Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Biomaterials 35 (2014) 4465e4476

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Review

Biomaterials in co-culture systems: Towards optimizing tissue


integration and cell signaling within scaffolds
Kyle G. Battiston a,1, Jane W.C. Cheung a,1, Devika Jain a, J. Paul Santerre a, b, *
a
Institute of Biomaterials and Biomedical Engineering, University of Toronto, 124 Edward Street, Room 461, Toronto, Ontario, Canada M5G 1G6
b
Department of Biomaterials, Faculty of Dentistry, University of Toronto, 124 Edward Street, Room 464D, Toronto, Ontario, Canada M5G 1G6

a r t i c l e i n f o a b s t r a c t

Article history: Most natural tissues consist of multi-cellular systems made up of two or more cell types. However, some
Received 4 January 2014 of these tissues may not regenerate themselves following tissue injury or disease without some form of
Accepted 12 February 2014 intervention, such as from the use of tissue engineered constructs. Recent studies have increasingly used
Available online 3 March 2014
co-cultures in tissue engineering applications as these systems better model the natural tissues, both
physically and biologically. This review aims to identify the challenges of using co-culture systems and to
Keywords:
highlight different approaches with respect to the use of biomaterials in the use of such systems. The
Co-culture
application of co-culture systems to stimulate a desired biological response and examples of studies
Biomaterials
Tissue engineering
within particular tissue engineering disciplines are summarized. A description of different analytical co-
Cellular interactions culture systems is also discussed and the role of biomaterials in the future of co-culture research are
Stem cells elaborated on. Understanding the complex cellecell and cellebiomaterial interactions involved in co-
Monocytes culture systems will ultimately lead the field towards biomaterial concepts and designs with specific
biochemical, electrical, and mechanical characteristics that are tailored towards the needs of distinct co-
culture systems.
Ó 2014 Elsevier Ltd. All rights reserved.

1. Introduction physical interactions [8]. Due to various physiological events such


as disease or injury, some natural tissues, like neural tissues, may
In the physiological environment, natural tissues most often not regenerate themselves or, in some cases, the body may attempt
consist of multi-cellular systems of two or more types of cells which to repair damage but the resulting tissues do not have the same
interact with one another to facilitate viability, proliferation [1,2], as properties and function of the original tissue (e.g. the generation of
well as generating growth factors and proteins [3,4], promoting the fibrotic tissues in the myocardium after an infarction [9]). In an
differentiation [5], and enabling other key biological tasks. For attempt to regenerate these damaged tissues and organs, tissue
example, the multi-cellular system of endothelial cells (ECs), engineering approaches have attracted great interest.
vascular smooth muscle cells (VSMCs), and fibroblasts make up the For a long time, many biomaterial studies have focused on single
blood vessels [6], and the organized structure of epithelial cells, cell studies with such constructs. However, recent directions in the
gingival fibroblasts, periodontal ligament cells, ECs, and osteoblasts field have increasingly used co-cultures in tissue engineering as
generate periodontal tissues [7]. In arteries, cellular cross-talk be- these systems better model the natural tissues, physically and
tween ECs and VSMCs regulates the proliferation of the VSMCs [2], biologically, through interactions between different cell types.
preventing the hyperproliferative VSMC phenotypic state that is Despite the increasing interest in using co-culture systems for tis-
characteristic of pathological conditions. In periodontal tissues, sue engineering purposes, challenges have been identified from
gingival fibroblasts regulate the migration of epithelial cells via these co-culture studies (Table 1), which place constraints on the
design of the experiment, and ultimately influence the effective
analysis of the experimental results. In particular, co-culture sys-
* Corresponding author. Department of Biomaterials, Faculty of Dentistry, Uni- tems involve multiple interactions (Fig. 1) that can be difficult to
versity of Toronto, 124 Edward Street, Room 464D, Toronto, Ontario, Canada M5G distinguish from one another without the use of proper experi-
1G6. Tel.: þ1 416 979 4903x4341; fax: þ1 416 979 4760.
mental design, which can lead to the misinterpretation of results.
E-mail addresses: paul.santerre@utoronto.ca, paul.santerre@dentistry.utoronto.
ca (J.P. Santerre). For example, a significant barrier to the effective analysis of co-
1
These authors contributed equally to this work. culture systems is being able to distinguish the contribution of the

http://dx.doi.org/10.1016/j.biomaterials.2014.02.023
0142-9612/Ó 2014 Elsevier Ltd. All rights reserved.
4466 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

Table 1
Challenges associated with co-culture experiments.

Issue Challenge References for relevant studies

Medium optimization If two cell types have different optimized growth media, for co-culture experiments [5,23,27,29,35,55,72,78,90,113]
which growth medium (or combination of growth media) should be used to
optimize growth and cell phenotype? Should growth supplements still be
added since cellecell contact between cell types may already promote
specific responses?
Cell type/origin The anatomical origin of the cells (ex. microvascular ECs, HUVECs, HAMECs, etc.) [1,3,4,9,23,25,27,31,34,46,57,
may influence the outcome of the experiment, and the appropriate cell type 59,71,87,92,98]
should be chosen for the organ system that the co-culture experiment
needs to address.
Cell ratio What cell ratio will lead to optimal experimental outcomes? [23,30,31,39,72,78,90,92,93,160]
Cell number If experiments are continued from a monoculture, does one maintain the [23,67,94,99,121,140]
same cell number used in the monoculture study for each cell type? Or is
an optimized cell ratio required? Should the total cell population vary?
Seeding protocol Should cells be seeded simultaneously or sequentially? If sequentially, which [2,14,20,28,32,36,38,55,67,70,
cell type should be seeded first? 78,88,108,138,139,160,161]
Static or dynamic Should additional mechanical stimulus be introduced in the culture? [75,81,96,108,129,162]
3D vs. 2D Will the cells in the co-culture system behave differently in a 2D vs. 3D setting? [30,54,77,82,107,108,128,155]
Location of cells within scaffold Is spatial patterning necessary? [28,36,75,77,89,100,101,104,106,
108,115,120e123,133,135e138,
140,149,161]
Purpose of the experiment One must consider if it is necessary to precisely mimic the physiological system [125e129,131,132,163]
in order to maximize specific experimental outcomes.
Optimization conditions When optimizing experimental conditions, it is important to choose the correct [25,33,36,38,56,67,85,93,99,104,
analysis parameters for the optimization (i.e. maximized cell growth, enhanced 105,114,124,134,136,139,
cellular differentiation, maximized phenotypic marker expression, maximized 140,147]
matrix synthesis, etc.).
Distinguishing cell types from one another Appropriate and selective phenotypic markers must be chosen to distinguish [85,93]
between the cell types.
Contribution of different cell types to If both cell types express the same phenotypic markers, co-staining for multiple [10,11]
proliferation or phenotypic markers may be a possible solution. Careful selection of stains (e.g. fluorescent labels)
marker expression and sample preparation are important. Vital dyes can be used to label different cell
types prior to seeding onto biomaterials and imaging or flow cytometry can be used
to determine spatial distribution of each cell as well as the increase in cell number
of each cell type individually.
Distinguishing paracrine vs. cellecell In typical co-culture systems, the effects mediated by cellecell contact vs. paracrine [1,21,25,31,35,69,71,72,105,
contact mediated effects signaling cannot easily be separated. Strategic experimental design is required to 116,121,122,135,136,146,
disentangle the two from one another to determine mechanistically what is 148,155]
happening within the co-culture system to properly inform future research.

different cell types to the measured responses. More specifically, importance in the outcome of the co-culture system, in addition to
typical assays used to assess proliferation or metabolic activity may the effects caused by the interaction of the two cell types.
only measure the response of the system as a whole, but will not Synthetic and natural biomaterials play a critical role in tissue
provide insight into which cell type in the system is proliferating and engineering as they can modulate cell response via different ma-
at what rate. There are a number of strategies that can be employed terial properties such as surface chemistry (i.e. presence of func-
to differentiate between cell types to address this issue. Perhaps one tional groups, hydrophobicity, ionic and hydrogen bonding, etc.),
of the most powerful involves the use of vital dyes [10,11], whereby topography, roughness, compliance, porosity, isotropy, etc [6]. Cell-
labeling each cell type allows the fate of each cell type to be tracked. biomaterial interactions also affect cellecell interactions in co-
It may also be necessary to perform immunostaining for cell type- culture systems, in that all cell types in the co-culture environ-
specific markers to determine if the initial seeding ratio has been ment will behave uniquely upon interaction with different bio-
maintained throughout the length of the experiment. materials, which may affect how one cell type interacts with the
Another factor that can be lost during data interpretation is the other. This can be advantageous in developing new approaches for
relative contributions of cellecell interactions versus cellebioma- promoting tissue regeneration.
terial interactions to the observed responses. This issue can This review aims to highlight the different approaches to co-
partially be addressed through the use of control monoculture culture systems with biomaterials, while identifying key studies
conditions, wherein the cell types present in the co-culture system that have been able to overcome some of the challenges associated
are cultured in parallel by themselves but otherwise under identical with such systems (Table 1). Specifically, the use of co-culture
conditions (medium type, biomechanical stimulation, culture systems to stimulate a desired biological response and examples
substrate, etc.) to the co-culture. This will aid the experimenter in of studies within particular tissue engineering disciplines will be
determining if the effects of the co-culture are altering the effects of summarized and key studies highlighted. Furthermore, the use of
the cells compared to how they are stimulated by the biomaterial different analytical systems to probe cellecell interactions in co-
substrate alone. However, this type of experimental design will not cultures, and their relative advantages and disadvantages will be
determine if cellecell communication or the stimulus imparted by discussed. Lastly, this review will report on the future promise of
the biomaterial is the dominant factor in the culture system. While strategically directed biomaterial development towards enhancing
the biomaterial substrate is always likely to play an important role, biological outcomes in co-culture systems and the use of bioma-
conducting the same co-culture on different biomaterials will help terial concepts to address some of the challenges inherent to this
determine if the stimulus provided by the biomaterial has potential field of work.
K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476 4467

Fig. 1. Different interactions that occur in co-culture systems. Cells have multiple ways of interacting with one another. It can be difficult in co-culture systems to distinguish how
the cells are communicating, and whether the effects observed are primarily due to cellecell communication, cellebiomaterial interactions, celleprotein interactions, or autocrine
effects, such as when a stimulatory molecule for one cell type can be produced by both cells.

2. Promotion of biological responses paracrine signals have been shown to be important for maintain-
ing EC phenotype in a hepatocellular model involving hepatocytes
Co-culture systems can be used with the intent of using one cell and fibroblasts [25], while other studies have also shown similar
type to provide a desired stimulus to a second cell type. This effects [26,27]. The hepatocellular model illustrates the use of a
strategy takes advantage of the naturally occurring cross-talk be- unique system to allow for the deconvolution of effects mediated
tween cell types, either through soluble factors or direct cellecell by cellecell contact vs. soluble signals [28]. This involves seeding
contact, which occurs in healthy tissues [12], regeneration [13], cells on separate silicone microchips which can then be brought
wound healing [14], and during developmental processes [15]. This into contact or kept at a distance (‘gap culture’, 80 mm separation) in
can often be a less cost-intensive strategy than dosing specific order to assess any effects mediated by cellecell contact or through
growth factors into culture medium to provide a similar effect. paracrine mechanisms. In other systems, however, the effects of
different cell types on ECs have been shown to be dependent on
2.1. Promotion of vasculogenesis and angiogenesis cellecell contact rather than through paracrine signaling [29e31].
It should also be noted that paracrine mechanisms have been
The long-term success of tissue engineering strategies both shown to be responsible for enhanced angiogenesis [19,25], while
in vitro and following implantation relies significantly on the ability reduced angiogenesis and lack of responsiveness to angiogenic
of cells to receive an adequate supply of nutrients, particularly for factors have been linked to cellecell contact [29e31]. Co-culture
large and deep tissues that are susceptible to diffusive limitations systems designed for promoting angiogenesis should therefore
[16]. Most tissues are highly vascularized (e.g. bone [17]) and thus look to provide an environment with appropriate cell distribution
need to be engineered with a vascular network in place to support and density to allow for the soluble signal-mediated pro-angio-
nutrient transport and to ease the connection with the host vascu- genic effects of co-culture, without the suppressive effects induced
lature upon implantation. Seeding endothelial cells (ECs) on by cellecell contact. This need for fine control of the cellecell ratios
biomaterial scaffolds for this purpose has not been an effective in co-cultures provides an opportunity to capitalize on the field’s
strategy due to the inability of ECs to survive and proliferate for long ability to conceive synthetic and natural biomaterial strategies that
culture times, as well as their inability, in monoculture, to self- can perturb cell proliferation, migration, and distribution by vary-
assemble and form tube-like structures [18]. However, a strategy ing the chemical, electrical, and mechanical properties, as well as
that has proven effective involves the co-culture of ECs with the interconnectivity, of biomaterial constructs.
different cell types, which provides stimulation through the release An example of in vitro vascularization providing benefits post-
of soluble factors (e.g. vascular endothelial growth factor (VEGF) and implantation was demonstrated by Guillemette et al., where a
platelet-derived growth factor (PDGF)), resulting in the self- fibroblast-cell sheet was seeded with ECs, resulting in the forma-
assembly of ECs into capillary-like structures in vitro [19,20]. Pri- tion of capillary-like structures after 7-days [32]. When the cell
marily, ECs have been successfully co-cultured with fibroblasts [3] sheet was then rolled up to a form a tissue-engineered vascular
and osteoblasts [21] to support cell proliferation and capillary for- construct, the result was a vascularized blood vessel that had
mation, though stem cells have shown promising results as well [22]. improved blood circulation and enhanced integration with the
Exogenous supplementation of growth factors to promote EC surrounding environment following implantation compared to
function in vitro often involves VEGF, basic fibroblast growth factor non-pre-seeded constructs [32]. The success of EC co-culture sys-
(bFGF), epithelial growth factor (EGF), insulin-like growth factor-1 tems, primarily with fibroblasts or osteoblasts, is an example of
(IGF-1), or some combination thereof [23]. The effects of fibro- how co-culture systems can be used to provide stimuli to one cell
blasts and osteoblasts supporting angiogenesis in co-culture sys- type through culture with a second cell type exhibiting a pheno-
tems with ECs have primarily been linked to paracrine signaling type conducive to supporting a particular biological outcome.
mechanisms [24]. Co-cultures of fibroblasts with ECs have been While this section has focused specifically on the role of cells
shown to stimulate VEGF release by fibroblasts, upregulate VEGF supporting angiogenesis when co-cultured with ECs, it should be
receptor 2 on ECs, and support endothelial nitric oxide synthase noted that ECs in these systems are also playing important roles in
and nitric oxide production from ECs [24]. Similarly, short-range regulating the phenotypic state of the other cells [33e36].
4468 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

2.2. Monocytes and co-culture systems differences of up to two orders of magnitude in the release of
specific proteins [48]. Proteins in human sera in some cases have
Monocytes are a key cellular component involved in the wound been shown to be more bioactive towards human cells than the
healing response to biomaterials following implantation [37]. bovine source of the same proteins [49].
Depending on their activation state, monocytes and their derived In addition to the use of immune cells ex vivo to investigate celle
macrophages will release a spectrum of pro-/anti-inflammatory biomaterial interactions, immune cell cultures can also be used to
and/or pro-/anti-wound healing growth factors and cytokines that understand cellular cross-talk between immune cells and other cell
will determine, to a significant extent, the ability of the material to types in different co-culture systems. The use of immune cells for
support tissue integration and regeneration. As a result, in vitro co- this purpose, however, should be viewed with some skepticism, as
cultures containing monocytes can be used as a strategy to promote the previously mentioned used of xenogeneic media can produce
other cell types to adopt a phenotypic state that is conducive to misleading results. Furthermore, the outcome of poly-
supporting tissue formation and regeneration, as well as cell morphonuclear leukocyte-monocyte co-cultures has been shown
growth or other properties depending on the application [38,39]. In to have a biomaterial dependence [50], such that any insight pro-
particular, there has been a recent focus in the biomaterials liter- vided by the in vitro culture system is relative to the culture sub-
ature leading to an understanding that monocyte/macrophage strate that was used, and does not necessarily provide insight with
dysfunction with respect to monocyte/macrophage activation in regards to the cellular cross-talk that will occur in other environ-
healthy tissue is associated with adverse outcomes, such as the case ments. Even the use of TCPS, the most common culture substrate
of obesity where macrophages in adipose tissue have been polar- for evaluating cellecell interactions, is known to vary depending on
ized to an M1 (pro-inflammatory) phenotype, compared to lean the manufacturer, with numerous reports demonstrating varied
individuals where macrophages in adipose tissue adopt a pre- cell responses, including its use with monocytes, to TCPS from
dominantly M2 (pro-wound healing, regulatory) phenotype [40]. different manufacturers [51e53]. Care must thus be taken when
Co-culture systems with monocytes/macrophages that are induced interpreting data from reports involving the use of immune cells on
into a pro-wound healing, anti-inflammatory state by biomaterials different biomaterials as well as in the presence of different media
have potential as strategies for tissue regeneration [41]. types, while also giving thought to the source of the monocytes and
Surface properties, such as topography and chemical function- the distribution of their phenotypes.
ality, have been shown to be important mediators of the monocytic
response to synthetic biomaterials [42e45]. The diverse phenotypic 2.3. Co-culture to induce stem cell growth and differentiation
range of monocytes/macrophages and the ability of biomaterials
with specific properties to elicit distinct activation states provides a Stem cell growth in vitro, in order to maintain the undifferen-
unique opportunity to develop biomaterial strategies to promote a tiated state of the stem cell, requires the presence of certain growth
monocyte/macrophage activation that can support a desired effect factors, cytokines, and nutrients. This is often accomplished with
in different co-culture systems. It has recently been shown that the use of feeder cells, typically fibroblasts, in a process where the
monocytes can promote the migration and growth of VSMCs when feeder cell layer is used as a substrate to grow the stem cells. The
they are co-cultured on a degradable polyurethane [38]. Further- effects of the feeder layer on stem cell survival and expansion can
more, monocytes seeded on an acrylamide-based hydrogel that be mediated through cellecell contact, feeder layer-assembled
were highly expressing CD163, an M2 marker of activation, were extracellular matrix (ECM), or various cytokines and growth fac-
shown to support human umbilical vein EC (HUVEC) proliferation tors [54]. In contrast to this more traditional use of co-culture to
through the release of VEGF, without releasing pro-inflammatory maintain an undifferentiated stem cell state, co-culture systems can
cytokines, compared to control monoculture conditions [46]. also be used with the goal of inducing stem cell differentiation.
Enhanced cell growth can accelerate the process of tissue formation Several examples include the co-culture of osteoblasts with
in vitro, which is essential before cell-seeded biomaterials are ready mesenchymal stem cells to promote osteoblastic differentiation
to be implanted. Furthermore, monocytes can non-invasively be [55], osteoblasts enhancing hematopoietic progenitor cell prolif-
isolated from a patient’s peripheral blood, making them an easily eration and function [56], ECs regulating cardiomyocyte develop-
accessible, autologous source of stimulatory cytokines and growth ment from embryonic stem cells (ESCs) [57], and the differentiation
factors. In comparison, the addition of exogenous growth factors of placenta-derived multipotent cells to neuronal and glial cells
such as VEGF, PDGF-BB, or EGF can quickly become expensive, under co-culture with rat brain cells [58].
particularly with long culture periods, which is further complicated Stem cell differentiation or self-renewal can be managed by
by the possibility of endotoxin contamination if the proteins are using the appropriate co-culture system. This has recently been
obtained from a recombinant source [47]. illustrated by Sneddon et al., where it was shown that the prolif-
Culturing monocytes and other cells of the inflammatory system eration and self-renewal of ESC-derived progenitors, without dif-
on biomaterials ex vivo is useful for providing initial insight into ferentiation, could be promoted by co-culture with organ-matched
how a new biomaterial is going to be seen by elements of the body’s mesenchyme [59]. This study also illustrates that not only is the cell
immune system, and whether an adverse foreign body reaction is type being co-cultured an important, but also that the source of this
to be expected or if beneficial wound healing and tissue regener- cell can have important implications. To promote differentiation,
ation could be anticipated. However, there are several issues ESCs have been co-cultured with ECs to promote the formation of
associated with the use of immune cells that can compromise the pancreatic insulin producing cells [60]. Co-cultures of ESCs with
integrity of the collected data. A large amount of studies reporting primary bone-derived cells on PLGA/HA composite scaffolds,
the use of immune cells in vitro rely on the use of fetal bovine serum however, has been shown to be efficient for osteogenic differenti-
or other xenogeneic protein sources to culture their cells. While ation [61].
traditional in vitro culture systems are already limited in terms of Within these co-culture systems there are two important factors
their predictive capacity for how biomaterialecell interactions will that can be used to favor a particular differentiated state, namely
occur in vivo, the use of xenogeneic media with immune cells in the cell type being used for the co-culture, as well as the bioma-
particular can give skewed results. It has recently been shown that terial substrate upon which the culture will be performed, where
culturing cells on the same biomaterial substrate, but in the pres- the different mechanical, electrical, chemical, and morphological
ence of human serum vs. xenogeneic serum, can result in properties of the substrate can play important roles. To optimize
K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476 4469

the different aspects of the co-culture system, including the cell followed by the seeding of ECs to generate a tissue-engineered
type and designed properties of the biomaterial, the in vivo differ- vascular graft (TEVG) [4]. Scaffold design plays an important role
entiation conditions under which stem cell fate is specified should in the generation of a TEVG. For example, the pore interconnectivity
be taken into consideration. For example, substrate elasticity has and the porosity of the scaffolds allow interactions between cell
been shown to favor ESC differentiation to different germ layers types [38,79] and uniform cell distribution [4]. The structure of the
[62]. In a study by Zoldan et al., 3D scaffolds were engineered to scaffold (i.e. layered and porous [75,80,81] is also important for
possess different stiffness thresholds which are similar to the mimicking the natural blood vessel.
different forces that cells would be exposed to during the process of Mechanical stimuli such as perfusion and cyclic strain have also
gastrulation, where it is known that mechanical forces are critical been added into co-culture systems in order to promote cell pro-
to embryogenesis [62,63]. liferation [75,76,80] and to modulate the phenotype of VSMCs
As discussed in Section 2.2, monocytes/macrophages can be a [75,76]. Work by Williams and Wick used a perfusion bioreactor
source of growth factors and cytokines, with the exact make-up of and co-cultures with ECs in order to promote cell proliferation,
their secretome dependent on their activation state [64]. These uniform cell distribution, and induce a more contractile phenotype
cells have recently been shown to provide directed differentiation in VSMCs [76]. The use of pulsatile flow with a silk fibroin scaffold
of stem and progenitor cells dependent on their culture environ- was also shown to allow for the formation of an EC layer on top of
ment and their particular phenotype. Harnessing the power of the VSMCs, while promoting the proliferation and alignment of
secreted products from a particular phenotype has also been VSMCs, retaining the contractile phenotype of the VSMCs, as well as
exploited for MSCs [65]. When using collagen-based materials for enhancing extracellular matrix production [75]. In order to further
cell entrapment, macrophages were shown to attenuate MSC dif- stimulate cell responses, additional cell types such as monocytes
ferentiation to chondrocytes, but enhance osteoblast differentia- have also been introduced into co-cultures with ECs and VSMCs to
tion [66]. The ability of macrophages to direct differentiation was modulate the cells’ phenotypes. Monocytes in the wound healing
shown to be material-dependent, with cultures contained within a state have been shown to induce a less contractile phenotype in
gelatin/PEG-based matrix promoting adipocyte differentiation [66]. VSMCs [79]. Also, polystyrene-activated monocytes and neutro-
Macrophage effects on MSCs has also shown a polarization de- phils (i.e. in a pro-inflammatory state) have been reported to
pendency, with M1 macrophages inhibiting growth and M2 mac- downregulate EC proliferation [4], indicating that a less pro-
rophages promoting growth [67]. In other studies, MSCs have been inflammatory monocyte phenotype may be beneficial in EC-
shown to be important mediators of macrophage polarization [68e VSMC co-culture systems.
70], such as promoting the regulatory, or M2, state [68,69]. In
addition to supporting adipocyte, osteoblast, and chondrocyte 3.1.2. Cardiac tissues
differentiation, macrophage co-culture has also been shown to After myocardial infarction, cardiac tissues become damaged
support MSC differentiation to SMCs when the macrophages have but they have difficulty to be regenerated by the body as the car-
been activated with IL-1b [71]. Macrophage effects on MSC differ- diomyocytes are terminally differentiated. Instead, fibrosis occurs
entiation have been shown to be mediated by both cellecell contact and results in the formation of a fibrotic scar and organ dysfunction.
[72] and the release of soluble factors in a paracrine manner [71,72], Cardiac tissue engineering has been focused on the use of car-
where effects in this particular study were determined to be cell diomyocytes to generate a cardiac patch that can be used to replace
contact dependent based on the use of a transwell system with a the damaged tissue; however, these cells could not remain viable in
0.4 mm semipermeable membrane [72]. long-term monoculture and sometimes could not contract syn-
chronously [9]. It has been shown that co-culturing cardiomyocytes
3. Rebuilding of natural tissues with cardiac fibroblasts on two-dimensional tissue culture plastic
and in three-dimensional collagen matrices promoted synchro-
Most co-culture systems are developed to repair, regenerate, or nized contraction in the cardiomyocytes with the presence of
mimic the natural tissues. These systems often involve two or more intercalated discs [82]. Recently, a similar co-culture model was
types of cells cultured in a biomaterial but vary in terms of the built on electrospun chitosan nanofiber scaffolds. In this study, the
sequence in which the cells are seeded, the spatial distribution of cardiomyocytes exhibited polarized morphology and their lifespan
different cell types, and the structure of the biomaterials. In this was prolonged by the presence of fibroblasts [9]. The co-culture of
section, examples of co-culture systems used with biomaterials in engineered cardiac progenitor cells with neonatal myocytes has
the regeneration of various types of tissues will be discussed. been shown to promote the commitment of the myocyte lineage in
the cardiac progenitor cells and the resulting construct had elec-
3.1. Cardiovascular tissue regeneration trophysiological properties that are similar to mature cardiac tis-
sues [83].
3.1.1. Vascular tissues
A good number of co-culture systems have been established for 3.2. Bone and cartilage tissue regeneration
vascular tissue regeneration with the goal of replacing damaged
vessels [73] and reconstructing the vessel environment [2,4,74e76]. 3.2.1. Bone tissues
Co-cultures of vascular ECs with VSMCs have exhibited a higher Bone is a calcified matrix consisting mainly of osteoblasts
survival rate for the vascular ECs [4,74e77] and promoted the residing in a highly vascularized network. In bone regeneration,
proliferation of VSMCs [2,77], indicating their potential to generate osteoblasts are often co-cultured with ECs to induce capillary for-
vessel-like structures. In most studies, ECs and VSMCs were seeded mation [19,21]. ECs, however, can similarly affect osteoblast
in a mixture, and the cells were allowed to rearrange themselves to phenotype and proliferation. Osteoblast-EC co-cultures have been
form two distinct layers of ECs and VSMCs via cellular crosstalk reported to upregulate the expression of osteogenic markers (e.g.
[77]. However, ECs may initially migrate into 3D scaffolds and, ALP) and to support osteoblastic proliferation of osteoprogenitors
instead of forming a monolayer, they may establish a capillary-like [33,84]. Similar co-cultures on silk fibroin scaffolds consisting of
network [78], which results in a different tissue structure from the porous hydroxyapatite, calcium phosphate and nickel-titanium
natural blood vessel. As such, sequential seeding was performed in showed that osteoblast-EC crosstalk prolonged the lifespan of ECs
which the VSMCs were seeded first to form a basement layer, and promoted the formation of microcapillary structures, while ECs
4470 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

in the monoculture began to die after one week of culture [85]. In exogenous fibroblast growth factor 9 was still required to inhibit
addition, fibroblasts have been reported to support osteoblast dif- mineralization in the engineered cartilage [95]. In addition, a me-
ferentiation and ectopic bone formation [86] and to support lacuna chanical stimulus (intermittent hydrostatic pressure) has been
structure by co-culture with chondrocytes on biodegradable PLGA introduced to the co-culture of chondrocytes and MSCs in alginate
(poly-lactic-glycolic acid) scaffolds [87]. beads, and at higher magnitudes, the intermittent hydrostatic
While co-cultures of osteoblasts with other cell types have pressure has been shown to enhance the proliferation and differ-
shown promising results in bone regeneration, the osteochondral entiation of MSCs into chondrocytes [96].
interface (i.e. cell interaction between the bone and cartilage) is not
well established. Studies of osteoblast-chondrocyte co-cultures are 3.3. Neural tissue regeneration
currently being assessed to regenerate the osteochondral interface
[88]. It has been shown that by co-culturing osteoblasts with Co-culture has also proven to be an important strategy towards
chondrocytes, ALP activity was maintained in osteoblasts, but achieving neural regeneration. PLGA multiple-channel conduits
glycosaminoglycan (GAG) content was decreased, along with have been used to co-culture Schwann cells and MSCs, which pro-
decreased mineralization in general. The mechanisms that resulted mote MSCs to differentiate into neuron-like cells as well as forma-
in the aforementioned cell behavior remain to be investigated. In tion of synapse-like structures [97]. Schwann cells have also been
addition, the spatial distribution of the two cell types is important co-cultured with adipose-derived stem cells (ASCs) in a transwell
in regulating cell behavior such as maintaining the chondrogenic system (Millicell) to promote Schwann cell-like differentiation [98].
phenotype in chondrocytes on one side of a bilayered agarose- Schwann cells co-cultured with neurons in a hydrogel network of
PDMS scaffold while promoting ossification on the other side [89]. collagen and hyaluronic acid also showed improved neurite growth
Various challenges in co-culture systems for bone regeneration [99]. Patterned biomaterials such as multi-channel-patterned PLG
should be addressed during experimental design and these include, (Poly-Lactic-Glycolic) have been used to direct neurite extension in
but are not limited to, the sequence of cell seeding as well as the cell co-cultures of primary neurons and accessory cells [100]. Other co-
seeding ratio (i.e. osteoblast:EC). For regenerating bone with a culture systems for neural regeneration have been developed such
microcapillary network, osteoblasts and ECs are often seeded as those used for encouraging the formation of neuromuscular
together [19,21,33,85], but for generating an osteochondral inter- synapses in vitro [101,102]. Intrafusal muscle fibers were co-cultured
face, sequential seeding should be considered [88]. Various seeding with type Ia sensory neurons on a surface-patterned PEG-silane
ratios have been explored in these co-culture systems. A recent substrate as well as MEMs device to study the conversion of me-
study has shown that the highest mineralization was observed for a chanical information from the intrafusal fibers to electrical action
50:50 ratio of osteoprogenitor cells and ECs [90]. The choice of potential in neuromuscular synapses. The resulting cell morphology
culture medium is also important in the co-cultures for bone and calcium ion response in the co-culture system were similar to
regeneration as the medium should (i) promote bone mineraliza- those observed in physiological synapses [101]. As neurons can be
tion and (ii) EC proliferation, along with subsequent capillary electrically stimulated, biomaterials such as hydrogels can be
formation. positively charged to stimulate neurite outgrowth [103]. It should
also be noted that different spacing widths between these micro-
3.2.2. Cartilage tissues channels/patterns can affect electrophysiological interactions be-
Similar to the cardiac tissues, articular cartilage tissue has tween neurons and other cell types.
limited regenerative capacity and the phenotype of chondrocytes, a
major cell type in articular cartilage, is difficult to maintain in a 3.4. Other tissue regeneration
tissue-engineered construct for long-term culture [91]. In cartilage
tissue engineering, chondrocytes have been co-cultured with 3.4.1. Periodontal tissues
various cell types for their phenotypic maintenance and to rebuild Periodontal tissue engineering involves the regeneration of the
cartilage-like tissue. For example, chondrocytes embedded in gingival tissue, the periodontal ligament and the alveolar bone [7].
agarose gel have been co-cultured with human MSCs in separate In order to reconstruct the damaged gingiva, gingival fibroblasts
bilayers (5  3 mm) such that the MSCs were physically separated have been co-cultured with gingival keratinocytes on poly-
from the chondrocytes. It was found that the MSCs provided dimethylsiloxane (PDMS) pillars to promote epithelial cell
paracrine signals to the chondrocytes, which resulted in chondro- morphogenesis via cellecell contact between the two cell types
genesis and promotion of type II collagen, a phenotypic marker of [8,104]. Co-cultures have also been used to promote osteogenesis
chondrocytes, with no mineralization in the chondrocyte layers and address the bone loss caused in destructive periodontal dis-
[89]. While this study used a 1:1 cell ratio, a similar study by eases. For example, periodontal ligament stem cells (PDLSCs) were
Meretoja et al. showed that a 1:3 ratio of chondrocytes:MSCs in co-cultured with human umbilical vein ECs in a transwell setting
porous polycaprolactone scaffolds (8 mm in diameter) had with polycarbonate membranes to induce osteogenesis in PDLSCs
enhanced cartilage-like matrix production [92]. There is no [105]. Furthermore, in order to prevent keratinocytes or gingival
consensus on the optimized ratio that enhances chondrocyte pro- fibroblasts from migrating into the periodontal ligament and over-
liferation and cartilaginous ECM production. Chondrocyte-MSC co- populating the periodontal defect, a method called guided tissue
culture has also regulated the hypertrophic behavior in MSCs, thus regeneration was used such that the biomaterial scaffold, typically
reducing mineralization in the engineered cartilage [93]. Other ePTFE (expanded polytratra-fluoro-ethylene) or PLGA, provided a
types of stem cells such as adipose-derived stromal cells have been barrier between the periodontal ligament cells and the gingival
co-cultured with chondrocytes in polyglycolic acid/polylactic acid keratinocytes and fibroblasts [106].
scaffolds. After eight weeks, the resulting tissue-engineered con-
structs were cartilage-like with rich type II collagen and GAG 3.4.2. Liver tissues
contents [94]. Furthermore, chondrocytes from different parts of The regeneration of liver tissues requires careful spatial distri-
the body have also been used to regenerate articular cartilage. bution of different cell types and the use of a 3D structure is
Costal chondrocytes were harvested and co-cultured with dental important for priming specific cell behavior as well as establishing
pulp stem cells at a ratio of 5:5 or 3:7 to enhance chondrogenesis. A cellular crosstalk between the cells of interest. Studies have been
ratio of 5:5 was chosen to minimize mineralization; however carried out to mimic liver tissue by co-culturing rat hepatocytes
K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476 4471

with rat stellate cells on a PLGA substrate [107]. Viable 3D spheroids seeded in the channels where the coating was not present. The
were formed at a faster rate in co-culture as compared to hepato- entire surface was then covered with chitosan and a layer of fibro-
cyte monoculture, and higher expression of hepatocyte growth blasts were seeded on top to investigate fibroblast-induced capillary
factor mRNA was observed in the co-culture, which indicates the formation in a well-oriented space [120]. Micropatterning can also
potential of this co-culture to regenerate liver tissue. Hepatocytes be used to control the area in which cellecell interaction can occur as
have also been co-cultured with ECs in PDMS honeycomb micro- well as the cell populations that are allowed to interact with one
wells to form endothelialized aggregates, which were then mixed another. Using micropatterning, fibroblasts were confined at the
with biodegradable poly-L-lactide fibers such that the aggregates periphery of hepatocyte islands and by controlling the area of pro-
were spaced out [108]. The hepatocyte-EC interaction in this spatial tein coating, the distance between these islands and fibroblast
orientation and the influence of the biomaterial had increased al- density changed. These arrangements were useful for investigating
bumin production and the aggregates remained spherical. fibroblastehepatocyte interactions with controllable parameters
[121]. Similarly, in a study by Lamponi et al., ECs and fibroblasts were
3.4.3. Skin tissues seeded in rectangles of various areas that were micropatterned on
Co-culture systems for skin regeneration were extensively silanized glass. By varying the size of the areas, the morphology of
developed in the 90’s and they predominately involved co-cultures fibroblasts changed and the number of fibroblasts allowed to
of fibroblasts and keratinocytes. Early co-cultures of keratinocytes interact with ECs was limited by the area of the rectangle [122].
and dermal fibroblasts on three-dimensional constructs showed Microfluidic systems can create a physiologically-relevant
success in reconstructing the epidermis [109e112]. For example, microenvironment that models the vasculature while reducing
after 5 weeks of in vitro culture on 3D nylon mesh, the co-culture of size and cost [123]. At this micro-scale, the spatial and temporal
human dermal fibroblasts with human keratinocytes showed aspect of the cell environment such as cell distribution and diffu-
epidermal differentiation; and the basal lamina, anchoring zone, sion rate can be better controlled to mimic three-dimensional tis-
and dermis were distinctly present with corresponding markers sue architecture and study cell behavior [124]. Microfluidic systems
[109,110]. This type of co-culture system has also been used to have often been used to study paracrine effects induced in the co-
reconstruct the stratified epidermis for cytotoxicity testing culture of ECs and various cell types on hydrogels [125,126]. For
[111,112]. Fibroblast-keratinocyte co-culture systems have been example, stromal cells embedded in fibrin gel channels promoted
shown to regulate collagen synthesis [113], induce myofibroblast capillary formation from the ECs cultured in the adjacent channel
differentiation [114], and to induce fibroblast migration [115]. [126]. Microfluidic devices have also been used to study how
In addition to fibroblast-keratinocyte co-culture, dermal different growth factors patterned on the microfluidic platform can
fibroblast-EC and fibroblast-stem cell co-culture systems have been affect ECs in co-cultures. Extracellular matrix proteins have also
used [116e118]. Studies on dermal fibroblast-EC co-culture systems been used to form microstructured platforms to spatially organize
could be dated as early as the 1990’s. In early studies of angio- breast cancer cells adjacent to ECs for investigating tumor angio-
genesis, dermal fibroblasts were co-cultured with human umbilical genesis [127].
vein ECs in vitro on 2D culture flasks to demonstrate the potential of
this co-culture system for capillary formation in skin regeneration 4.2. Tissue modeling
[117]. Vascularization was also observed in dermal fibroblast-EC co-
culture systems in 3D collagen scaffolds, and the resulting construct Co-cultures have also been used in tissue modeling for (i) drug
showed integration with the host vasculature 4 days after im- screening and (ii) studying various diseases such as cancer
plantation [118]. metastasis and respiratory diseases. In terms of drug screening, co-
Different biomaterials such as fibrin gels [115] and collagen cultures of cancer cells have been established for testing the effi-
matrices [119] have been used in co-culture systems for skin cacy of drugs. For example, lymphoma cancer cells were co-
regeneration. The cell behavior in co-culture systems is dependent cultured with neonatal stromal cells in a three-dimensional poly-
on the biomaterial used. For example, stratified layers of kerati- styrene scaffold to establish a blood cancer model for screening
nocytes were formed on the surface of both fibrin gels and collagen various commercially available drugs [128]. Also, a “lung-on-a-
matrices, along with fibroblast migration into the core of the chip” microfluidic system was created with lung alveolar epithelial
scaffolds. However, more fibroblast migration was observed in the cells and endothelial cells to study the drug treatment of respira-
collagen matrices in a shorter time frame than in the fibrin gels. In tory diseases [129]. Drug toxicity was also studied using a co-
addition, the use of different media may enhance or retard fibro- culture system [130], where hepatocytes were co-cultured with
blast migration [115]. other cell types such as ECs in the nylon scaffolds of a transwell
setting, thus creating a “three-dimensional liver” for testing the
4. Analytical systems for studying cell response in co-culture effect of different drugs in inducing hepatotoxicity [130].
Furthermore, tissue modeling with co-cultures has been used to
In addition to the use of co-culture systems for tissue regener- study various diseases. For example, a co-culture of alveolar
ation and stimulation of biological responses, these systems can epithelial cells, monocyte-derived macrophages, and dendritic cells
also be applied to study the fundamental aspects of cellecell was established in a transwell system to model the alveolar
interaction in specific tissues, developmental biology, as well as to epithelial barrier and study infectious disease [131]. In addition, a
examine cell responses to specific stimuli (tissue modeling). In this tissue model was built with breast cancer cells circulating through
section, the use of micropatterning and microfluidic systems with microfluidic channels coated with ECs to study adhesion of these
co-cultures used for tissue modeling will be discussed. cancer cells, angiogenesis, and how different chemokines can
stimulate cancer metastasis [132].
4.1. Micropatterning and microfluidic systems e vascular
microenvironment 5. The role of biomaterials in the future of co-culture
research
Micropatterning is often used to study the interactions between
two cell types in a co-culture. For example, gelatin films were coated Co-culture systems are often employed without much consid-
with bands of a cell-resistant polyelectrolyte coating and ECs were eration with regards to the potential influence of the substrate and
4472 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

its effect on experimental outcomes. In spite of this, it is well known with features on the order of 100 mm. A particularly powerful
that the biomaterial surface plays an important role influencing cell strategy involves the design of 3D matrices that allows for the
phenotype, proliferation, and other factors [45,133]. As an example, addition or removal of biological cues (ex. growth factors, extra-
the success of osteoblast:osteoclast co-culture systems has been cellular matrix ligands) in a bioorthogonal manner (i.e. not inter-
shown to be dependent on the biomaterial substrate (silk fibroin vs. fering with biochemical processes in a living system) while cells
chitosan vs. PLLA) [134]. In a study by Jones et al., both silk fibroin have already been growing on the biomaterial substrate (termed
and chitosan supported growth in osteoblast and osteoclast mono- ‘4D matrices’) [142]. This allows for the biomaterial scientist to
and co-cultures, whereas PLLA showed comparatively poorer temporally modulate the biological characteristics of the scaffold in
osteoclast differentiation in both mono- and co-cultures. Synthetic a controlled manner in order to facilitate a change in cell phenotype
biomaterials thus have the potential to significantly advance the when necessary.
field of co-culture research. By designing biomaterials with specific Though not as controlled as site-specific patterning techniques,
properties, the biomaterials can be used to impart one cell type control over a biomaterial scaffold’s porosity can also help to induce
with a desirable phenotype which in turn can influence the func- specific effects. Scaffold porosity has been shown to be important in
tional response of the second cell type. Biomaterials scientists have regulating cell migration [143], diffusion rate, cell differentiation
the ability to control numerous factors that can be used to optimize (pre-osteoblasts) [144], and cell distribution (fibroblasts) [144].
a biomaterial substrate for a particular co-culture system. These Using PLGA-CaP scaffolds, Sicchieri et al. suggested the importance
include surface chemistry, surface morphology, spatial patterning, of heterogenous pore size distribution for optimizing cell growth
porosity, the incorporation of cell-responsive peptides, surface and bone healing and demonstrated the importance of pore size in
wettability, electrical, and mechanical properties, amongst others. promoting bone and blood vessel formation [145]. Porosity is thus
The different properties can be varied to suit the specific needs of an important design strategy to consider, particularly for systems
different co-culture systems, depending on the aim of the experi- requiring the promotion of vascularization [146].
ments and the different cell types involved.
5.2. Biomaterial mechanical properties
5.1. Spatial patterning
The elastic modulus of biomaterials is particularly important for
The ability to pattern multiple cell types simultaneously by controlling stem cell differentiation. MSCs, for example, favor dif-
providing a biomaterial substrate conducive to supporting a ferentiation to the osteoblastic lineage when cultured on stiff
particular geometric configuration allows for the control of devel- substrates with modulus comparable to that of bone tissue [147].
oping tissue structure, as well as the ability to investigate cellecell Further control of a biomaterial’s mechanical properties, such as
interactions in a controlled manner. While typical co-culture sys- introducing mechanical gradients within a surface, has also been
tems involve two cell types, the use of superhydrophobic structures shown to have an effect on differentiation capacity, with narrow
(using photoinitiated grafting of 2,2,3,3,3-pentafluoropropyl mechanical gradients (70e90 kPa) inducing osteogenic differenti-
methacrylate) has recently been reported to have the ability to ation, whereas mechanical gradients with a broader range (10e
support patterns of up to twenty different cell types on one surface 90 kPa) showed protein-dependent effects on osteogenic differ-
[135]. Such systems may provide the necessary control for unrav- entiation, with inhibition observed on collagen vs. fibronectin
eling some of the more complex interactions involved in co-culture coated substrates [148]. These altered states of differentiation were
systems. Patterning has also been applied to three-dimensional shown to be due to changes in paracrine signaling mechanisms,
biomaterials through the use of photopatterned oligo(poly- which could take advantage of the upregulation of specific secreted
ethylene glycol)-fumarate:poly(ethylene glycol)-diacrylate hydro- cytokines from one cell type, which is dependent on changes in
gels [136] using photomasks containing polygonal features, which substrate properties, to elicit a desired response from another cell
can be tailored to the needs of specific experiments. Furthermore, type in co-culture.
substrates can be patterned to direct cell behavior, such as the use Mechanical cues have also recently been reported to produce
of patterned PLG substrates for directed neurite extension in co- self-patterning co-culture systems [149]. In a study by Palama et al.,
cultures of neurons and accessory cells [100], as well as the PDMS surfaces coated with laminin were prepared with areas
spatial control of neuron-glial cell co-cultures [137]. differing in stiffness. By creating stiff and soft regions on the PDMS
Another approach to spatial patterning involves the use of substrate, it was found that KU812 cells in the co-culture system
temperature-sensitive substrates. Patterning culture substrates preferred stiff microdomains, while 3T3 cells preferred soft regions.
with different thermo-responsive polymers has been used to pre- Patterning surfaces such as these with areas of varying stiffness
pare patterned cultures of ECs and fibroblasts, with such patterns could allow for the spatial control and segregation for the growth of
potentially providing insight into cellecell communication phe- different cell types, and provide useful design criteria when trying
nomena in co-culture systems [138]. In a study by Tsuda et al., to mimic tissue structures characterized by spatial patterning of
thermo-responsive micropatterned surfaces using polyacrylamide cells.
(PAAm) and thermally-responsive PIPAAm allow for the prepara-
tion of patterned endothelial cell sheets that can be layered with 5.3. Surface functionality
fibroblast sheets. Microwell systems have also proven useful for
ESC differentiation studies and could provide useful information if Changes in surface functionality can be achieved through two
applied to co-culture systems [139,140]. While the use of bioma- broad strategies: (1) the presentation of functional groups, with the
terial strategies to confine cells to particular geometries on two- goal of presenting an optimized mixture of functional character or
dimensional substrates provides significant promise in terms of surface wettability, and (2) the integration of cell-responsive pep-
disentangling the effects of cellular cross-talk occurring in co- tides and biochemical cues into the biomaterial to provide cell-
culture systems, three-dimensional systems will be necessary for specific effects, such as the use of REDV peptides to target endo-
recapitulating important structural features for tissue engineering thelial cells [150]. General trends have been reported in the bio-
purposes. materials literature suggesting that certain surface chemistries
Spatial patterning can also be achieved by patterning proteins [133,151e153] or a specific range in surface wettability, are more
that are cell-type specific [141]. Such patterns have been produced supportive of cell growth and attachment [133,151,154]. However,
K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476 4473

the literature is far from conclusive on specific chemical function- this reason it is important that biomaterials be considered as a
alities and their ratios that are necessary for inducing targeted more integral element in the experimental design of co-culture
biological responses from multiple cell types simultaneously. The systems, rather than the focus solely centred around the
defined effects of certain functional groups are likely also influ- biochemical reactions of the cellular components.
enced by the combination of other factors, such as material me-
chanical properties. There is not likely a generalization that can be
Acknowledgments
applied to a single strategy for all types of synthetic and natural
biomaterials. A more strategic approach involves the use of specific
This review was supported from funding by the Natural Sciences
biochemical functionalities being incorporated into a biomaterial’s
and Engineering Research Council (NSERC) Discovery (360520),
chemical architecture [155,156]. Such approaches have been sug-
NSERC Alexander Graham Bell Canada Graduate Scholarship (CGS
gested to be critical to the development of biomaterials for the
D3), Canadian Institutes of Health Research (CIHR) Grant (230762),
targeted differentiation of stem cells [156], and are a strategy that
Ontario Graduate Scholarship Program, and CIHR-Cell Signals
will be key in developing biomaterials for co-culture systems.
Training Fellowship program (TGF-53877).
Specific biochemical ligands can be chosen that have known
cell-specific effects. Such a strategy has been employed by Aizawa
and Shoichet to elucidate the role of ECs in governing retinal References
stem and progenitor cell (RPSC) fate [155]. In the latter study, 3D
agarose-sulfide hydrogels were modified to contain a uniform [1] Schubert SY, Benarroch A, Ostvang J, Edelman ER. Regulation of endothelial
cell proliferation by primary monocytes. Arterioscler Thromb Vasc Biol
distribution of RGD and a concentration gradient of immobilized 2008;28:97e104.
VEGF. These hydrogels supported EC tubular-like morphologies, [2] Nam M-H, Lee H-S, Seomun Y, Lee Y, Lee K-W. Monocyte-endothelium-
while also demonstrating that ECs inhibit RPSC proliferation and smooth muscle cell interaction in co-culture: proliferation and cytokine
productions in response to advanced glycation end products. Biochim Bio-
differentiation [155]. Similar targeting strategies have also been phys Acta Gen Subj 2011;1810:907e12.
employed to guide ECs in 3D scaffolds [157]. By providing such [3] Sorrell JM, Baber MA, Caplan AI. A self-assembled fibroblast-endothelial cell
guidance, EC distribution within the 3D scaffold can potentially co-culture system that supports in vitro vasculogenesis by both human
umbilical vein endothelial cells and human dermal microvascular endothe-
be controlled, allowing the scaffold to provide spatial patterning lial cells. Cells Tissues Organs 2007;186:157e68.
of the ECs, while a second cell type, such as fibroblasts, can be [4] Rose SL, Babensee JE. Smooth muscle cell phenotype alters cocultured
used to promote their survival or particular functional character endothelial cell response to biomaterial-pretreated leukocytes. J Biomed
Mater Res A 2008;84:661e71.
[3,146].
[5] Traphagen SB, Titushkin I, Sun S, Wary KK, Cho M. Endothelial invasive
response in a co-culture model with physically-induced osteodifferentiation.
5.4. 2D vs. 3D culture systems J Tissue Eng Regen Med 2013;7:621e30.
[6] Ratner BD, Hoffman AS, Schoen FJ, Lemons JE, editors. Biomaterials science.
1st ed. Toronto: Academic Press; 1996.
When trying to recapitulate cellular cross-talk that occurs in vivo [7] Schroeder HE. The periodontium. Germany: Springer-Verlag Berlin Heidel-
with an in vitro culture system, there are numerous issues that need berg; 1986.
to be taken into consideration. Many co-culture systems are con- [8] Müssig E, Tomakidi P, Steinberg T. Gingival fibroblasts established on
microstructured model surfaces: their influence on epithelial morphogenesis
ducted under 2D conditions, while tissues are inherently 3D and and other tissue-specific cell functions in a co-cultured epithelium: a model.
contain several different topographical cues that cells can sense and J Orofac Orthop 2009;70:351e62.
respond to [158]. As a result, when primary cells are isolated and [9] Hussain A, Collins G, Yip D, Cho CH. Functional 3-D cardiac co-culture model
using bioactive chitosan nanofiber scaffolds. Biotechnol Bioeng 2013;110:
plated onto 2D substrates there is generally a loss of mature 637e47.
phenotypic character and certain functionality, such as with he- [10] Wein F, Bruinink A. Human triple cell co-culture for evaluation of bone
patocytes [36] and SMCs [159], which makes it difficult to mimic implant materials. Integr Biol (Camb) 2013;5:703e11.
[11] Burguera EF, Bitar M, Bruinink A. Novel in vitro co-culture methodology to
the response of the cells in vivo. This loss of function provides an
investigate heterotypic cell-cell interactions. Eur Cell Mater 2010;19:
opportunity in the context of biomaterial and co-culture strategies 166e79.
to maintain cell function and phenotypic character, as has been [12] Nakahama K. Cellular communications in bone homeostasis and repair. Cell
Mol Life Sci 2010;67:4001e9.
achieved by some in with primary hepatocytes, a cell type that is
[13] Arai K, Jin G, Navaratna D, Lo EH. Brain angiogenesis in developmental and
notoriously difficult to maintain in culture [28,107,130]. The use of pathological processes: neurovascular injury and angiogenic recovery after
3D culture systems in combination with an optimization of the stroke. FEBS J 2009;276:4644e52.
aforementioned biomaterial properties (surface functionality, me- [14] Anderson JM, McNally AK. Biocompatibility of implants: lymphocyte/
macrophage interactions. Semin Immunopathol 2011;33:221e33.
chanical properties, spatial patterning) will be important in the [15] Lee HS, Han J, Bai H, Kim K. Brain angiogenesis in developmental and
next generation of biomaterials, whether they are developed to pathological processes: regulation, molecular and cellular communication at
mimic natural tissue structure or promote the growth and devel- the neurovascular interface. FEBS J 2009;276:4622e35.
[16] Novosel EC, Kleinhans C, Kluger PJ. Vascularization is the key challenge in
opment of new tissues in the lab. tissue engineering. Adv Drug Deliv Rev 2011;63:300e11.
[17] Das A, Botchwey E. Evaluation of angiogenesis and osteogenesis. Tissue Eng
6. Summary Part B Rev 2011;17:403e14.
[18] Janvier R, Sourla A, Koutsilieris M, Doillon C. Stromal fibroblasts are required
for PC-3 human prostate cancer cells to produce capillary-like formation of
Some of the issues facing co-culture experiments can be endothelial cells in a three-dimensional co-culture system. Anticancer Res
addressed by including appropriate controls, while others require 1997;17:1551e7.
[19] Ghanaati S, Fuchs S, Webber MJ, Orth C, Barbeck M, Gomes ME, et al. Rapid
proper experimental design and innovative methodologies for vascularization of starch-poly(caprolactone) in vivo by outgrowth endothe-
retrieving maximum information from the co-culture systems lial cells in co-culture with primary osteoblasts. J Tissue Eng Regen Med
(Table 1). In all cases, however, it must be understood that while a 2011;5:E136e43.
[20] Dohle E, Fuchs S, Kolbe M, Hofmann A, Schmidt H, Kirkpatrick CJ. Compar-
co-culture system can be optimized by varying seeding density, cell
ative study assessing effects of sonic hedgehog and VEGF in a human co-
type, and different media, the optimized culture conditions will be culture model for bone vascularisation strategies. Eur Cell Mater 2011;21:
specific to the biomaterial system which the cultures will be con- 144e56.
ducted on. Different substrates will elicit different functional re- [21] Santos MI, Unger RE, Sousa RA, Reis RL, Kirkpatrick CJ. Crosstalk between
osteoblasts and endothelial cells co-cultured on a polycaprolactone-starch
sponses from cells which may alter the balance that was achieved scaffold and the in vitro development of vascularization. Biomaterials
when the system was initially optimized on a different material. For 2009;30:4407e15.
4474 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

[22] Luo JZQ, Xiong F, Al-Homsi AS, Roy T, Luo LG. Human BM stem cells initiate [48] Schmidt D, Joyce EJ, Kao WJ. Fetal bovine serum xenoproteins modulate
angiogenesis in human islets in vitro. Bone Marrow Transplant 2011;46: human monocyte adhesion and protein release on biomaterials in vitro. Acta
1128e37. Biomater 2011;7:515e25.
[23] Eckermann CW, Lehle K, Schmid SA, Wheatley DN, Kunz-Schughart LA. [49] Meszaros K, Aberle S, White M, Parent J. Immunoreactivity and bioactivity of
Characterization and modulation of fibroblast/endothelial cell co-cultures for lipopolysaccharide-binding protein in normal and heat-inactivated sera.
the in vitro preformation of three-dimensional tubular networks. Cell Biol Int Infect Immun 1995;63:363e5.
2011;35:1097e110. [50] Cohen HC, Joyce EJ, Kao WJ. Biomaterials selectively modulate interactions
[24] Li H, Chang J. Bioactive silicate materials stimulate angiogenesis in fibroblast between human blood-derived polymorphonuclear leukocytes and mono-
and endothelial cell co-culture system through paracrine effect. Acta Bio- cytes. Am J Pathol 2013;182:2180e90.
mater 2013;9:6981e91. [51] Clinchy B, Youssefi M, Hakansson L. Differences in adsorption of serum proteins
[25] March S, Hui EE, Underhill GH, Khetani S, Bhatia SN. Microenvironmental and production of IL-1ra by human monocytes incubated in different tissue
regulation of the sinusoidal endothelial cell phenotype in vitro. Hepatology culture microtiter plates. J Immunol Methods 2003;282:53e61.
2009;50:920e8. [52] Battiston KG, McBane JE, Labow RS, Santerre JP. Differences in protein
[26] Clarkin CE, Emery RJ, Pitsillides AA, Wheeler-Jones CPD. Evaluation of VEGF- binding and cytokine release from monocytes on commercially sourced
mediated signaling in primary human cells reveals a paracrine action for tissue culture polystyrene. Acta Biomater 2012;8:89e98.
VEGF in osteoblast-mediated crosstalk to endothelial cells. J Cell Physiol [53] Zeiger AS, Hinton B, Van Vliet KJ. Why the dish makes a difference: quan-
2008;214:537e44. titative comparison of polystyrene culture surfaces. Acta Biomater 2013;9:
[27] Grellier M, Ferreira-Tojais N, Bourget C, Bareille R, Guillemot F, Amedee J. Role 7354e61.
of vascular endothelial growth factor in the communication between human [54] Khademhosseini A, Ferreira L, Blumling III James, Yeh J, Karp JM, Fukuda J,
osteoprogenitors and endothelial cells. J Cell Biochem 2009;106:390e8. et al. Co-culture of human embryonic stem cells with murine embryonic
[28] Hui EE, Bhatia SN. Micromechanical control of cell-cell interactions. Proc Natl fibroblasts on microwell-patterned substrates. Biomaterials 2006;27:
Acad Sci U S A 2007;104:5722e6. 5968e77.
[29] Wenger A, Kowalewski N, Stahl A, Mehlhorn A, Schmal H, Stark G, et al. [55] Sinclair SSK, Burg KJL. Effect of osteoclast co-culture on the differentiation of
Development and characterization of a spheroidal coculture model of human mesenchymal stem cells grown on bone graft granules. J Biomater Sci
endothelial cells and fibroblasts for improving angiogenesis in tissue engi- Polym Ed 2011;22:789e808.
neering. Cells Tissues Organs 2005;181:80e8. [56] Chitteti BR, Cheng Y, Streicher DA, Rodriguez-Rodriguez S, Carlesso N,
[30] Korff T, Kimmina S, Martiny-Baron G, Augustin H. Blood vessel maturation in Srour EF, et al. Osteoblast lineage cells expressing high levels of Runx2
a 3-dimensional spheroidal coculture model: direct contact with smooth enhance hematopoietic progenitor cell proliferation and function. J Cell
muscle cells regulates endothelial cell quiescence and abrogates VEGF Biochem 2010;111:284e94.
responsiveness. FASEB J 2001;15:447e57. [57] Chen K, Bai H, Arzigian M, Gao Y, Bao J, Wu W, et al. Endothelial cells regulate
[31] Nehls V, Herrmann R, Huhnken M, Palmetshofer A. Contact-dependent in- cardiomyocyte development from embryonic stem cells. J Cell Biochem
hibition of angiogenesis by cardiac fibroblasts in three-dimensional fibrin 2010;111:29e39.
gels in vitro: implications for microvascular network remodeling and coro- [58] Yen BL, Chien C, Chen Y, Chen J, Huang J, Lee F, et al. Placenta-derived
nary collateral formation. Cell Tissue Res 1998;293:479e88. multipotent cells differentiate into neuronal and glial cells in vitro. Tissue
[32] Guillemette MD, Gauvin R, Perron C, Labbe R, Germain L, Auger FA. Tissue- Eng Part A 2008;14:9e17.
engineered vascular adventitia with vasa vasorum improves graft integra- [59] Sneddon JB, Borowiak M, Melton DA. Self-renewal of embryonic-stem-cell-
tion and vascularization through inosculation. Tissue Eng Part A 2010;16: derived progenitors by organ-matched mesenchyme. Nature 2012;491:
2617e26. 765e8.
[33] Guillotin B, Bareille R, Bourget C, Bordenave L, Amedee J. Interaction between [60] Talavera-Adame D, Wu G, He Y, Ng TT, Gupta A, Kurtovic S, et al. Endothelial
human umbilical vein endothelial cells and human osteoprogenitors triggers cells in co-culture enhance embryonic stem cell differentiation to pancreatic
pleiotropic effect that may support osteoblastic, function. Bone 2008;42: progenitors and insulin-producing cells through BMP signaling. Stem Cell
1080e91. Rev 2011;7:532e43.
[34] Guillotin B, Bourget C, Remy-Zolgadri M, Bareille R, Fernandez P, Conrad V, [61] Kim S, Kim S, Lee S, Ahn SE, Gwak S, Song J, et al. In vivo bone formation from
et al. Human primary endothelial cells stimulate human osteoprogenitor cell human embryonic stem cell-derived osteogenic cells in poly(D,L-lactic-co-
differentiation. Cell Physiol Biochem 2004;14:325e32. glycolic acid)/hydroxyapatite composite scaffolds. Biomaterials 2008;29:
[35] Steiner D, Lampert F, Stark GB, Finkenzeller G. Effects of endothelial cells on 1043e53.
proliferation and survival of human mesenchymal stem cells and primary [62] Zoldan J, Karagiannis ED, Lee CY, Anderson DG, Langer R, Levenberg S. The
osteoblasts. J Orthop Res 2012;30:1682e9. influence of scaffold elasticity on germ layer specification of human em-
[36] Kim K, Ohashi K, Utoh R, Kano K, Okano T. Preserved liver-specific functions bryonic stem cells. Biomaterials 2011;32:9612e21.
of hepatocytes in 3D co-culture with endothelial cell sheets. Biomaterials [63] Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, and forces
2012;33:1406e13. combine and control stem cells. Science 2009;324:1673e7.
[37] Anderson JM, Rodriguez A, Chang DT. Foreign body reaction to biomaterials. [64] Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activa-
Semin Immunol 2008;20:86e100. tion. Nat Rev Immunol 2010;10:460.
[38] McBane JE, Cai K, Labow RS, Santerre JP. Co-culturing monocytes with [65] Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal
smooth muscle cells improves cell distribution within a degradable poly- stem cell secretome for the treatment of cardiovascular disease. Cell Stem
urethane scaffold and reduces inflammatory cytokines. Acta Biomater Cell 2012;10:244e58.
2012;8:488e501. [66] Cantu DA, Hematti P, Kao WJ. Cell encapsulating biomaterial regulates
[39] McBane JE, Battiston KG, Wadhwani A, Sharifpoor S, Labow RS, Santerre JP. mesenchymal stromal/stem cell differentiation and macrophage immuno-
The effect of degradable polymer surfaces on co-cultures of monocytes and phenotype. Stem Cells Transl Med 2012;1:740e9.
smooth muscle cells. Biomaterials 2011;32:3584e95. [67] Freytes DO, Kang JW, Marcos-Campos I, Vunjak-Novakovic G. Macrophages
[40] Brown BN, Ratner BD, Goodman SB, Amar S, Badylak SF. Macrophage po- modulate the viability and growth of human mesenchymal stem cells. J Cell
larization: an opportunity for improved outcomes in and regenerative Biochem 2013;114:220e9.
medicine. Biomaterials 2012;33:3792e802. [68] Zhang Q, Su W, Shi S, Wilder-Smith P, Xiang AP, Wong A, et al. Human
[41] Brown BN, Londono R, Tottey S, Zhang L, Kukla KA, Wolf MT, et al. Macro- gingiva-derived mesenchymal stem cells elicit polarization of M2 mac-
phage phenotype as a predictor of constructive remodeling following the rophages and enhance cutaneous wound healing. Stem Cells 2010;28:
implantation of biologically derived surgical mesh materials. Acta Biomater 1856e68.
2012;8:978e87. [69] Maggini J, Mirkin G, Bognanni I, Holmberg J, Piazzon IM, Nepomnaschy I,
[42] MacEwan MR, Brodbeck WG, Matsuda T, Anderson JM. Monocyte/lympho- et al. Mouse bone marrow-derived mesenchymal stromal cells turn activated
cyte interactions and the foreign body response: in vitro effects of bioma- macrophages into a regulatory-like profile. PLoS One 2010;5:e9252.
terial surface chemistry. J Biomed Res A 2005;74A:285e93. [70] Hanson SE, King SN, Kim J, Chen X, Thibeault SL, Hematti P. The effect of
[43] Brodbeck WG, Nakayama Y, Matsuda T, Colton E, Ziats NP, Anderson JM. mesenchymal stromal cell-hyaluronic acid hydrogel constructs on immu-
Biomaterial surface chemistry dictates adherent monocyte/macrophage nophenotype of macrophages. Tissue Eng Part A 2011;17:2463e71.
cytokine expression in vitro. Cytokine 2002;18:311e9. [71] Lee MJ, Kim MY, Heo SC, Kwon YW, Kim YM, Do EK, et al. Macrophages
[44] Schutte RJ, Parisi-Amon A, Reichert WM. Cytokine profiling using mono- regulate smooth muscle differentiation of mesenchymal stem cells via a
cytes/macrophages cultured on common biomaterials with a range of sur- prostaglandin F-2 alpha-mediated paracrine mechanism. Arterioscler
face chemistries. J Biomed Res A 2009;88A:128e39. Thromb Vasc Biol 2012;32:2733e40.
[45] Bota PCS, Collie AMB, Puolakkainen P, Vernon RB, Sage EH, Ratner BD, et al. [72] Nicolaidou V, Wong MM, Redpath AN, Ersek A, Baban DF, Williams LM, et al.
Biomaterial topography alters healing in vivo and monocyte/macrophage Monocytes induce STAT3 activation in human mesenchymal stem cells to
activation in vitro. J Biomed Res A 2010;95A:649e57. promote osteoblast formation. PLoS One 2012;7:e39871.
[46] Mayer A, Hiebl B, Lendlein A, Jung F. Support of HUVEC proliferation by pro- [73] Guyton AC, Hall JE, editors. Textbook of medical physiology. 11th ed. Phila-
angiogenic intermediate CD163(þ) monocytes/macrophages: a co-culture delphia: Elsevier; 2006.
experiment. Clin Hemorheol Microcirc 2011;49:423e30. [74] Wen S-J, Zhao L-M, Wang S-G, Li J-X, Chen H-Y, Liu J-L, et al. Human vascular
[47] Wakelin SJ, Sabroe I, Gregory CD, Poxton IR, Forsythe JLR, Garden OJ, et al. smooth muscle cells and endothelial cells cocultured on polyglycolic acid
“Dirty little secrets” e endotoxin contamination of recombinant proteins. (70/30) scaffold in tissue engineered vascular graft. Chin Med J 2007;120:
Immunol Lett 2006;106:1e7. 1331e5.
K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476 4475

[75] Zhang X, Wang X, Keshav V, Wang X, Johanas JT, Leisk GG, et al. Dynamic [101] Rumsey JW, Das M, Bhalkikar A, Stancescu M, Hickman JJ. Tissue engineering
culture conditions to generate silk-based tissue-engineered vascular grafts. the mechanosensory circuit of the stretch reflex arc: sensory neuron
Biomaterials 2009;30:3213e23. innervation of intrafusal muscle fibers. Biomaterials 2010;31:8218e27.
[76] Williams C, Wick TM. Endothelial cell-smooth muscle cell co-culture in a [102] Umbach JA, Adams KL, Gundersen CB, Novitch BG. Functional neuromuscular
perfusion bioreactor system. Ann Biomed Eng 2005;33:920e8. junctions formed by embryonic stem cell-derived motor neurons. PLoS One
[77] Xu T, Zhao W, Zhu J-M, Albanna MZ, Yoo JJ, Atala A. Complex heterogeneous 2012;7:e36049.
tissue constructs containing multiple cell types prepared by inkjet printing [103] Dadsetan M, Knight AM, Lu L, Windebank AJ, Yaszemski MJ. Stimulation of
technology. Biomaterials 2013;34:130e9. neurite outgrowth using positively charged hydrogels. Biomaterials
[78] Kunz-Schughart LA, Schroeder JA, Wondrak M, Van Rey F, Lehle K, 2009;30:3874e81.
Hofstaedter F, et al. Potential of fibroblasts to regulate the formation of [104] Mussig E, Steinberg T, Schulz S, Spatz JP, Ulmer J, Grabe N, et al. Connective-
three-dimensional vessel-like structures from endothelial cells in vitro. Am J tissue fibroblasts established on micropillar interfaces are pivotal for
Physiol Cell Physiol 2006;290:C1385e98. epithelial-tissue morphogenesis. Adv Funct Mater 2008;18:2919e29.
[79] Song Y, Feijen J, Grijpma DW, Poot AA. Tissue engineering of small- [105] Wu Y, Cao H, Yang Y, Zhou Y, Gu Y, Zhao X, et al. Effects of vascular
diameter vascular grafts: a literature review. Clin Hemorheol Microcirc endothelial cells on osteogenic differentiation of noncontact co-cultured
2011;49:357e74. periodontal ligament stem cells under hypoxia. J Periodontal Res 2013;48:
[80] Hoerstrup SP, Zünd G, Sodian R, Schnell AM, Grünenfelder J, Turina MI. 52e65.
Tissue engineering of small caliber vascular grafts. Eur J Cardio-thorac Surg [106] Taba Jr M, Jin Q, Sugai JV, Giannobile WV. Current concepts in periodontal
2001;20:164e9. bioengineering. Orthod Craniofac Res 2005;8:292e302.
[81] Niklason LE, Abbott W, Gao J, Klagges B, Hirschi KK, Ulubayram K, et al. [107] Thomas RJ, Bhandari R, Barrett DA, Bennett AJ, Fry JR, Powe D, et al. The
Morphologic and mechanical characteristics of engineered bovine arteries. effect of three-dimensional co-culture of hepatocytes and hepatic stellate
J Vasc Surg 2001;33:628e38. cells on key hepatocyte functions in vitro. Cells Tissues Organs 2005;181:
[82] Van Luyn MJA, Tio RA, Gallego Y, Van Seijen XJ, Plantinga JA, De Leij LFMH, 67e79.
et al. Cardiac tissue engineering: characteristics of in unison contracting two- [108] Pang Y, Montagne K, Shinohara M, Komori K, Sakai Y. Liver tissue engi-
and three-dimensional neonatal rat ventricle cell (co)-cultures. Biomaterials neering based on aggregate assembly: efficient formation of endothelialized
2002;23:4793e801. rat hepatocyte aggregates and their immobilization with biodegradable fi-
[83] Tufan H, Zhang X-H, Haghshenas N, Sussman MA, Cleemann L, Morad M. bres. Biofabrication 2012;4.
Cardiac progenitor cells engineered with Pim-1 (CPCeP) develop cardiac [109] Fleischmajer R, MacDonald II ED, Contard P, Perlish JS. Immunochemistry of
phenotypic electrophysiological properties as they are co-cultured with a keratinocyte-fibroblast co-culture model for reconstruction of human skin.
neonatal myocytes. J Mol Cell Cardiol 2012;53:695e706. J Histochem Cytochem 1993;41:1359e66.
[84] Ohno M, Cooke JP, Dzau VJ, Gibbons GH. Fluid shear stress induces endo- [110] Contard P, Bartel RL, Jacobs II L, Perlish JS, MacDonald II ED, Handler L, et al.
thelial transforming growth factor beta-1 transcription and production. Culturing keratinocytes and fibroblasts in a three-dimensional mesh results
Modulation by potassium channel blockade. J Clin Invest 1995;95:1363e9. in epidermal differentiation and formation of a basal lamina-anchoring zone.
[85] Unger RE, Sartoris A, Peters K, Motta A, Migliaresi C, Kunkel M, et al. Tissue- J Invest Dermatol 1993;100:35e9.
like self-assembly in cocultures of endothelial cells and osteoblasts and the [111] Gay R, Swiderek M, Nelson D, Ernesti A. The living skin equivalent as a model
formation of microcapillary-like structures on three-dimensional porous in vitro for ranking the toxic potential of dermal irritants. Toxicol Vitro
biomaterials. Biomaterials 2007;28:3965e76. 1992;6:303e15.
[86] Kinto N, Iwamoto M, Enomoto-Iwamoto M, Noji S, Ohuchi H, Yoshioka H, [112] Donnelly T, Decker D, Stemp M, Rheins L, Logemann P. A three-dimensional
et al. Fibroblasts expressing sonic hedgehog induce osteoblast differentiation in vitro model for the study of ocular cytotoxicity and irritancy. Toxicol Vitro
and ectopic bone formation. FEBS Lett 1997;404:319e23. 1994;8:631e3.
[87] Liu X, Zhou G, Liu W, Zhang W, Cui L, Cao Y. In vitro formation of lacuna [113] Tandara AA, Kloeters O, Mogford JE, Mustoe TA. Hydrated keratinocytes
structure by human dermal fibroblasts co-cultured with porcine chon- reduce collagen synthesis by fibroblasts via paracrine mechanisms. Wound
drocytes on a 3D biodegradable scaffold. Biotechnol Lett 2007;29:1685e90. Repair Regen 2007;15:497e504.
[88] Jiang J, Nicoll SB, Lu HH. Co-culture of osteoblasts and chondrocytes modu- [114] Shephard P, Martin G, Smola-Hess S, Brunner G, Krieg T, Smola H. Myofi-
lates cellular differentiation in vitro. Biochem Biophys Res Commun broblast differentiation is induced in keratinocyte-fibroblast co-cultures and
2005;338:762e70. is antagonistically regulated by endogenous transforming growth factor-
[89] Sheehy EJ, Vinardell T, Buckley CT, Kelly DJ. Engineering osteochondral beta and interleukin-1. Am J Pathol 2004;164:2055e66.
constructs through spatial regulation of endochondral ossification. Acta [115] Sun T, Haycock J, MacNeil S. In situ image analysis of interactions between
Biomater 2013;9:5484e92. normal human keratinocytes and fibroblasts cultured in three-dimensional
[90] Ma J, Van Den Beucken JJ, Yang F, Both SK, Cui F-Z, Pan J, et al. Coculture of fibrin gels. Biomaterials 2006;27:3459e65.
osteoblasts and endothelial cells: optimization of culture medium and cell [116] Smith AN, Willis E, Chan VT, Muffley LA, Isik FF, Gibran NS, et al. Mesen-
ratio. Tissue Eng Part C Methods 2011;17:349e57. chymal stem cells induce dermal fibroblast responses to injury. Exp Cell Res
[91] Darling EM, Athanasiou KA. Rapid phenotypic changes in passaged articular 2010;316:48e54.
chondrocyte subpopulations. J Orthop Res 2005;23:425e32. [117] Bishop ET, Bell GT, Bloor S, Broom IJ, Hendry NFK, Wheatley DN. An in vitro
[92] Meretoja VV, Dahlin RL, Kasper FK, Mikos AG. Enhanced chondrogenesis in model of angiogenesis: basic features. Angiogenesis 1999;3:335e44.
co-cultures with articular chondrocytes and mesenchymal stem cells. Bio- [118] Tremblay P, Hudon V, Berthod F, Germain L, Auger F. Inosculation of tissue-
materials 2012;33:6362e9. engineered capillaries with the host’s vasculature in a reconstructed skin
[93] Fischer J, Dickhut A, Rickert M, Richter W. Human articular chondrocytes transplanted on mice. Am J Transplant 2005;5:1002e10.
secrete parathyroid hormone-related protein and inhibit hypertrophy of [119] Kempf M, Miyamura Y, Liu P-Y, Chen AC, Nakamura H, Shimizu H, et al.
mesenchymal stem cells in coculture during chondrogenesis. Arthritis A denatured collagen microfiber scaffold seeded with human fibroblasts and
Rheum 2010;62:2696e706. keratinocytes for skin grafting. Biomaterials 2011;32:4782e92.
[94] Lv X, Zhou G, Liu X, Liu H, Chen J, Liu K, et al. Chondrogenesis by co-culture of [120] Co CC, Wang Y-C, Ho C-C. Biocompatible micropatterning of two different
adipose-derived stromal cells and chondrocytes in vitro. Connect Tissue Res cell types. J Am Chem Soc 2005;127:1598e9.
2012;53:492e7. [121] Bhatia SN, Balis UJ, Yarmush ML, Toner M. Microfabrication of hepatocyte/
[95] Dai J, Wang J, Lu J, Zou D, Sun H, Dong Y, et al. The effect of co-culturing fibroblast co-cultures: role of homotypic cell interactions. Biotechnol Prog
costal chondrocytes and dental pulp stem cells combined with exogenous 1998;14:378e87.
FGF9 protein on chondrogenesis and ossification in engineered cartilage. [122] Lamponi S, Di Canio C, Forbicioni M, Barbucci R. Heterotypic interaction of
Biomaterials 2012;33:7699e711. fibroblasts and endothelial cells on restricted area. J Biomed Mater Res A
[96] Jeong JY, Park SH, Shin JW, Kang YG, Han K-H, Shin J-W. Effects of inter- 2010;92:733e45.
mittent hydrostatic pressure magnitude on the chondrogenesis of MSCs [123] Wong KHK, Chan JM, Kamm RD, Tien J. Microfluidic models of vascular
without biochemical agents under 3D co-culture. J Mater Sci Mater Med functions. Annu Rev Biomed Eng 2012;14:205e30.
2012;23:2773e81. [124] Choi NW, Cabodi M, Held B, Gleghorn JP, Bonassar LJ, Stroock AD. Micro-
[97] Zhang Y-Q, He L-M, Xing B, Zeng X, Zeng C-G, Zhang W, et al. Neurotrophin-3 fluidic scaffolds for tissue engineering. Nat Mater 2007;6:908e15.
gene-modified Schwann cells promote TrkC gene-modified mesenchymal [125] Chung S, Sudo R, MacK PJ, Wan C-R, Vickerman V, Kamm RD. Cell migration
stem cells to differentiate into neuron-like cells in poly(lactic-acid-co- into scaffolds under co-culture conditions in a microfluidic platform. Lab
glycolic acid) multiple-channel conduit. Cells Tissues Organs 2012;195: Chip 2009;9:269e75.
313e22. [126] Carrion B, Huang CP, Ghajar CM, Kachgal S, Kniazeva E, Jeon NL, et al. Rec-
[98] Wei Y, Gong K, Zheng Z, Liu L, Wang A, Zhang L, et al. Schwann-like cell reating the perivascular niche ex vivo using a microfluidic approach. Bio-
differentiation of rat adipose-derived stem cells by indirect co-culture with technol Bioeng 2010;107:1020e8.
Schwann cells in vitro. Cell Prolif 2010;43:606e16. [127] Dickinson LE, Lütgebaucks C, Lewis DM, Gerecht S. Patterning microscale
[99] Suri S, Schmidt CE. Cell-laden hydrogel constructs of hyaluronic acid, extracellular matrices to study endothelial and cancer cell interactions
collagen, and laminin for neural tissue engineering. Tissue Eng Part A in vitro. Lab Chip 2012;12:4244e8.
2010;16:1703e16. [128] Caicedo-Carvajal CE, Liu Q, Remache Y, Goy A, Suh KS. Cancer tissue engi-
[100] Houchin-Ray T, Swift LA, Jang J-H, Shea LD. Patterned PLG substrates for neering: a Novel 3D polystyrene scaffold for in vitro isolation and amplifi-
localized DNA delivery and directed neurite extension. Biomaterials cation of lymphoma Cancer cells from heterogeneous cell mixtures. J Tissue
2007;28:2603e11. Eng 2011;2011:362326.
4476 K.G. Battiston et al. / Biomaterials 35 (2014) 4465e4476

[129] Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Yuan Hsin H, [147] Trappmann B, Gautrot JE, Connelly JT, Strange DGT, Li Y, Oyen ML, et al. Extra-
Ingber DE. Reconstituting organ-level lung functions on a chip. Science cellular-matrix tethering regulates stem-cell fate. Nat Mater 2012;11:642e9.
2010;328:1662e8. [148] Sharma RI, Snedeker JG. Paracrine interactions between mesenchymal stem
[130] Kostadinova R, Boess F, Applegate D, Suter L, Weiser T, Singer T, et al. A long- cells affect substrate driven differentiation toward tendon and bone phe-
term three dimensional liver co-culture system for improved prediction of notypes. PLoS One 2012;7:e31504.
clinically relevant drug-induced hepatotoxicity. Toxicol Appl Pharmacol [149] Palama IE, D’Amone S, Coluccia AML, Biasiuccia M, Gigli G. Cell self-
2013;268:1e16. patterning on uniform PDMS-surfaces with controlled mechanical cues.
[131] Lehmann AD, Daum N, Bur M, Lehr C-M, Gehr P, Rothen-Rutishauser BM. An Integr Biol (Camb) 2012;4:228e36.
in vitro triple cell co-culture model with primary cells mimicking the human [150] Ceylan H, Tekinay AB, Guler MO. Selective adhesion and growth of vascular
alveolar epithelial barrier. Eur J Pharm Biopharm 2011;77:398e406. endothelial cells on bioactive peptide nanofiber functionalized stainless steel
[132] Song JW, Cavnar SP, Walker AC, Luker KE, Gupta M, Tung Y-C, et al. Micro- surface. Biomaterials 2011;32:8797e805.
fluidic endothelium for studying the intravascular adhesion of metastatic [151] Lee JH, Khang G, Lee JW, Lee HB. Interaction of different types of cells on
breast cancer cells. PLoS One 2009;4:e5756. polymer surfaces with wettability gradient. J Colloid Interface Sci 1998;205:
[133] Arima Y, Iwata H. Effect of wettability and surface functional groups on 323e30.
protein adsorption and cell adhesion using well-defined mixed self- [152] Barrias CC, Martins MCL, Almeida-Porada G, Barbosa MA, Granja PL. The
assembled monolayers. Biomaterials 2007;28:3074e82. correlation between the adsorption of adhesive proteins and cell behaviour
[134] Jones GL, Motta A, Marshall MJ, El Haj AJ, Cartmell SH. Osteoblast: osteoclast on hydroxyl-methyl mixed self-assembled monolayers. Biomaterials
co-cultures on silk fibroin, chitosan and PLLA films. Biomaterials 2009;30: 2009;30:307e16.
5376e84. [153] Fuse Y, Hirata T, Kurihara H, Okazaki M. Cell adhesion and proliferation
[135] Efremov AN, Stanganello E, Welle A, Scholpp S, Levkin PA. Micropatterned patterns on mixed self-assembled monolayers carrying various ratios of
superhydrophobic structures for the simultaneous culture of multiple cell types hydroxyl and methyl groups. Dent Mater J 2007;26:814e9.
and the study of cell-cell communication. Biomaterials 2013;34:1757e63. [154] Xu L, Siedlecki CA. Effects of surface wettability and contact time on protein
[136] Hammoudi TM, Lu H, Temenoff JS. Long-term spatially defined coculture adhesion to biomaterial surfaces. Biomaterials 2007;28:3273e83.
within three-dimensional photopatterned hydrogels. Tissue Eng Part C [155] Aizawa Y, Shoichet MS. The role of endothelial cells in the retinal stem and
Methods 2010;16:1621e8. progenitor cell niche within a 3D engineered hydrogel matrix. Biomaterials
[137] Yang I, Co C, Ho C. Spatially controlled co-culture of neurons and glial cells. 2012;33:5198e205.
J Biomed Mater Res A 2005;75A:976e84. [156] Lutolf MP, Gilbert PM, Blau HM. Designing materials to direct stem-cell fate.
[138] Tsuda Y, Shimizu T, Yarnato M, Kikuchi A, Sasagawa T, Sekiya S, et al. Cellular Nature 2009;462:433e41.
control of tissue architectures using a three-dimensional tissue fabrication [157] Aizawa Y, Wylie R, Shoichet M. Endothelial cell guidance in 3D patterned
technique. Biomaterials 2007;28:4939e46. scaffolds. Adv Mater 2010;22:4831e5.
[139] Moeller H, Mian MK, Shrivastava S, Chung BG, Khademhosseini A. A microwell [158] Ventre M, Causa F, Netti PA. Determinants of cell-material crosstalk at the
array system for stem cell culture. Biomaterials 2008;29:752e63. interface: towards engineering of cell instructive materials. J R Soc Interface
[140] Bauwens CL, Peerani R, Niebruegge S, Woodhouse KA, Kumacheva E, 2012;9:2017e32.
Husain M, et al. Control of human embryonic stem cell colony and aggregate [159] Dinardo CL, Venturini G, Omae SV, Zhou EH, da Motta-Leal-Filho JM,
size heterogeneity influences differentiation trajectories. Stem Cells Dariolli R, et al. Vascular smooth muscle cells exhibit a progressive loss of
2008;26:2300e10. rigidity with serial culture passaging. Biorheology 2012;49:365e73.
[141] Alge DL, Azagarsamy MA, Donohue DF, Anseth KS. Synthetically tractable [160] Iyer RK, Chiu LLY, Vunjak-Novakovic G, Radisic M. Biofabrication enables
click hydrogels for three-dimensional cell culture formed using tetrazine- efficient interrogation and optimization of sequential culture of endothelial
norbornene chemistry. Biomacromolecules 2013;14:949e53. cells, fibroblasts and cardiomyocytes for formation of vascular cords in
[142] Tibbitt MW, Anseth KS. Dynamic microenvironments: the fourth dimension. cardiac tissue engineering. Biofabrication 2012;4:035002.
Sci Transl Med 2012;4. 160ps24. [161] Shao-Jun Wen, Li-min Zhao, Shen-guo Wang, Jing-xing Li, Hua-ying Chen,
[143] Bergmeister H, Schreiber C, Grasl C, Walter I, Plasenzotti R, Stoiber M, et al. Jie-lin Liu, et al. Human vascular smooth muscle cells and endothelial cells
Healing characteristics of electrospun polyurethane grafts with various po- cocultured on polyglycolic acid (70/30) scaffold in tissue engineered vascular
rosities. Acta Biomater 2013;9:6032e40. graft. Chin Med J 2007;120:1331e5.
[144] Choi S, Zhang Y, Xia Y. Three-dimensional scaffolds for tissue engineering: [162] Maidhof R, Tandon N, Lee EJ, Luo J, Duan Y, Yeager K, et al. Biomimetic
the importance of uniformity in pore size and structure. Langmuir 2010;26: perfusion and electrical stimulation applied in concert improved the as-
19001e6. sembly of engineered cardiac tissue. J Tissue Eng Regen Med 2012;6:e12e23.
[145] Sicchieri LG, Crippa GE, de Oliveira PT, Beloti MM, Rosa AL. Pore size regu- [163] Rauch MF, Hynes SR, Bertram J, Redmond A, Robinson R, Williams C, et al.
lates cell and tissue interactions with PLGA-CaP scaffolds used for bone Engineering angiogenesis following spinal cord injury: a coculture of neural
engineering. J Tissue Eng Regen Med 2012;6:155e62. progenitor and endothelial cells in a degradable polymer implant leads to an
[146] Kirkpatrick CJ, Fuchs S, Unger RE. Co-culture systems for vascularization e increase in vessel density and formation of the blood-spinal cord barrier. Eur
learning from nature. Adv Drug Deliv Rev 2011;63:291e9. J Neurosci 2009;29:132e45.

You might also like