layer, no immune [13,14,16]
models UVa) damage, bacterial adhesion, cells
epidermal permeability
–  Keratinocytes, melanocytes
Full-thickness skin models – Normal or disease patient derived Dermal penetration, absorption No immune cells [17,19–25]
differentiated keratinocytes on testing, fibrosis, autoimmune disease,
fibroblast-populated dermis wound healing, skin infection
Full-thickness skin model – Fibroblasts, keratinocytes, RAW Test model for skin inflammation RAW 264.7 is a mouse cell [30]
containing macrophages 264.7 cell line
Full-thickness skin models – Collagen, fibroblasts, keratinocytes, Solar UV radiation testing, drug Other immune cells missing [32,35,36]
containing LCb) and/or DCc) mo-LCs and mo-DCsd) and chemical testing
–  MUTZ-3 derived LC
– mo-DC
Full-thickness skin models – Normal or iPSCe)-derived fibroblasts, High potential for drug discovery, Poor melanocyte viability, [26,27,29]
containing melanocytes and/ keratinocytes, melanocytes, and comparison of normal versus cancerous no immune cells
or endothelial cells endothelial cells melanocytes, wound healing
Full-thickness skin model – Collagen, fibroblasts, and Fungal infections Other immune cells missing [34]
containing T cells keratinocytes, CD4+ T cells
Organotypic tumor coculture – Collagen, fibroblasts, macrophages, Tumor progression No healthy keratinocytes; only [31]
squamous cell carcinoma cells represents cancerous skin

a)UV: ultraviolet; b)LC: Langerhans cell; c)DC: dendritic cell; d)mo-DC: monocyte derived DC; e)iPSC: induced pluripotent stem cell.

in the evaluation of UV exposure. 3D full thickness skin models Only a handful of full-thickness skin equivalent (FTSE)
which contain both epidermal and dermal layers have also been models that contain immune cells have been created (Table 1,
developed for a variety of applications and studies including Section 3). While these models also lack the structural and
dermal penetration, absorption testing, fibrosis, autoimmune immune complexity of native skin, they retain the foundations
disease, wound healing, and skin infection[19–22] (Table 1). required for the eventual realization of physiologically relevant
Dermal penetration studies and absorption tests are more rele- and immune competent skin.
vant in these models compared to 3D epidermal models, as they
better recapitulate native skin thickness and cell–cell interac-
tions. Skin infection and disease models have also been created, 3. Current Immune Competent 3D Skin Models
including a model of percutaneous worm invasion,[23] bacterial
infection,[24] and psoriasis.[25] In vitro studies comparing normal 3.1. Construction Methods
skin cells to psoriatic biopsy-derived cells in 3D constructs have
led to better characterization of the disease at a cellular level.[25] 3D culture provides the opportunity to coculture multiple cell
However, these models also lack other cell types including types leading to more complex cell systems. In 2D culture, cells
immune cells. are usually grown in monolayers on polystyrene or glass, while
Other 3D full thickness models have incorporated addi- 3D cultures grow in scaffold-free aggregates and spheroids,
tional cell types to understand the role of these cells in skin or scaffold-based matrices.[12] In the context of infection and
health and disease, and to increase the complexity of cur- inflammation, most current 3D models of immune competent
rent skin models. Deeper understanding of the interaction skin are reduced systems that are generated in Transwell,[30–34]
between melanocytes and the resident skin cell populations, Snapwell,[35] or CellCrown[36] tissue culture inserts and plates.
fibroblasts and keratinocytes, has been achieved using 3D The majority of these models comprise a fibroblast populated,
in vitro models[26,27] and currently, a standardized protocol biologically derived collagen scaffold (dermal layer), seeded
even enables the reproducible creation of a skin melanoma with keratinocytes (epidermal layer) that are eventually cultured
model.[28] Melanocytes and endothelial cells have also been at an air–liquid interface to generate a FTSE (Figure 1a). Others
used to create a more complex wound model to study cell have placed epidermal sheets from skin biopsies onto acellular
mobility in wound healing.[29] More comprehensive lists of dermis (stratum corneum side up) and cultured them exposed
2D and 3D skin models and their applications and limita- to air. Subsequently, these are placed onto fibroblast cultures
tions have been reviewed by Bergers et al.[2] and Mohammadi to allow for repopulation of the dermis and the generation of
et al.[11] It is apparent that in all of these models, including a FTSE (Figure 1b).[37] Generally, these models are validated
those that have been used to study immune-based disease, against native skin by hematoxylin and eosin staining, and
have largely neglected the role of immune cells and inflam- immunostaining for markers expressed in the different skin
matory processes. layers (Figure 1c).

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (3 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 1.  Full thickness skin equivalent generation (FTSE). Fibroblast populated a) collagen or b) decellularized dermis are seeded with keratinocytes in
culture. After culture at air–liquid interface the differentiated keratinocytes represent the epidermis and the fibroblast-populated scaffolds represent the
dermis. c) Native skin and FTSEs generated with primary cells or cell lines after hematoxylin and eosin (H&E) staining and immunohistochemical staining
for specific markers vimentin, keratin 5, and keratin 10. Adapted with permission.[60] Copyright 2015, the author; published by Mary Ann Liebert, Inc.

To create immunocompetent models, up to two immune cell Macrophages have been placed into models either via
types have been incorporated during or after the generation of seeding at the bottom of transwell plates,[30] or seeding into col-
the FTSE. These are usually sourced from blood or are incor- lagen scaffolds together with fibroblasts (Figure 3a,b).[31] They
porated as cell lines (Section 3.3) and include langerhans cells have been visualized using cell-specific markers including the
(LC),[32,35,37–39] dendritic cells (DC),[35,36] macrophages,[30,31] macrophage non-inflammation marker, CD209, and pan mac-
and T cells.[34] Whole blood and peripheral blood mononu- rophage marker CD68 (Figure 3b).[31] Whole blood, PBMCs, or
clear cells (PMBCs) (which contain different immune cell T cells have been incorporated into collagen and placed under-
types) have also been incorporated.[34] LCs and DCs have been neath FTSEs in transwell inserts (Figure 3c).[34] During model
co-seeded with keratinocytes on the dermal layer during epi- generation, the addition of these cells into the scaffold represents
dermal equivalent generation (Figure 2a).[32,35,38] LCs have also a more 3D-like approach that mimics natural skin structure
been placed beneath FTSEs and allowed to migrate to the epi- compared to cells seeded at the bottom of transwell plates,
dermis (Figure 2b).[37] These cells are usually visualized in the which does not allow migration and cell–cell contact. Never-
models using immunostaining against cell-specific markers theless, while these immune competent models may resemble
including langerin, human leukocyte antigen DR-1, and den- some components of native skin, in terms of immune cell inte-
dritic-cell-specific intercellular adhesion molecule-3 grabbing gration their entirety remains more reminiscent of “sandwich”
non-integrin (Figure 2a,b). Dendritic cells have also been style cocultures rather than true 3D representations. Others
incorporated into an agarose-fibronectin layer that was placed have used human skin explants to generate full-thickness
between a fibroblast-seeded synthetic microfiber scaffold and models that recapitulate complexity of human skin, including
a keratinocyte-populated microfiber scaffold, forming a sand- the presence of innate LCs and T cells, however culture times
wich style structure (Figure 2c). These cells were tracked in the of these models are limited to 7 d before cell viability becomes
model using a fluorescent dye (Figure 2c).[36] an issue.[33] Future developments should examine alternative
Spatial organization and the physical constraints provided by methods of construction that represent more complex and rel-
3D culture influence gene expression and cellular behavior.[12] evant skin. This may be achieved by integrating immune cells
In this regard, keratinocytes and fibroblasts require cross talk that can form interactions with adjacent cells types with min-
in a bi-layered cellular construct for production of several rel- imal support and interference (see Section 3.4).[41]
evant growth factors and cytokines.[40] Thus, the presence of an
agarose-fibronectin immune cell layer in the FTSE (Figure 2)
may impede on fibroblast-keratinocyte cross talk. Furthermore, 3.2. Scaffold Choice
the presence of high water content gel layers within the struc-
ture may complicate skin impedance readings.[36] While “sand- The majority of immune competent FTSEs contain a scaffold
wich” style construction may be advantageous for separation generated from rat tail collagen I gels.[30–32,34,39] Collagen I
of cells for analysis post use, ultimately the separation and/or hydrogels are a popular extracellular matrix choice for 3D fibro-
location of the inflammatory cells could be expected to influ- blast culture, as collagen I is the most abundant ECM compo-
ence cell–cell signaling. This, along with other processes needs nent found in native skin tissue and because these hydrogels
to be evaluated and validated for the specific application of the can be formed with appropriate matrix densities, stabilize cells,
skin models. and contain cell adhesion sites.[42] Collagen gels also undergo

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (4 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 2.  Dendritic and Langerhans cell locations in full thickness skin equivalent (FTSE) models. a) The schematic (left panel) shows that dendritic
cells and Langerhans cells are coseeded with keratinocytes during FTSE generation. After culture at air–liquid interface they migrate into the dermis
and epidermis, respectively. Experimental results (right panel) show that monocyte-derived Langerhans cells (mo-LC) are visualized in the epidermis
after immunostaining of langerin and human leukocyte antigen DR-1 (HLA-DR). Dermal dendritic cells (mo-DDC) are visualized in the dermis after
staining of HLA-DR, and dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) markers. The dotted line represents
the dermal–epidermal junction. Scale bar: 20 µm (Adapted with permission.[35] Copyright 2007, Mary Ann Liebert, Inc.). b) The schematic (left panel)
shows that FTSEs cultured with decellularized dermis are placed onto cultured Langerhans cells, which migrate to the epidermis during culture at
air–liquid interface. Experimental results (right panel) show immunostaining of markers for Langerhans cells including langerin and HLA-DR. Scale
bar: 20 µm (Reproduced with permission.[37] Copyright 2012, Elsevier). c) The schematic (left panel) shows that microfiber scaffolds are seeded with
keratinocytes or fibroblasts to form separate layers, while dendritic cells are incorporated into an agarose-fibronectin layer. These layers are assembled
in a “sandwich” style so that the dendritic cells sit between the dermis and epidermis. Experimental results (right panel) show monocyte-derived
DCs (middle layer) visualized using Cell Tracker Red between microfiber scaffolds (top and bottom layers) that contain keratinocytes and fibroblasts,
respectively. Scale bar: 20 µm (Adapted under the terms of the CC BY 3.0 license.[36] Copyright 2013, IOP Publishing Ltd.).

cell-mediated contraction, which occurs as a result of mechan- collagen type-III, elastin, laminin, or fibronectin play important
ical interaction of keratinocytes and fibroblasts with the col- roles in native skin but are largely neglected in current FTSEs.
lagen hydrogel.[43,44] Cell-mediated contraction is an important Yet, these components are likely important for such models,
process in wound healing[45] and has been used as a measure of as different ECM fragments are known to be involved in skin
cell activation in many models.[46–48] While this feature is neces- inflammatory responses.[52] For example, Almine et al. showed
sary in wound healing processes, it can be a source of variation that the interaction of elastin with human dermal fibroblasts
that impedes on the generation of standardized skin models. induces the expression of matrix metalloproteinase-12 and
Moreover, the variability associated with the use of different chemokines that trigger a pro-inflammatory response.[53]
batches of collagen makes it difficult to achieve the same struc- To address some of the issues associated with the use of
tural and mechanical properties. Hence, collagen alone may collagen-only scaffolds, immune competent FTSEs have been
not be useful for models with applications that require testing generated using modified collagens. Bechetoille et al. have
skin responses to drugs. Additionally, fibroblasts, keratinocytes, generated a dermal scaffold in the form of a porous sponge,
and macrophages secrete collagenases such as matrix metal- composed of types I and II bovine collagen containing chi-
loproteinases that degrade collagen gels,[49–51] therefore this tosan and the glycosaminoglycan, chondroitin-4 sulfate.[35]
scaffold may also not be useful for models requiring long-term While Kuhbacher et al. have combined the glycosaminoglycans,
skin culture or skin survival. Other ECM components such as chondroitin-4 sulfate, and chondroitin-6-sulfate with rat tail

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (5 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 3.  Macrophage and T cell locations in full thickness skin equivalents (FTSEs). The schematics show that macrophages are a) seeded at the bottom of
transwell plates or b) (left panel) coseeded with keratinocytes, and migrate to the dermis after culture at air–liquid interface. b) (right panel) Experimental results
show macrophage migration to the tumor epithelium (TE) and the pan macrophage marker CD68 and macrophage M2 polarization marker, CD209 were
visualized in the dermal equivalent (DE) in the presence or absence of interleukin 4 (IL-4) treatment (Reproduced under the terms of the CC BY 3.0 license.[31]
Copyright 2012, the authors, published by PLOS). c) The schematic shows that T cells are incorporated into a collagen scaffold and placed underneath the FTSE.

type I collagen.[34] The addition of these components endows Decellularized tissue scaffolds have been used both in vitro
mechanical strength to the scaffold without compromising cell and in vivo to investigate the cellular interactions, organ forma-
attachment and growth.[54] During culture, the collagen-glycosa- tion and transplantation in tissue engineering and regenerative
minoglycan-chitosan based dermal scaffold does not contract, medicine.[57] While human decellularized dermis is currently
unlike its collagen-only counterpart.[35] This scaffold also allows the best scaffold option for the creation of clinically utilized
for long-term culture (up to 49 d) of the FTSE, which may be bioengineered skin constructs,[58] whether it is the best scaffold
valuable in infection and inflammation studies that incorporate option for the creation of immune competent skin models that
immune cells and living infectious agents.[34] are needed to ascertain immunological mechanisms in disease
Others have facilitated a FTSE with similar characteristics remains to be determined. Human dermis is usually harvested
to native skin by cross-linking succinimidyl glutarate poly- from cadavers, surgical skin donations, or animal organs and
ethylene glycol with a collagen hydrogel to create mechanical thus may not be easily sourced or applicable for skin models
strength and less cell-mediated contraction.[55] Modified col- that require high throughput applications. Decellularized
lagens may improve model variability and the complexity of dermis has been used as a scaffold to create an immune com-
3D immune competent skin by allowing the incorporation of petent skin model to study chemokine-driven migration of
vasculature (see Section 3.4). This was exhibited with the use LC-like cells.[37] The same group has also used a collagen-based
of a cross-linked porous bovine I collagen gel that was seeded scaffold to study the migration of these cells.[32] While this indi-
with endothelial cells suspended in a fibrin gel.[56] Using col- cates that decellularized dermis may not be required for cer-
lagen in conjunction with fibrin provided the model with a tain applications, a decellularized porcine jejunum scaffold has
greater vascular capacity compared to fibrin alone. As com- been successfully used to create a perfusable vascularized skin
plexity increases, scaffold choice and how it is utilized becomes model and such vascularization may be necessary for the crea-
increasingly important with respect to physiological relevance. tion of immune cell flow (see Section 3.4).[59]
Considering this, Chau et al. have separately populated a syn-
thetic, porous microfiber-based scaffold with fibroblasts or
keratinocytes as shown in Figure 2. This scaffold is commer- 3.3. Cell Types and Sources
cially available and does not significantly affect cell viability.[36]
However, this scaffold allowed keratinocytes to permeate The cells used to generate immune competent FTSEs are usu-
through it, consequently disrupting the brick-and-mortar like ally partially or fully sourced from donor skin samples and
structure of the epidermis.[36] Even though the authors show peripheral blood.[30–32,34,35] Primary cells may be harder to
staining of some differentiation markers, this model is not source regularly and are endowed with donor variation.[5] To
ideal when comparing the structure to native skin in terms of overcome donor variation in LCs sourced from blood, Ouwe-
promoting a more natural 3D structure. hand et al. incorporated LCs differentiated from the human

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (6 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

acute myeloid leukemia cell line, MUTZ-3 (MUTZ-LC) into it difficult to fine-tune and maintain cell differentiation toward
their FTSE. However, the FTSE itself was generated using the desired phenotype. For example, macrophages are respon-
primary skin samples.[32] Others have used cell lines to create sible for both pro-inflammatory (M1) responses as well as the
models in a disease context, where mouse or human FTSEs switch from a pro-inflammatory to a reparative (M2) environ-
were generated to resemble skin in a tumor-associated envi- ment, and the resolution of inflammation in skin wounds.[63]
ronment.[31] These contained an epidermal equivalent gen- However, this dualistic role and functional heterogeneity also
erated from the mouse or human squamous cell carcinoma represents a limitation when considering their implementation
cell lines PDVA and A-5RT3, respectively, but the monocytes/ into 3D in vitro skin models. Nonpolarized (M0) macrophages
macrophages in these models were sourced from peripheral that are derived from peripheral blood monocytes can easily
blood.[31] be isolated from blood and differentiated into M1 or M2-like
Using the telomerase reverse transcriptase (TERT)-immortal- macrophages. While macrophage colony stimulating factor or
ized, N/TERT-1 keratinocyte and BJ-5ta fibroblast cell lines, an granulocyte-macrophage colony stimulating factor is required
all cell line FTSE was developed that was comparable to native for monocyte survival and differentiation into macrophages,[64]
skin (Figure 1c).[60] However, it did not contain any immune both cytokines distinctly influence the cells’ expression profiles
cells. To the best of our knowledge, no one has created an and by that, prime them toward M1- or M2-like macrophage
immune competent FTSE composed entirely out of cell lines. populations.[65] The M1-like phenotype is normally induced
It would be vital to determine whether incorporating immune by supplementing the medium with interferon-γ [66] and LPS
cell lines such as MUTZ-LC into an all cell line model, allevi- or tumor necrosis factor-α,[67] while M2-like macrophages are
ates the variability associated with donor samples, and whether obtained when supplementing the medium with the cytokines
the immune response in such a model is comparable to that interleukin-4 and/or IL-13.[68] Cell survival also depends on
of its primary counterparts. It is also important to consider macrophage types, M2-like macrophages survive longer than
whether the cell types being used to create immune competent M0 and M1-like macrophages.[69] Thus, defining the purpose
skin are physiologically relevant. In this regard, Chung et al. and biological processes involved in skin infection, injury and
used a mouse macrophage cell line within a human FTSE to disease is critical before the introduction of relevant immune
study lipopolysaccharide (LPS)-induced immune response.[30] cell types.
Mouse and human macrophages have a different response to
LPS treatment,[61] so using human monocyte/macrophage cell
lines such as THP-1 or U937 would be more physiologically 3.4. Requirements for Physiologically Relevant Skin Models
relevant in such a model. Others have created FTSEs using
HaCaT keratinocytes, which display deficient epidermal differ- The ideal skin model would contain many different cell types,
entiation[62] and therefore do not appropriately mimic the native appendages, adipose tissue, and vasculature that recapitulate
epidermis. skin structure and function. This model should support long-
Despite considerable efforts to create immunocompetent term culture and encompass devices that monitor real-time cel-
skin models to date, a limited variety of immune cells (max- lular behavior in health and disease (Figure 4).[11] Yet, several
imum of two) have been incorporated into current immune hurdles are faced when trying to construct the ideal or physio-
competent skin models.[35] To increase the complexity of skin logically relevant skin model for research purposes. Skin models
models, several immune cell types should be incorporated. Con- tend to be designed in a highly context dependent manner and
versely, increasing the number of cell types may also increase thus require unique design concerns. First, the biological con-
variability, thus a trade-off exists between model complexity and text and complexity of the model must be established. Once
variability. Recently, Kuhbacher et al. incorporated whole blood established, several considerations are apparent including the
and PBMCs into their FTSEs to study skin infection by Candida effect of multiple cell types, that is, cell–cell ratios and cell–cell
albicans (C. albicans).[34] In terms of immune cell variety, this interactions, which will affect the expression of physiological
FTSE represents the most sophisticated immune model to date. surface markers and the migration of cells within the model.[70]
Additionally, it is the only FTSE containing immune cells that Concurrently, matrix considerations including scaffold choice
has incorporated a living infectious agent to study skin infec- and spatial interactions of cells within the model (2D vs 3D),
tion. It was demonstrated that whole blood or PBMCs (which as well as the effect of culture requirements including culture
contain T cells), and CD4+ T cells alone inhibited C. albicans- times, media constituents, and media diffusion are relevant.[70]
induced dermal invasion to similar degrees. As efficient protec- These considerations are required to maintain a physiologically
tion was achieved with the incorporation of CD4+ T cells alone, relevant model while achieving research purposes.
FTSEs containing these cells were used to determine mecha- To the best of our knowledge, a complex immunocom-
nisms of C. albicans invasion.[34] Although less physiologically petent skin model, containing more than a few immune cell
representative, one cell type achieved the same response and types in long-term culture has not been generated. Incorpo-
allowed a simpler approach to relevant model generation. rating a wide variety of cells to generate complexity may be
Avoiding the inclusion of redundant cell types may be advanta- realized with the use of induced pluripotent stem cells. iPSCs
geous in terms of reproducibility, while achieving similar out- are derived from differentiated somatic cells that are repro-
comes. Such an approach to cell choice may also be useful when grammed to a pluripotent state with the potential to become
generating a model of skin infection where immune response many different cell types.[71] iPSCs have been used to gen-
mechanisms are undefined. It is challenging to precisely regu- erate keratinocytes and fibroblasts to create a whole iPSC-
late the microenvironment where cells are introduced, making derived FTSE,[27] iPSC-derived melanocytes,[27] and endothelial

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (7 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

of inflammatory diseases with vascular


involvement remains a major challenge
in skin immunology. Additionally, the vas-
cularization of skin models is required to
address diffusion limitations of media in
models with matrix-rich environments and
remains a solid requirement for the suc-
cessful implementation of shared nutrients
or media, and immune cell flow.[70] There
are several vascularization strategies in skin
tissue engineering including those that are
scaffold-based, cell-based, and emerging.[78]
Modifying scaffolds can increase intrinsic
vascularization (see Section 3.2)[56] and cell-
based technologies such as iPSCs can be
used to create endothelial cells that generate
vascular structures.[72] Likewise, emerging
strategies such as 3D bioprinting, cell sheet
engineering, and microfluidics are impor-
Figure 4.  The ideal skin model. The ideal skin model should encompass native skin structure tant for the realization of more complex
and function by containing many different cell types, appendages, adipose tissue, and vasculature.
immunocompetent skin.[78] The bioprinting
It should support immune cell flow, long-term culture, and encompass devices that monitor
real-time cellular behavior and biomarkers in health and disease. of skin models is gaining traction as a result
of the need to reduce the amount and cost
of animal experimentations.[79] Several skin
cells [72] have also been integrated into FTSEs, exemplifying constructs have been bioprinted including models that mimic
the capability of iPSCs to produce different skin cell types and wound healing, in vitro models for validation and testing, and
increase the complexity of current skin models. In the near constructs that encompass appendages, pigmentation, and vas-
future, iPSCs may be used to introduce immune cells into culature, all of which, to some extent recapitulate the native
the skin models. Furthermore, stem cells have been used to skin. Nevertheless, bioprinting faces challenges in replicating
generate 3D models that can be cultured for up to 45–50 d, native skin, particularly the complex ECM structure.[79] To intro-
and therefore may be advantageous for developing models in duce vasculature into skin models, the major approaches have
long-term culture.[73,74] Currently, it is known that iPSCs can be been printing endothelial and smooth muscle cell on hydrogel
differentiated into multiple functional lymphocyte lineages,[75] layers[80] and printing channels that are subsequently seeded
however skin-specific immune cells such as LCs have not with cells.[81,82]
been generated. Human iPSC-derived macrophages, which are The aforementioned strategies will provide additional user
developmentally related to tissue resident macrophages such as control of construction factors and scaffold choice,[11] hence a
LCs[76] may warrant use in FTSEs. iPSCs may also facilitate the combination of vascularization strategies may generate physi-
development of personalized skin models, enabling drug and ologically relevant models. This is becoming evident with the
other treatment methodologies to be evaluated and tailored to construction of several user-controlled vascular models. A
the specific patient’s needs.[5] Specialized medium is required human FTSE that contains spatially controlled perfusable vas-
to maintain different cell types, thus the combination of many culature was produced using microfabrication and micropat-
cell types in one model containing one type of medium may terning techniques and iPSC-derived endothelial cells.[72] Addi-
result in aberrant marker expression that may not be physio- tionally, a novel FTSE containing blood and lymph-like capil-
logically relevant. This will subsequently affect cell differentia- laries was constructed using a cell accumulation technique
tion, viability, and migration within the model. As iPSCs allow whereby cell surface coating was produced using layer-by-layer
for directed differentiation,[71] they may be used as a future tool assembled nanofilms of extracellular matrix and the coculture
to control marker expression of cells. Although for this to be of human umbilical vein endothelial cells and human dermal
possible, the use of this tool must be accompanied with the lymphatic microvascular endothelial cells.[83] This process
creation of a medium or a blood substitute that can successfully eliminated the use of collagen hydrogels and provided more
maintain many cell types at a time. reliability between models.[83] Moreover, this model contained
The cutaneous vascular system, which is missing in estab- lymph-like capillaries that may subsequently form lymphatic
lished immunocompetent skin models, is critical for homeo- structures that allow immune cell travel. Spatial control exhib-
stasis and inflammatory responses in native skin. Blood and ited by these models will also be useful in mimicking skin het-
lymphatic vessels supply oxygen and nutrients, and allow erogeneity in disease versus healthy states.
immune cell and cytokine infiltration to the skin during While these models are relevant for the analysis of static top-
pathogen invasion. Many inflammatory skin diseases are ical delivery of drugs or infectious agents, systemic delivery of
associated with insufficient or overactive vasculature.[77] Due drugs, infectious agents, or immune cells cannot be achieved
to the absence of vasculature systems in current immuno- without blood and lymphatic flow. Recently, a model of vascu-
competent skin models, understanding the mechanisms larized skin was generated that may allow flow of immune cells

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (8 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

through the construct. Groeber et al. described a FTSE based wide range of responses, including the expression of cytokines
on decellularized porcine jejunum with a perfused vascular such as IL-1β, IL-6, and IL-8.[34]
network generated using a bioreactor for dynamic culture.[59] The pro-inflammatory cytokines TNF-α, IL-1α, IL-1β, IL-6,
Using this system they were able to obtain similar physiolog- and IL-8, as well as the immune suppressive IL-10 have been
ical perfusion of capillary blood flow and bidirectional mass measured in response to a variety of inflammation or infection
transport.[59] By increasing the complexity of models through inducing agents including UV radiation,[35] LPS,[30] and sensi-
the introduction of flow, the stress applied from flow force on tizing agents.[36] Soluble cytokines, chemokines, and their recep-
cells may affect cell viability and activate cells through mecha- tors can act in complex networks making it difficult to target a
notransduction.[70] The mechanical signal to cells is converted particular cytokine or chemokine as a biomarker of inflamma-
to signaling pathways that regulate gene expression, cell prolif- tion and infection.[87] In future immunocompetent models, tar-
eration, and differentiation.[84] Understanding the mechanisms geting a variety of inflammatory biomarkers including cytokines
involved in mechanotransduction signaling in skin models may be more effective. When the skin barrier is compromised
and controlling the duration and frequency of the flow rate in during inflammation and disease, this affects skin pH, protease
models may eventually help to control cell viability and main- activity, and cytokine secretion.[87] Other biological indicators of
tain physiological levels of cell proliferation and differentiation. inflammation include the increased production and release of
Microfluidics platforms are being utilized to create skin- antimicrobial peptides and damage associated molecular pat-
on-chip models in pursuit of a controlled environment and an terns.[87] These features may be useful as potential novel bio-
exchange of immune cells (reviewed by van den Broek et al.).[5] markers of skin inflammation and disease in new generation
Noticeably, many of these are reduced models that lack a FTSE skin models. Further, the activation properties of immune cells
and immune cells. One skin-on-chip model, dubbed as being may also be explored as potential novel biomarkers. Incidentally,
immunocompetent, includes the U937 monocyte/macrophage several smart fluorescent probes have been developed for detec-
cell line with the HaCaT keratinocyte cell line in a device with tion of macrophage activity including migration, reactive oxygen
a perfusion setup.[85] This device allowed the monitoring of and nitrogen species release, and matrix metalloproteinase and
monocyte and keratinocyte interactions after UV irradiation or cathepsin activation.[88] Others have created reporter cell lines
LPS stimulation in dynamic culture. It also contained an in- that have been integrated into FTSEs to elucidate mechanisms
built barrier function readout using transepithelial electrical of infection progression. Kuhbacher et al. used photometric
resistance.[85] While this device did not utilize a FTSE, real-time measurement of secreted alkaline phosphatase by S1F fibro-
monitoring of skin-relevant processes is evident. Ultimately, a blasts containing the nuclear factor kappa-light-chain-enhancer
combination of the aforementioned technologies and real-time of activated B cells (NF-κB) reporter gene, which allowed meas-
monitoring of skin processes can lead to the creation of the urement of toll-like receptor activity.[34] Methods such as these
ideal skin model (Figure 4). may be useful for future immunocompetent skin designs that
take advantage of in situ measurements of activity.[2]

4. Monitoring Infection and Inflammation in


Immunocompetent Skin Models 5. Conclusion
The barrier function provided by skin is an important mech- Much of research and development in skin health and disease has
anism of protection against infection and disease.[86] Many traditionally been centered on the use of 2D cultures, cocultures,
skin models have used transepithelial impedance measure- and animal models. Since the European Union’s ban on animal
ments as an indicator of barrier function in response to treat- testing of cosmetics in 2013, there has been a rise in the construc-
ment.[36,59,83,85] Skin impedance measurements are minimally tion and implementation of more physiologically relevant and 3D
invasive and permit real-time measurement. However, when human skin models. This has resulted in the development of a
used alone, this technique does not serve as a relevant bio- variety of epidermal and FTSE models to study disease, as well as
marker of infection and inflammation in the skin. Many of the skin substitutes for clinical applications. However, current models
methods used to study skin immune responses are endpoint do not fully recapitulate native skin. A handful of groups have
measurements that involve deconstructing the 3D structure constructed skin models that have integrated several immune
of models. Cell fluorescence imaging and immunostaining cell types including LCs, DCs, macrophages, and T cells into 3D
are abundantly used to track the migration and phenotype of FTSEs, paving the way for the development of more relevant and
DCs[32,35] and macrophages.[30,31] Furthermore, antibody array immunocompetent skin models. Construction approaches of
assays and enzyme-linked immunosorbent assays are routinely these models focus heavily on the use of transwell systems, col-
used to show that the microenvironment of the models con- lagen-based scaffolds, and human-sourced primary cells. Although
taining immune cells successfully maintain their phenotypes, useful, coculture type systems are not truly representative of native
and that the model responds to treatment through the meas- 3D skin. Further, the need exists to design scaffolds that withstand
urement of cytokine expression and secretion.[30,35,36] Other long-term culture and treatment with infectious agents, as well
methods used to detect immune responses include zymog- as appropriate and easily sourced cells. Together, these alterations
raphy, which has been used to observe collagenolytic activity may deliver more representative and reproducible results.
and expression of matrix metalloproteinases after macrophage Successful future creation of relevant models lies in inte-
integration into FTSEs,[31] and dual RNA-sequencing of FTSEs grating and implementing knowledge from materials science,
infected with C. albicans, which allowed the assessment of a engineering, and cell biology. Designing and incorporating

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (9 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

appropriate scaffolds and cells into models of disease is con- [2] L. Bergers, C. M. A. Reijnders, L. J. van den Broek, S. W. Spiekstra,
text-dependent and requires a solid understanding of biological T. D. de Gruijl, E. M. Weijers, S. Gibbs, Drug Discovery Today 2016,
processes involved in the disease, as well as materials engi- 21, 1479.
neering and design needs. Several technologies are paramount [3] P. A. Gerber, B. A. Buhren, H. Schrumpf, B. Homey, A. Zlotnik,
P. Hevezi, Biol. Chem. 2014, 395, 577.
in designing physiologically relevant immunocompetent skin
[4] J. M. Richmond, J. E. Harris, Cold Spring Harbor Perspect. Med.
models. The integration of iPSCs into models may allow for faster 2014, 4, a015339.
sourcing of cells, as well as more relevant cell choice to model [5] L. J. van den Broek, L. I. J. C. Bergers, C. M. A. Reijnders, S. Gibbs,
disease. It may also provide a more personalized or patient-spe- Stem Cell Rev. Rep. 2017, 13, 418.
cific approach to study disease, which would offer industry and [6] V. Lee, G. Singh, J. P. Trasatti, C. Bjornsson, X. Xu, T. N. Tran,
clinicians alternative test models for drug development. Using S. S. Yoo, G. Dai, P. Karande, Tissue Eng., Part C 2014, 20, 473.
current and newly emerging bioengineering techniques such as [7] Z. Zhang, Y. Hu, R. Yan, L. Dong, Y. Jiang, Z. Zhou, M. Liu,
bioprinting, cell sheet engineering, and microfluidics to integrate T. Zhou, N. Dong, Q. Wu, Sci. Rep. 2017, 7, 45262.
vasculature into immunocompetent models will eventually allow [8] Q. M. Hu, W. J. Yi, M. Y. Su, S. Jiang, S. Z. Xu, T. C. Lei, Cell Prolif.
immune cell flow and more relevant cell–cell interactions in skin 2017, 50, e12372.
[9] Y. Lai, A. L. Cogen, K. A. Radek, H. J. Park, D. T. Macleod,
models. Further, in situ measurements of model activity at the cel-
A. Leichtle, A. F. Ryan, A. Di Nardo, R. L. Gallo, J. Invest. Dermatol.
lular and molecular level will provide important biomarker read- 2010, 130, 2211.
outs in skin disease without the need to disturb or process the [10] A. Baroni, E. Buommino, V. De Gregorio, E. Ruocco, V. Ruocco,
model. Taking advantage of cellular processes and engineering R. Wolf, Clin. Dermatol. 2012, 30, 257.
requirements during the construction of new and more relevant [11] M. H. Mohammadi, B. H. Araghi, V. Beydaghi, A. Geraili, F. Moradi,
models will allow smart ways of integrating in situ sensors into P. Jafari, M. Janmaleki, K. P. Valente, M. Akbari, A. Sanati-Nezhad,
future skin models. Together the aforementioned aspects may Adv. Healthcare Mater. 2016, 5, 2459.
lead to the creation of an ideal skin model that is physiologically [12] R. Edmondson, J. J. Broglie, A. F. Adcock, L. J. Yang, Assay Drug
relevant and biologically informative in its contextual use. Dev. Technol. 2014, 12, 207.
[13] F. Netzlaff, C. M. Lehr, P. W. Wertz, U. F. Schaefer, Eur. J. Pharm.
Biopharm. 2005, 60, 167.
[14] J. Rehder, L. R. Souto, C. M. Issa, M. B. Puzzi, Sao Paulo Med. J.
Supporting Information 2004, 122, 22.
[15] F. Bernerd, D. Asselineau, J. Am. Acad. Dermatol. 2008, 58, S155.
Supporting Information is available from the Wiley Online Library or
[16] G. Lerebour, S. Cupferman, M. N. Bellon-Fontaine, J. Appl. Microbiol.
from the author.
2004, 97, 7.
[17] P. J. Hayden, M. Bachelor, S. Ayehunie, S. Letasiova, Y. Kaluzhny,
M. Klausner, H. Kandarova, Appl. In Vitro Toxicol. 2015, 1, 226.
Acknowledgements [18] M. Brenner, V. J. Hearing, Photochem. Photobiol. 2008, 84, 539.
[19] K. Ackermann, S. L. Borgia, H. C. Korting, K. R. Mewes,
A.P. and S.L.M. gratefully acknowledge funding from the Commonwealth M. Schäfer-Korting, Skin Pharmacol. Physiol. 2010, 23, 105.
Scientific and Industrial Research Organization (CSIRO) Probing [20] V. J. Moulin, Methods Mol. Biol. 2013, 961, 287.
Biosystems Future Science Platform, CSIRO OCE Science Leader [21] A. El Ghalbzouri, M. Jonkman, J. Kempenaar, M. Ponec, Am. J.
Program and Swinburne University of Technology. B.S. and M.R.
Pathol. 2003, 163, 1771.
gratefully acknowledge funding from the Commission for Technology
[22] Y. Xie, S. C. Rizzi, R. Dawson, E. Lynam, S. Richards,
and Innovation CTI (Grant No. 18524.2 PFLS-LS). S.L.M. and M.R.
D. I. Leavesley, Z. Upton, Tissue Eng., Part C 2010, 16, 1111.
gratefully acknowledge the Swiss National Science Foundation for the
Visiting Fellowship (Grant No. IZK0Z1_171124) with which support this [23] M. Jannasch, F. Groeber, N. W. Brattig, C. Unger, H. Walles,
review was developed. J. Hansmann, Exp. Parasitol. 2015, 150, 22.
[24] J. Shepherd, I. Douglas, S. Rimmer, L. Swanson, S. MacNeil, Tissue
Eng., Part C 2009, 15, 475.
[25] C. L. Barker, M. T. McHale, A. K. Gillies, J. Waller, D. M. Pearce,
Conflict of Interest J. Osborne, P. E. Hutchinson, G. M. Smith, J. H. Pringle, J. Invest.
Dermatol. 2004, 123, 892.
The authors declare no conflict of interest. [26] P. Eves, C. Layton, S. Hedley, R. A. Dawson, M. Wagner,
R. Morandini, G. Ghanem, S. Mac Neil, Br. J. Dermatol. 2000, 142,
210.
[27] K. Gledhill, Z. Y. Guo, N. Umegaki-Arao, C. A. Higgins, M. Itoh,
Keywords A. M. Christiano, PLoS One 2015, 10, e0136713.
3D skin models, inflammation, inflammatory cells, skin immunity [28] L. Li, M. Fukunaga-Kalabis, M. Herlyn, J. Visualized Exp. 2011, 54,
2937.
Received: December 1, 2017 [29] H. N. Monsuur, M. A. Boink, E. M. Weijers, S. Roffel, M. Breetveld,
Revised: January 29, 2018 A. Gefen, L. J. van den Broek, S. Gibbs, J. Biomech. 2016, 49, 1381.
Published online: March 15, 2018 [30] E. Chung, H. Choi, J. E. Lim, Y. Son, Tissue Eng. Regener. Med 2014,
11, 87.
[31] N. Linde, C. M. Gutschalk, C. Hoffmann, D. Yilmaz, M. M. Mueller,
PLoS One 2012, 7, e40058.
[1] F. Groeber, M. Holeiter, M. Hampel, S. Hinderer, K. Schenke- [32] K. Ouwehand, S. W. Spiekstra, T. Waaijman, R. J. Scheper,
Layland, Adv. Drug Delivery Rev. 2011, 63, 352. T. D. de Gruijl, S. Gibbs, J. Leukocyte Biol. 2011, 90, 1027.

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (10 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[33] B. De Wever, S. Kurdykowski, P. Descargues, Appl. In Vitro Toxicol. [60] C. M. Reijnders, A. van Lier, S. Roffel, D. Kramer, R. J. Scheper,
2015, 1, 26. S. Gibbs, Tissue Eng., Part A 2015, 21, 2448.
[34] A. Kuhbacher, H. Henkel, P. Stevens, C. Grumaz, D. Finkelmeier, [61] A. C. Thomas, J. T. Mattila, Front. Immunol. 2014, 5, 2448.
A. Burger-Kentischer, K. Sohn, S. Rupp, J. Infect. Dis. 2017, 215, 1742. [62] N. Maas-Szabowski, A. Starker, N. E. Fusenig, J. Cell Sci. 2003,
[35] N. Bechetoille, C. Dezutter-Dambuyant, O. Damour, V. Andre, 116, 2937.
I. Orly, E. Perrier, Tissue Eng. 2007, 13, 2667. [63] T. J. Koh, L. A. DiPietro, Expert Rev. Mol. Med. 2011, 13, e23.
[36] D. Y. Chau, C. Johnson, S. MacNeil, J. W. Haycock, [64] K. S. Akagawa, Int. J. Hematol. 2002, 76, 27.
A. M. Ghaemmaghami, Biofabrication 2013, 5, 035011. [65] A. J. Fleetwood, H. Dinh, A. D. Cook, P. J. Hertzog, J. A. Hamilton,
[37] K. Ouwehand, S. W. Spiekstra, T. Waaijman, M. Breetveld, J. Leukocyte Biol. 2009, 86, 411.
R. J. Scheper, T. D. de Gruijl, S. Gibbs, Eur. J. Cell Biol. 2012, 91, 765. [66] D. M. Mosser, J. P. Edwards, Nat. Rev. Immunol. 2008, 8, 958.
[38] V. Laubach, N. Zoller, M. Rossberg, K. Gorg, S. Kippenberger, [67] A. Mantovani, A. Sica, S. Sozzani, P. Allavena, A. Vecchi,
J. Bereiter-Hahn, R. Kaufmann, A. Bernd, Arch. Dermatol. Res. M. Locati, Trends Immunol. 2004, 25, 677.
2011, 303, 135. [68] S. Gordon, P. R. Taylor, Nat. Rev. Immunol. 2005, 5, 953.
[39] I. J. Kosten, S. W. Spiekstra, T. D. de Gruijl, S. Gibbs, Toxicol. Appl. [69] S. C. C. Huang, B. Everts, Y. Ivanova, D. O’Sullivan,
Pharmacol. 2015, 287, 35. M. Nascimento, A. M. Smith, W. Beatty, L. Love-Gregory,
[40] A. M. Wojtowicz, S. Oliveira, M. W. Carlson, A. Zawadzka, W. Y. Lam, C. M. O’Neil, C. Yan, H. Du, N. A. Abumrad,
C. F. Rousseau, D. Baksh, Wound Repair Regener. 2014, 22, 246. J. F. Urban, M. N. Artyomov, E. L. Pearce, E. J. Pearce, Nat.
[41] E. Knight, S. Przyborski, J. Anat. 2015, 227, 746. Immunol. 2014, 15, 846.
[42] M. E. Smithmyer, L. A. Sawicki, A. M. Kloxin, Biomater. Sci. 2014, [70] R. D. Abbott, D. L. Kaplan, Trends Biotechnol. 2015, 33, 401.
2, 634. [71] M. Brouwer, H. Zhou, N. N. Kasri, Stem Cell Rev. 2016, 12, 54.
[43] J. M. Souren, M. Ponec, R. van Wijk, In Vitro Cell Dev. Biol. 1989, [72] H. E. Abaci, Z. Guo, A. Coffman, B. Gillette, W. H. Lee, S. K. Sia,
25, 1039. A. M. Christiano, Adv. Healthcare Mater. 2016, 5, 1800.
[44] I. A. Schafer, A. Shapiro, M. Kovach, C. Lang, R. B. Fratianne, [73] S. Neuss, R. Stainforth, J. Salber, P. Schenck, M. Bovi, R. Knuchel,
Exp. Cell Res. 1989, 183, 112. A. Perez-Bouza, Cell Transplant. 2008, 17, 977.
[45] P. Martin, Science 1997, 276, 75. [74] C. K. Abrahamsson, F. Yang, H. Park, J. M. Brunger, P. K. Valonen,
[46] G. E. Davis, K. A. P. Allen, R. Salazar, S. A. Maxwell, J. Cell Sci. R. Langer, J. F. Welter, A. I. Caplan, F. Guilak, L. E. Freed, Tissue
2001, 114, 917. Eng., Part A 2010, 16, 3709.
[47] E. Garbuzenko, A. Nagler, D. Pickholtz, P. Gillery, R. Reich, [75] T. S. Lan, L. B. Wang, L. Xu, N. Jin, G. L. Yan, J. J. Xia, H. L. Wang,
F. X. Maquart, F. Levi-Schaffer, Clin. Exp. Allergy 2002, 32, 237. G. H. Zhuang, C. Gao, L. X. Meng, F. F. Du, Q. Zhou, Z. Q. Qi,
[48] A. Margulis, K. H. Nocka, N. L. Wood, S. F. Wolf, S. J. Goldman, Stem Cells Dev. 2016, 25, 462.
M. T. Kasaian, Am. J. Physiol. Lung Cell Mol. Physiol. 2009, 296, L236. [76] J. Buchrieser, W. James, M. D. Moore, Stem Cell Rep. 2017, 8, 334.
[49] D. Lindner, C. Zietsch, P. M. Becher, K. Schulze, H. P. Schultheiss, [77] R. Huggenberger, M. Detmar, J. Invest. Dermatol. Symp. Proc.
C. Tschope, D. Westermann, Biochem. Res. Int. 2012, 2012, 875742. 2011, 15, 24.
[50] A. A. Tandara, T. A. Mustoe, J. Plast. Reconstr. Aes. 2011, 64, 108. [78] F. S. Frueh, M. D. Menger, N. Lindenblatt, P. Giovanoli,
[51] P. J. Bergin, W. Sicheng, P. H. Qiang, Q. J. Marianne, FEMS M. W. Laschke, Crit. Rev. Biotechnol. 2017, 37, 613.
Immunol. Med. Microbiol. 2005, 45, 159. [79] R. F. Pereira, A. Sousa, C. C. Barrias, A. Bayat, P. L. Granja,
[52] T. L. Adair-Kirk, R. M. Senior, Int. J. Biochem. Cell Biol. 2008, 40, P. J. Bártolo, Biomanuf. Rev. 2017, 2, 1.
1101. [80] P. K. Wu, B. R. Ringeisen, Biofabrication 2010, 2, 014111.
[53] J. F. Almine, S. G. Wise, M. Hiob, N. K. Singh, K. K. Tiwari, S. Vali, [81] V. K. Lee, D. Y. Kim, H. Ngo, Y. Lee, L. Seo, S. S. Yoo, P. A. Vincent,
T. Abbasi, A. S. Weiss, FASEB J. 2013, 27, 3455. G. Dai, Biomaterials 2014, 35, 8092.
[54] R. Parenteau-Bareil, R. Gauvin, F. Berthod, Materials 2010, 3, [82] D. B. Kolesky, R. L. Truby, A. S. Gladman, T. A. Busbee,
1863. K. A. Homan, J. A. Lewis, Adv. Mater. 2014, 26, 3124.
[55] C. Lotz, F. F. Schmid, E. Oechsle, M. G. Monaghan, H. Walles, [83] M. Matsusaki, K. Fujimoto, Y. Shirakata, S. Hirakawa,
F. Groeber-Becker, ACS Appl. Mater. Interfaces 2017, 9, 20417. K. Hashimoto, M. Akashi, J. Biomed. Mater. Res., Part A 2015, 103,
[56] E. C. Chan, S. M. Kuo, A. M. Kong, W. A. Morrison, G. J. Dusting, 3386.
G. M. Mitchell, S. Y. Lim, G. S. Liu, PLoS One 2016, 11, e0149799. [84] J. Rosinczuk, J. Taradaj, R. Dymarek, M. Sopel, BioMed Res. Int.
[57] Y. Yu, A. Alkhawaji, Y. Ding, J. Mei, Oncotarget 2016, 7, 58671. 2016, 2016, 3943481.
[58] H. Debels, M. Hamdi, K. Abberton, W. Morrison, Plast. Reconstr. [85] Q. Ramadan, F. C. Ting, Lab Chip 2016, 16, 1899.
Surg. Glob. Open 2015, 3, e284. [86] J. A. Segre, J. Clin. Invest. 2006, 116, 1150.
[59] F. Groeber, L. Engelhardt, J. Lange, S. Kurdyn, F. F. Schmid, [87] M. Wittmann, D. McGonagle, T. Werfel, Cytokine Growth Factor Rev.
C. Rucker, S. Mielke, H. Walles, J. Hansmann, ALTEX: Altern. Anim. 2014, 25, 443.
Exp. 2016, 33, 415. [88] A. Fernandez, M. Vendrell, Chem. Soc. Rev. 2016, 45, 1182.

Adv. Healthcare Mater. 2018, 7, 1701405 1701405  (11 of 11) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like