Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

International Journal of Polymeric Materials and

Polymeric Biomaterials

ISSN: 0091-4037 (Print) 1563-535X (Online) Journal homepage: https://www.tandfonline.com/loi/gpom20

Polymeric microgels for bone tissue engineering


applications – a review

Lalit Kumar Meena, Hilal Rather, Dhaval Kedaria & Rajesh Vasita

To cite this article: Lalit Kumar Meena, Hilal Rather, Dhaval Kedaria & Rajesh Vasita (2019):
Polymeric microgels for bone tissue engineering applications – a review, International Journal of
Polymeric Materials and Polymeric Biomaterials

To link to this article: https://doi.org/10.1080/00914037.2019.1570512

Published online: 03 Feb 2019.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=gpom20
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS
https://doi.org/10.1080/00914037.2019.1570512

Polymeric microgels for bone tissue engineering applications – a review


Lalit Kumar Meena, Hilal Rather, Dhaval Kedaria, and Rajesh Vasita
Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, India

ABSTRACT ARTICLE HISTORY


Microgels have recently gained exposure as a potential scaffold either alone or in combination Received 15 October 2018
with other scaffold for bone tissue engineering. Microgel has been defined as a small size particle Accepted 13 January 2019
of cross linked polymer chains with the ability of swelling and holding the large amount of solv-
KEYWORDS
ent. This review summarizes the microgel based scaffolds specifically for bone tissue engineering.
Bone regeneration; cell
Microgels for growth factor delivery, cell transplantation and hybrid scaffold synthesis have been delivery; hydrogel; microgel;
discussed in details. The success in recreating potential bone regenerative scaffold have been osteogensis
explored for clinical applications, future perspectives and its concurrent view has been deliberated
in detail.

GRAPHICAL ABSTRACT

1. Introduction morbidity, dearth of grafts and nonunion complications


Bone comes second most transplanted tissue globally after restrict their utility. Allograft provides an alternative but it
blood with over two million bone grafting surgeries per also has issues like immune rejection and pathogen trans-
annum[1]. Bone regeneration is the complex phenomenon in mission[2,3]. Clinically need of alternative bone regeneration
which active signaling from different biomolecules is strategies keeps increasing with time and population ages
required at various stages of bone development. Small size globally. To compensate the demand of bone regeneration,
bone fracture whether it might be pathological or accidental bone tissue engineering has made the substantial growth
can be repaired by bone’s own regeneration capability. and remains a matter of hope in comparison to traditional
However, critical size bone defect compromise the active methods from last decades[4,5].
blood supply and infection lead to delayed unions or non- Bone substitute materials have been a subject of augmen-
unions. To assist the complete bone regeneration, autografts tation of the natural bone repair by providing space filler
and allograft have been used clinically for bone reconstruc- and inert support. Bone substitute materials industry claims
tion. Autologous bone grafts remain gold standard techni- more than two billion revenue from worldwide market in
ques to achieve successful bone repair however donor site 2013[6]. Bone tissue engineering is the cocktail of biological

CONTACT Rajesh Vasita rajesh.vasita@gmail.com Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, sector-30,
Gandhinagar, Gujarat, 382030 India.
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/gpom.
ß 2019 Taylor & Francis Group, LLC
2 L. K. MEENA ET AL.

and engineering principles which provide the basis for rapid


bone regeneration. The “diamond concept” of bone tissue
engineering proposes functional bone substitute mater-
ial[7–11]. Tissue engineering approach utilizes varieties of
synthetic and natural bone substitute materials for bone
regeneration. The synthetic bone substitutes like calcium
sulfate[12], calcium phosphate (CaP)[13,14], ceramics[15–17],
bioactive glass[18,19], calcium phosphate cement (CPC)[20–23]
provide osteoconductivity supports for space filling and
mechanical stability to the fractured bone. However, the lack
of osteoinductivity remains a challenge to prove as an alter-
native of natural bone substitute materials. The use of bio-
logical growth factors as osteoinductive molecules with these
synthetic bone substitutes would be desirable to augment
the bone regeneration. The bone substitute materials based
on natural polymers also considered next generation bone
Figure 1. Light microscopic images of core-shell (collagen 0.5% for core and
grafts. Natural polymers like collagen, alginate, and chitosan chitosan 3% for shell) microgels with intact shell structure and spherical shape.
are extensively explored for bioresorption and homing envir- Core-shell structures were indicated by arrows. Images were captured with 5X
onment to the bone progenitor cells[24–29]. The composites objective lens (scale ¼ 500 mm).
of synthetic bone substitute and natural polymers are also
reported for bone regeneration. The use of b-tricalcium
phosphate (b-TCP), hydroxyapatite with biopolymers like
collagen, alginate were investigated extensively[30–40]. The
use of osteoinductive growth factors and drug molecules
with these bone substitute materials are also clinically viable
approach. The list of growth factors explored for bone tissue
engineering includes bone morphogenetic proteins (BMPs),
vascular endothelial growth factors (VEGF), fibroblast
growth factors (FGF), parathyroid hormone (PTH) and
platelet-rich plasma (PRP); however, BMP-2 and BMP-7
were only approved by US Food and Drug Administration
(FDA) for bone regeneration in 2002[41,42]. Figure 2. Schematic representation of internal structure of cross-linked micro-
The development of ideal scaffold with biocompatibility gel and swelling (size) behavior in response to external stimuli.
(support the normal cellular activity without cytotox-
icity)[43], biodegradability (providing the space for new bone ability of swelling and holding the large amount of good
growth)[44], osteoconductivity, osteoinductivity, scaffold solvent (e.g. water) (Figure 1)[50]. Because of these proper-
architecture (porosity) and mechanical properties[45–47] is ties, they have gained tremendous attention. The responsive
challenging and complex process. However scientists are try- swelling and de-swelling in response to surrounding
ing to develop scaffolds with most of these properties to medium pH, temperature and solvent makes it promising
augment the bone regeneration. As a bone substitute grafts deliver device for bioactive molecules (Figure 2). The cross
and local delivery of osteoinductive molecules by various linking of polymeric chains or cross linking density controls
types of osteoconductive scaffolds have been explored. These swelling and shape of microgel. The change in size of micro-
are the hydrogels, cryogels, microspheres, microgels and gel upon swelling is inversely proportional to cross linking
nanoparticles. Among all these, microgels recently have been density[51]. The nature of solvents also influences swelling
explored for bone tissue engineering either alone or in com- and size of microgel, for example, poly(N-isopropylacryla-
bination with other scaffold. Microgels small size, injectabil- mide) (PNIPAAm) based microgels demonstrated deswelling
ity, and live cell encapsulation efficiently, makes it and reduction in size in higher volume fraction of alcohol
promising scaffold for bone regeneration. Hence this review whereas, in low volume fraction of alcohol microgels re-
will be focused on microgel technology for bone tissue swells[52]. Therefore microgels are termed as “smart materi-
engineering and their commercial perspective. als” because of their tunable size, shape, stiffness and par-
ticular interactions under the influence of external stimuli.
Microgels are cross-linked soft particles which provide a
2. Microgel
three-dimensional (3D) network structure[53]. The porous
In 1935 Staudinger and Husemann first time fabricated network allows the cells to penetrate inside the microgel and
microgel through suspension polymerization process[48]. provides an advantage of nutrient and gaseous exchange,
However the term microgel was remained unused till which directly affects the cellular growth and proliferation.
1949[49]. Microgel has been defined as a small size particle Microgels are also called as ‘hydrogels’ because of water sol-
(0.1–1000 mm) of cross linked polymer chains with the uble nature. Microgels and hydrogels both belong to gel
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 3

Table 1. Comparative profile of microgels and hydrogels features for bone tissue engineering.
Feature Microgels Hydrogels
Shape and size Spherical shape and size in micron range, hige surface to Shape not defined and size range above mm, helpful for
volume ratio is helpful in higher loading of osteoinduc- providing bone microenvironment[56]
tive molecules and controlled release[54,55]
Swelling and porosity Due to confined shape and less porosity, swelling occurs in Due to the higher porosity swelling occurs vary rapid that
controlled manner hence lead to controlled and sus- may lead to burst release of osteoinductive molecules
tained release of osteoinductive molecules[57]

family and have capacity to hold the large amount of water. integration with host tissue. The regulated swelling and deg-
However some features such as size, shape, swelling and radation of microgels imparts controlled release of growth
porosity make certain difference for bone tissue engineering. factors and cells, hence enhanced the regeneration of local
These features are tabulated in following Table 1. tissues such as bone, and cartilage could be achieved. These
On the basis of charge, microgels may be neutral or properties of microgels could be used for bone regeneration.
charged that mainly depends on materials composition. A In following sections the focus will be on how microgels can
wide range of polymers and monomer/co-monomers have assist the bone regeneration.
been used to fabricate the microgels such as alginate[58], gel-
atin[59], collagen[60], heparin, polyethylene glycol (PEG)[61],
chitosan[62], hyaluronan[57] PNIPAAm, methylmethacrylate, 3. Requirements for bone tissue
methacrylic acid (MAA), styrene, divinylbenzene, acrylic engineering scaffold
acid (AA), N,N’-methylene bisacrylamide (MBA) and ethyl- Bone regeneration using biomimetic approach involves the
ene glycol dimethacrylate etc.[63] among them PNIPAAm is use of several scaffolds which mimics certain features of the
extensively used[64]. The methylenebisacrylamide is com- bone anatomy or physiology. It includes the use of 3D con-
monly used cross-linker to impart mechanical strength and structs, scaffolds or matrices for example, 3D printed scaf-
prevents the complete dissolution of PNIPAAm
folds, hydrogels, cryogels, microspheres and nanofibers.
microgels[63].
These constructs regulate cell migration, proliferation, and
Diverse methods from conventional in situ polymerization
differentiation[74–76]. Before considered for bone tissue
to microfludics have been employed for the fabrication of dif-
engineering, these scaffolds should fulfill certain criteria.
ferent types of microgels. The chitosan based genipin cross-
linked microgels prepared by in situ emulsion crosslinking
method. This method produces homogenously crosslinked 1. ECM mimicking: To imitate the fibrillary architecture
microgels. These microgels have been employed for the post of natural ECM components.
fabrication loading of vascular endothelial growth factor iso- 2. Biodegradability: To support the replacement of scaffold
forms and in vitro release[65]. Arginine–glycine–aspartic acid by the newly secreted ECM
(RGD) grafted alginate microgel has been fabricated by elec- 3. Surface chemistry: To support cell attachment, prolifer-
trospraying method. Electrospraying at 10 kV reported cyto- ation, migration and differentiation
compatible for encapsulation of human outgrowth 4. Osteoconductivity: To act as substrate for cells to grow
endothelial cells (OECs) in microgels[66]. Precipitation poly- and differentiate.
merization technique has been used for poly(N-isopropyla- 5. Osteoinductivity: Release of Osteoinductive molecules,
crylamide-co-acrylic acid) based vancomycin loaded which could promote the osteogenic differentiation of
microgels fabrication[67]. Collagen based cell encapsulated progenitor cells.
microgels has been prepared by aqueous two-phase system, 6. Mineral deposition: Biomimetic scaffolds should provide
in which soluble polymer phase thermodynamically drive a microenvironment to facilitate the ossification process.
aqueous phase to form two separated phases[60]. Gelatin and 7. Mechanical properties: To provide mechanical stability
poly(ethylene glycol)-maleimide (PEG-4MAL) microgels has for constructs in load-bearing sites prior to synthesis of
been synthesized with uniform size and reproducibility by new ECM by cells.
microfludics technique[59,68]. While solvent/water inverse 8. Porosity: For exchange of nutrients and gases in vitro
microemulsion cross-linking method has been used for the and infiltration of blood supply in vivo.
loading of labile growth factors such as BMP-2 in heparin- 9. Ease of fabrication: the fabrication process should be
conjugated hyaluronan microgels[57]. less sophisticated, less time consuming and least toxic
The control over size, composition and functionality of reagents should be used.
microgels enhances its applications in biomedical areas like Microgels are also used to fulfill the criteria as reported
tissue engineering[69], controlled drug delivery[70,71], spinal in the literature. The core-shell microgels mimic the hirer-
column implants[72] and cells delivery[73]. The aqueous chial structure of the bone ECM and acts as osteoconductive
internal environment of microgel mimics extracellular scaffolds. They can be easily fabricated from natural poly-
matrix that is desirable to deliver the physical and chemical mers, which enhances their biodegradability. Furthermore,
stress labile growth factors and progenitor’s cells. A high the polymeric microgels are functionalized to change the
surface area maximizes its interactions with cells and host surface chemistry and alter the functionality of the micro-
tissue that is prime requirement for biomedical device gel[67]. The osteoinductivity of the microgels could be
4 L. K. MEENA ET AL.

controlled by loading several osteoconductive molecules[77]. for larger scaffold implant. Hence injectable properties of
Moreover, microgels could be used with hydrogels to microgels enhances its applicability in bone regeneration[73].
improve the mechanical stability, release profile and immo- Further, microgels could be used as a micro carrier for
bilization at site of implanting[78]. Furthurmore, injectable growth factor and drug delivery in bone regeneration
form of microgel is considered to be advantageous over (Figure 5). Since microgels are confined structure of poly-
other bone substitutes. Hence, microgels fulfill the basic cri- mers as compared to bulk gel, the controlled release of bio-
teria related with the bone tissue engineering scaffolds. macromoleculs could be achieved[80]. The progenitor cell
transplantation is the most effective therapeutic strategy to
augment the bone regeneration. Stem cells such as mesen-
4. Microgel for bone regeneration chymal stem cells (MSCs) have been encapsulated in micro-
The extensive research is going on the use of microgel for gels during fabrication and these microgels can be injectable
various tissue regeneration applications because of its soft at fracture site by syringe[81,82]. Alternatively, solvent access-
internal structure, elastic nature and injectable properties. ible surface of microgel also provides an adhesive site for
Bone structure is composed of two types of bony parts such cells which can be used as scaffolding. On the other hand
as cortical or compact bone and cancellous or spongy bone the therapeutic biomolecules loaded microgel can be loaded
(Figure 3a). The compact bone envelops spongy bone and in bulk gel system during hybrid scaffold fabrication[78,83].
These hybrid scaffolds could be used for progenitor cell cul-
forms a border with lower porosity and surface area. The
ture. The release of biomolecules from loaded microgels in
higher density and lower porosity of compact bone enhances
bulk gel could be mimicking the bone (ECM) microenviron-
its resistance against mechanical forces. The spongy bone is
ment[77]. A detailed discussion on aforementioned microgel
encircled by compact bone and has higher porosity and
based scaffolding (Figure 5) is provided in subsequent sec-
large surface area. The spongy bone is the network of tra-
tion of this review.
becular struts with space between them ranges from 300 to
1500 mm[79]. The bones structural organization mimicking
remains prime aim for scaffolds designing. A core-shell
microgel with different microstructure and composition can
be considered as next generation scaffolding. In Figure 3b a
core-shell microgel showed the core is mimicking spongy
bone organization while shell shows structural similarity
with compact bone. This structural similarity between bone
and microgels could be helpful to mimic cellular behavior
and transportation of signaling molecules. Bone regeneration
is the research area where the microgel providing an alterna-
tive of existing hard and soft scaffolds.
The production of homogeneous size microgels with soft
and elastic nature increases the applicability of the microgel
for injection at fracture site. Most of the studies have
explored microgels because of the some salient features as
mentioned in (Figure 4). The injectability of monodisperse
microgels at fracture site minimizes handling effort and Figure 4. Highlights the salient features of microgel which have been explored
avoids any open surgical complications, which are essential to enhance the osteogenesis.

Figure 3. Bone structural organization, (a) schematic representation of internal structure of bone with clear demarcation between compact bone and spongy bone,
(b) SEM image of core-shell microgel internal structure with highly porous core and less porous and compact shell.
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 5

4.1. Microgel as delivery device presentation of growth factors to guide the bone regener-
ation in vivo[86]. Growth factors are the key to augment the
4.1.1. Microgel based growth factor delivery
bone regeneration (Figure 6) and maintain the
Growth factors are the bioactive polypeptides that transduce bone metabolism.
the signal by cell surface receptors and guide the cells fate During natural fracture healing mechanism, an array of
process in vitro as well as in vivo[84,85]. The bone ECM bioactive signaling molecules become operational to guide
provides homing environment and spatio-temporal the bone cells to repair the fractured bone. In critical
defects, the disruption of blood vessels could imbalance the
signaling of growth factors. This interrupted signaling many
times causes the delayed union or nonunion. Therefore
recreation of local microenvironment became essential for
complete bone fracture repair. To simulate the natural heal-
ing mechanism, therapeutically relevant growth factors are
administered by various means. Conventionally practiced
systemic delivery of growth factors is least preferred due to
lack of targeted delivery and bioavailability[87]. The longer
circulation time of growth factors in body fluid leads to
decrease in bioactivity and half life which increase the
requirement of higher dose size for effective signaling initi-
ation. The increase in dose size or supara-physiological dose
causes the systemic side effects like ectopic bone formation,
neoplasia and inflammatory reactions[88,89]. Alternatively
delivery vehicles such as collagen sponges, hydrogels, mem-
branes, microparticles, implant coating, nanoparticles,
fibrous scaffolds, microgels and nanogels were explored for
localized delivery. Since present review is focused on micro-
gels applications in bone regeneration the remaining scaf-
folds will not be discussed further. The water holding
capacity, large solvent accessible area and ECM mimicking
Figure 5. Schematic demonstration of different applications of microgel in environment inside the microgel[90] facilitates encapsulation
bone regeneration. and release of therapeutic biomolecules and growth

Figure 6. Involvement of growth factors in different bone regeneration stages.


6 L. K. MEENA ET AL.

factors[91,92]. Loading of therapeutic biomolecules in micro- reinforce the glycidyl-methacrylated hydrogel for sustained
gels is the critical step and mainly achieved by two ways. In delivery of BMP-2[78]. The in vitro release of BMP-2 showed
one way the therapeutic biomolecules can be loaded during 20% at day one from heparin-conjugated hyaluronan micro-
microgel fabrication, called prefabrication. Pre-fabrication gel reinforced hydrogel in comparison to bulk gel (glycidyl-
loading can be used to achieve higher loading and sustained methacrylated hyaluronan gel) with 60% burst release at day
release of therapeutic biomolecules. The exposure of fabrica- one. The controlled release achieved in this study is desir-
tion environment to the labile therapeutic biomolecules may able for osteogenic cells differentiation. Further this study
reduce its bioactivity. In another way loading can be also demonstrated the relationship between swelling ratio
achieved after microgel fabrication by immersing the micro- and BMP-2 loading. The higher swelling ratio of bulk gel
gels into therapeutic biomolecules solution is called as post- claims highest BMP-2 loading 43.4 ± 0.9 ng/mg, in compari-
fabrication loading. Due to the small size and higher surface son to microgels reinforced glycidyl-methacrylated hydrogel
area of microgels post-fabrication loading is the rapid and (without heparin) with 21.9 ± 3.3 ng/mg loading. But as hep-
mild process[55]. The lower amount adsorption and uncon- arin content increasing from 1 to 10%, BMP-2 loading also
trolled release of therapeutic biomolecules limits this process increased from 28.2 ± 1.6 ng/mg to 42.4 ± 2.2 ng/mg in
applicability. Since the interactions of functional groups of microgels reinforced glycidyl-methacrylated hydrogel. In
polymeric microgels with therapeutic biomolecules also plays spite of low swelling of microgels, reinforced glycidyl-metha-
important role in determining the release profile and loading crylated hydrogel with 10% heparin showed comparable
capacity of microgels. A range of natural and synthetic poly- BMP-2 loading as bulk gel[78]. In conclusion swelling ratio
mers including collagen[60], heparin, PEG[61], chitosan[62], was not the only factor for higher BMP-2 loading but nega-
hyaluronan[57], poly(N-isopropylacrylamide) have been tively charged heparin content in microgels also play a part.
explored for fabrication of growth factor loaded microgel. Study demonstrated that microgel reinforced hydrogel could
The growth factor BMP-2 is extensively studied growth fac- be desirable way to deliver the BMP-2 in more sustained
tor and participates in all phases of bone regeneration. manner for bone regeneration application.
BMP-2 also reported for its role in MSCs recruitment and
proliferation during inflammatory phase and MSCs differen- 4.1.2. Microgel based drug delivery
tiation in soft callus formation and mineralization (Figure The delivery of bioactive drug molecules is the first step
6). To modulate the release profile of BMP-2 functionally which decides the fate of elicited responses. The systemic
active polymers such as heparin has been used. In one study, delivery of drug associated without any vehicle raises sys-
Zhao et al. synthesized heparin-conjugated hyaluronan (HA- temic toxicity and associated side effects. The reduced bio-
Hp) microgels through inverse emulsion polymerization activity, bioavailability and repeated dosage further
technique. Three different concentration of heparin was complicated the patients complains. An answer to following
evaluated for its effect on loading efficiency and release of limitations could be vehicle based drug delivery. Polymeric
BMP-2. With increasing heparin concentration the loading microgel systems have been developed for extended drug
efficiency of BMP-2 increased from 53% to 90%[57]. BMP-2 delivery. Polymers such as poly(N-isopropylacrylamide-co-
possesses basic heparin binding domain that might electro- acrylic acid)[67], polyvinyl alcohol (PVA)[95], collagen[77] and
statically interact with acidic sulfate groups on heparin[93]. locust bean gum (LBG)[96] have been explored in the form
The availability of BMP-2 throughout the bone healing is of microgel for osteoinductive drug, antibiotics deliv-
desirable to achieve the complete bone regeneration. The ery[77,97]. Infection is the major barrier in bone recovery and
sustain release profile maintains a continuous supply of may lead to loss of limb[98,99]. During the open fracture sur-
BMP-2. In aforementioned study, the in vitro release pattern gery antibiotics such as cefazolin, pipercacillin, tazobactam,
of BMP-2 from HA-Hp microgels also showed sustained tobramycin and penicillin are extensively used via systemic
release with 7% at day 1 in compared to 26% at day 1 from route[100]. The higher dose size and frequent administration
HA microgels. The remaining BMP-2 showed sustained of antibiotics causes side effects and patient discomfort
release for 15 days from HA-Hp microgels. Results showed respectively. To combat the infection at open fracture site
that the increased content of heparin in HA microgels initial faster release followed by sustained release of anti-
imparts sustained release pattern for BMP-2[57]. It might be biotic molecules for longer time is desirable. In one study,
due to increased electrostatic interactions between heparins vencomycin has been used in three different ways to provide
N- and O-sulfated residues and BMP-2 lysine and arginine the antibiotic functionality to the BioglassV-based scaf-
R

residues[94]. In conclusion, biological material based micro- folds[67]. In first group vancomycin was directly coated with
gels showed more controlled release of BMP-2 and high Poly (D,L-lactide-co-glycolide) (PLGA) solution on
BioglassV-based scaffolds. In second group the vancomycin
R
loading efficiency that would be helpful in bone
regeneration[57]. loaded poly(N-isopropylacrylamide-co-acrylic acid) micro-
gels coated with PLGA solution on BioglassV-based scaffolds.
R
Although microgels have been used for growth factor
delivery alone but its use with hydrogel may improve the In third group vancomycin loaded microgel coated with
double layer of PLGA on BioglassV-based scaffolds. The
R
mechanical stability, release profile and immobilization at
site of implanting. In a different study, Yuan et al. used gly- drug release study of group one scaffold demonstrated initial
cidyl methacrylate to surface modification of heparin-conju- burst release of vancomycin at 40 min. and remaining drug
gated hyaluronan microgel. These microgels were used to was released within 8 h. On the contrary, group two and
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 7

three scaffolds showed initial slower burst release of vanco- effect of cross linker density on dexamethasone release was
mycin at 6 h, and further extended controlled release until also studied. Results showed higher release rate of dexa-
4 days. So vancomycin loaded microgel coating provides a methasone at lower crosslinking density. It was due to the
release profile where initial rapid release followed by long easier diffusion of drug from less dense polymer net-
term sustains release of vancomycin is desirable to prevent work[101]. These studies demonstrated the applicability of
the infection and achieve the healing[67]. The release behav- microgels as a drug delivery vehicle for osteoinductive and
ior of drug from microgels can be regulated further by antibiotic molecules in bone tissue engineering. In conclu-
changing the polymer. A study by Lv et al. presents polyvi- sion the micro sized geometry of gel provides high surface-
nyl alcohol-borax (PVA-B) based microgels that were inves- to-volume ratio and good diffusion properties to control the
tigated for burst release of vancomycin hydrochloride. 94% release of cargo. The ECM mimicking nature of microgel
of drug was released in 5 min and remaining 6% was helps to protect growth factors bioactivity for longer time.
released within 4 h. The rapid burst release can be attributed While the stimuli responsive microgels advances its applica-
to hydrophilic nature of microgels, which measured by con- tion as delivery vehicle.
tact angle analysis (38.2 ± 0.64 ). The hydrophilic nature
allows rapid swelling of microgels and facilitates burst
4.1.3. Microgel for cell transplantation
release of drug[95]. Such studies demonstrated that microgels
In addition to growth factors and drugs, cells are another
of different polymers could be used as delivery vehicle for
key player in bone tissue regeneration. The disturbed pool
antibiotic molecules. The microgel based initial burst release
of osteoblasts, osteocytes and osteoclasts and imbalanced
followed by sustain release of antibiotic is desirable to mini-
microenvironment of cellular talk in fractured bone leads to
mizes the chances of infection and rapid healing of
delayed union and non union. The cells transplantation is a
bone fracture. viable strategy to restore the bone cells population at frac-
In addition to antibiotics, microgels also have been inves- ture site. Stem cells like MSCs[102], murine embryonic stem
tigated to assist the delivery of osteoinductive drug such as cells (ESCs)[103,104] and primary cells like human umbilical
dexamethasone in bone regeneration. The collagen microgels vein endothelial cells (HUVECs)[105–107] have the potential
integrated with poly(L-Lactide) (PLLA)-Dexamethsone to regenerate the diseased and injured bone tissue[108–110].
(Dex)-collagen hybrid scaffold were used to induce osteo- However, their transplantations or delivery at site of regen-
genic differentiation in MSCs[77]. In this study author used eration remains challenging due to poor viability and reten-
PLLA-collagen scaffold without collagen microgel as control tion of cells at the site of injury[111]. Cytocompatible and
with 2.5% dexamethasone. While collagen microgel loaded hydrated microgel would be desirable carrier for encapsula-
PLLA-collagen hybrid scaffold with dexamethasone concen- tion of cells and their localized delivery[112,113]. Microgels
tration 0%, 2.5%, and 5% were used. The results demon- further facilitates large surface area to volume ratio, cellular
strated the initial burst followed by sustained release of microenvironment, cell-cell and cell-matrix interactions[114].
dexamethasone from all types of scaffolds. As the dexa- Due to these properties, microgels are more suitable for
methasone concentration increased in microgel loaded maintaining the encapsulated cells metabolically live for lon-
PLLA-Collagen hybrid scaffolds, the initial burst release was ger time period.
also increased. The initial rapid followed by sustained release Microgel based delivery of cells can be achieved by two
of dexamethasone could be desirable to maintain the thera- different ways. In first approach cells can be delivered via
peutic concentration. The loaded microgels assisted in sus- seeding on microgel surface and in second cells can be
tained release of dexamethasone in therapeutic window for encapsulated within microgel matrix and delivered at target
up to 28 days. While scaffold without microgel showed low site. Following section will discuss the recent investigation
initial burst and slowest sustained release, which could be explored by both methods.
due to hydrophobic PLLA. The effect of release pattern of
dexamethasone from Dex-microgel loaded PLLA-Collagen 4.1.3.1. Microgel for delivery of surface adhered cells.
hybrid scaffolds showed significantly increased expression of Microgels as microcarrier for cells transplantation offer sev-
COL1 (approximetly 1.7 fold) in cultured MSCs as com- eral advantages including large surface area in comparison
pared to without microgel scaffold after 21 days[77]. The ini- to conventional 2D surfaces. It enhances the chances of high
tial burst release from Dex-microgel loaded PLLA-Collagen number of cells culture and proliferation in small volume of
hybrid scaffolds maintains therapeutic (325 ± 27 nM) concen- culture media before transplantation. Microgels surface give
trations in culture medium that might have caused increased the opportunity to customize the chemical and physical
expression of COL1. In another study, the stimuli responsive properties relevant to true cellular microenvironment[59].
microgels were investigated for delivery of dexamethasone Microgel surfaces can be tailored to enhance cells adhesion,
and vencomycin. Poly(N-isopropylacrylamide-co-acrylic proliferation and differentiation (Figure 7a, b). Fabrication
acid) based temperature sensitive microgels prepared by pre- of specific growth factor loaded microgel with cells seeded
cipitation polymerization method showed higher release of on the surface could be the new approach. After injection of
dexamethasone at 37  C in compared to at 25  C. It is due this type of microgels, sustained release of growth factor to
to the release of isopropyl group bound water by thermal differentiate the cells population would be expected.
induction, which increases the intra and intermolecular However no such study has been reported until but in
hydrophobic interactions between isopropyl groups. The future this is possible for bone regeneration. In a study by
8 L. K. MEENA ET AL.

Figure 7. Microgel based cell delivery strategies, (a) SEM image of pre-osteoblast MC3T3-E1 cells cultured on surface of core-shell microgel (cells indicated by red
arrow), (b) schematic demonstration of MSCs adhered on ECM mimicking surface of microgel, (c) schematic demonstration of encapsulated MSCs in the ECM mim-
icking microgel bulk matrix.

Schmidt et al. the arginine-glycine-aspartic acid (RGD) pep- high cross linking density. The 5 kPa increase in compressive
tide were used to tailor the surface of alginate microgels. modulus of microgels with increasing concentrations of
They found increased cell growth rate and osteogenic differ- four-arm PEG-acrylate demonstrated mechano-compatibility
entiation as RGD peptide density increased on microgel sur- of microgel that is desirable for bone regeneration applica-
face from 6.22  107 to 6.22  108 RGD/mm2 on single tion. The cytocompatibility of GelMA and HA-SH microgel
microgel. On the microgel surface, RGD peptide enhances with hMSCs were evaluated. The viability of encapsulated
bone marrow-derived MSCs adhesion as compared to with- hMSCs was maintained in both types of microgel from day
out RGD microgel. The increased RGD density on microgel 1 to 7[117]. This result demonstrates the feasibility of micro-
surface also increased the secretion of osteocalcin and osteo- gels in oxygen and nutrient diffusion to maintain the cell
pontin as osteogenic differentiation markers. It might be viability. In one study Hou et al. prepared hMSCs and
due to the increased cells integrin-RGD interactions as com- BMP-2 loaded degradable microgel of poly(vinyl alcohol)
pared to without RGD modified microgels[115]. This study (PVA) through the micro-fluidic technology to provide
provides a new approach to design the novel cell adherent osteoinductive microenvironment for encapsulated hMSCs.
microgels for delivery of therapeutic multipotent stem cells The 50% release of BMP-2 in 10 days from microgel was
to enhance the bone tissue regeneration. observed. This controlled release of BMP-2 was able to
maintained osteoinductive concentration of BMP-2 inside
4.1.3.2. Microgels for delivery of encapsulated cells. the microgel for encapsulated hMSCs. In osteoinductive
Homing of precursor cells in three dimensional (3D) micro- environment the ALP activity was significantly increased
environments that could direct their osteogenic differenti- 2.0  103 nmol/ng, in compared to microgel without BMP-
ation is a major focus of tissue engineers. Various types of 2 with approximately 1.0  103 nmol/ng. The calcium con-
biomaterial based scaffolds have been developed to recreate tent also increased approximately 30 fold in treatment group
the bone tissue microenvironment; however, because of the compared to the control group at 28 days. The osteogenesis
harsh fabrication conditions, only few techniques have related genes expression of Runx2 (approximately 1 fold)
gained the success of cell encapsulation. Microgels have and OPN (approximately 2 fold) was also increased signifi-
gained some attention because they provide the three cantly after 3 week with treatment compared to the control
dimensionality to the cells entrapped in them[116] and entrap group. The results of study indicate that the long-term sus-
the live cells during the fabrication process without much tained release of BMP-2 enhanced hMSCs osteogenic differ-
damage to the cell viability (Figure 7c). The other advan- entiation. Therefore, osteoinductive microgel approach is a
tages for encapsulation of cells in microgel include the pro- promising strategy for controlled cell delivery in bone
tection of the cells from external stress during injection and regeneration[82].
transplantation. Furthermore, they avoid the faceoff with Bone tissue engineering is a complex process, which
host immune system hence increases their chance for inte- requires regulating three- dimensionality of cellular interac-
gration to the host body. The encapsulations of stem cells tions. Because during osteogenic differentiation cells
are more challenging as they are intends to self-renew and undergo cellular condensation, which amplifies the number
differentiate simultaneously. Guermani et al. engineered of osteogenic cells[118]. It is found that the early spheroid
microgel array to evaluate the differentiating potential of formation resulted from cellular condensation, leads to upre-
encapsulated human mesenchymal stem cells (hMSCs). They gulation of several bone related genes[119]. Furthermore, cell
used gelatin methacrylate (GelMA) and thiolated hyaluronic spheroid formation enhances bone formation both in vitro
acid (HA-SH) to engineer the hMSCs laden microgel arrays and in vivo[120]. Alginate- -RGD microgels were used to
through 3D printing system. Prepolymer GelMA with encapsulate human MSCs spheroids to retain their structure.
hMSCs in form of microdrops were crosslinked by UV The morphology of the spheroid was retained within micro-
irradiation while hMSCs laden prepolymer HA-SH micro- gel, since the solid gel formed by cross linking of alginate
drops were crosslinked by four-arm PEG-acrylate. In case of molecules around the spheroid. RGD mimics the extracellu-
HA-SH microgel, two different concentrations 0.5% and 1% lar component of the matrix; hence exhibit the cell-matrix
of four-arm PEG-acrylate was used. In results the compres- interactions. The encapsulation of human MSC spheroids in
sive modulus was improved from 5 kPa to 10 kPa due to the microgel demonstrate enhanced osteogenic
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 9

differentiation[121]. In summary, the microgel is the most regeneration because the hydroxyapatite formation is consid-
desirable delivery device for therapeutic cells delivery in ered as gold standard for determination of osteogenic differ-
bone regeneration. It is because of good diffusion properties entiation, as it is the major constituent of bone[122–124]. To
and ECM mimicking 3 D microenvironment that helps to the best of our knowledge, so far only few microgel hybrid
maintain cells viability. In future, there is ample opportunity scaffolds are being used for the osteogenic differentiation/
to develop the microgel based delivery system for multiple bone regeneration applications, which are mentioned above.
cells with signaling molecules delivery to enhance the bone However, few studies have reported only the formulations
regeneration. for developing microgel osteoconductive hybrid scaffolds.
Their applications for osteogenic differentiations are yet to
be reported. Birkholz et al. has fabricated microgel-b-TCP
4.2. Microgel as scaffolds
composite scaffolds. The microgel was fabricated using
It is proven that microgels are desirable vehicle for deliver- Poly(N-vinlycaprolactam), 2-(methacryloyloxy)ethyl acetoa-
ing the growth factors, drugs or even cells. However, their cetate and 1-vinylimidazol. The microgel dispersions were
immobilization at site of implant, integration at target tissue mixed with b-TCP and freeze dried or spray dried to yield
and poor sustained release remains open challenges for their composite granulates. They proved that the microgel chem-
use as implantable devices. Integration of microgels with ically interacted with b-TCP particles without use of any
existing scaffolding technology would be an interesting and binder. Therefore it is possible to use these hybrid scaffolds
innovative approach for bone tissue engineering. In prin- without any phase impurities for the manufacturing of bone
ciple, microgels can be used in combination with the other implants via powder based 3D-printing. Furthermore, the
osteoinductive or osteoconductive scaffolds, however only porosity of the printed scaffolds can be increased by burning
few studies have been performed so far and the field has a out of microgels in a sintering process, which can give an
potential to be explored more. Nanda et al used osteoinduc- advantage in the process of bone resorption[125].
tive microgels in combination with the osteoconductive Cell morphological changes are correlated with the cellu-
hybrid scaffold[77]. The collagen microgels were used to lar differentiation. Jhala et al. has reported that change in
mimic the osteoid microenvironment and assisted the the cell shape has influenced their differentiation potential.
release of dexamethasone which is osteoinductive in action. The cell shape changed from spindle to polygonal shaped
The incorporation of the collagen microgel was observed to during osteogenic differentiation. The cells shapes were
control the interconnected pore structure, which allowed influenced by growing them on different types of sub-
MSCs to infiltrate and distribute in the scaffold. The porous strates[76]. Microgels have also shown the potential to influ-
structure of the hybrid scaffold resulted in the increased cell ence the change in cell morphology to induce osteogenic
growth. Furthermore, the microgel facilitated spatio-tem- differentiation. In a study by Dai et al. cell culture dishes
poral control of dexamethasone release to induced the dif- were coated with Poly(N-isopropylacrylamide-co-acrylic
ferentiation of human bone marrow derived MSCs towards acid) microgels to culture MSCs. Cells grown on microgel-
osteogenic lineage. The collagen-dexamethasone microgel coated surface changed their morphology from spindle shape
loaded PLLA-Collagen hybrid scaffold showed increased to more stretched and elongated cell shape (Figure 8).
expression of alkaline phosphatase, osteopontin, integrin- Furthermore, cell morphological changes were accompanied
binding sialoprotein, and runt-related transcription factor 2 with the change in expression of osteopontin, Runt-related
after 21 days. Where, the expression of collagen type 1 was transcription factor 2 (RUNX2) and collagen type 1, which
highest in Dexamethasone-microgel loaded hybrid scaf- are osteogenesis related genes. Microgels were also observed
fold[77]. Another study performed by Olalde et al. has also to elevate the expression level of these genes without any
shown the potential of microgel hybrid scaffold in bone inducing factor[126].
regeneration/osteogenic differentiation. The microgel was For the bone defect regeneration, injectable gel systems
used as a hybrid scaffold in a different manner. They devel- have many advantages over the conventionally used scaffolds
oped a new family of scaffolds, incorporating biopolymer such as increased handling properties, controlled flowability
coatings on BioglassV derived foams. The biopolymer coat- and adaptability to the defect site[127,128]. Furthermore, the
R

ing was used as an antibiotic drug carrier. They found that injectable gel can be incorporated with growth factors or
the coating with PLGA with vancomycin imparts antibiotic any bioactive molecule which could aid and increase the
functions to the scaffold. The primary object of the study bone defect reconstruction[129]. Therefore, injectable form of
was to develop the multifunctional scaffold. They found that microgel is considered to be advantageous over other bone
coating of BioglassV with vancomycin loaded poly(N-isopro-
R
substitutes. Nanohydroxyapatite incorporated chitin-poly(e-
pylacrylamide-co-acrylic acid) microgel dispersed in PLGA caprolactone) (Chitin-PCL-nHAp) based injectable microgels
provides a rapid delivery of drug. They showed the antibac- has been used by Kumar, et al. for bone defect repair. Their
terial effect of the drug loaded hybrid microgel scaffold at results depicted that Chitin-PCL-nHAp microgel elicited an
the wound site. Although, no direct assay for bone regener- early osteogenic differentiation compared to Chitin-PCL
ation was performed; however, the microgel was found to gel[130]. In another study, Das et al. used injectable hybrid
increase the bioactivity of scaffold by acting as nucleation microgel/microspheres for the delivery of sphingolipid
center for hydroxyapatite crystal formation in simulated growth factor FTY720 to critical size cranial defects in rats.
body fluid[67]. Hence, it could promote the bone The microgel composed of chitosan and inorganic
10 L. K. MEENA ET AL.

Figure 8. Shows the immunostaining of F-actin in MSCs grown on normal culture plate (left) and plates coated with Poly(N-isopropylacrylamide-co-acrylic acid)
microgels (right), the pointed arrows show the increased density of actin which enhances the morphological changes in the cells. The figure is reproduced from
reference[126].

phosphate to deliver sphingolipid growth factor FTY720 address these limitations of versatility. Microarray technol-
loaded in poly(lactide-co-glycolic acid) microspheres. They ogy has enabled the development of versatile platforms to
showed that sustained release of FTY720 from microspheres address the limitations of conventional assays[143–145].
improves vascularization and bone formation in critical size However, microarray technology utilizes 2D substrate system
cranial defects in Sprague Dawley rats. Furthermore, these to understand the cellular behavior[146–150]. They provide
growth factor releasing microgel/microsphere hybrids valuable insight about the differentiation of stem cells but
reduced the infiltration of inflammatory cells and promoted fails to mimic the tissue architecture[146,147,150]. Current
recruitment of bone progenitor cells after stimulation with focus of microarray based technology for bone regeneration
BMP-2[62]. Therefore, injectable microgel has the potential is the development of 3D miniaturized cellular platforms.
to regenerate the bone defects which are irregularly shaped, For instance, Microgel arrays are utilized as 3D miniaturized
even in deeper planes and complex in nature. cellular platforms for off-the-shelf-tissue studies. The micro-
gel environments were composed in a manner to resemble
native vascularized and bone marrow tissue architectures.
4.3. Microgel as 3D microarrays
The complex heterogeneous tissue like structure lead to the
Microenvironmental cues determine the differentiation of localized cell spreading and osteogenic differentiation of
stem cells into the specialized cell types. These microenvir- hMSCs[117]. In another study, Gelatin methacrylate/nHA
onmental cues are governed by the cell-cell interactions, photocrosslinkable microgel arrays encapsulating human
cell-ECM interactions[131–133], soluble factors[131], matrix periodontal ligament stem cells were used for in vitro and in
topography and stiffness[132–135]. These cues act in a syner- vivo studies. The microgel arrays facilitated the in vitro cell
gistic manner to determine the fate of stem cells to form the viability, proliferation and osteogenic differentiation of stem
functional tissue. Particularly, the three dimentionality of cells; and promoted bone formation in vivo[151]. In another
the ECM regulate the cell behavior and differentiation[136]. study, combinatorial screening of hMSCs differentiation
It is a well-known fact that the cellular functions substan- using 3D cell-laden gel microarray platform in response to
tially deviate on 3D scaffolds compared to 2D sub- ECM components and growth factors has been performed.
strates[137,138]. Therefore, it is crucial to develop 3D scaffolds It used an automated printing technique to fabricate the
with controlled presentation of the environmental cues for microgel units composed of hMSCs embedded in methacry-
studying the stem cell response. Development of the 3D lated gelatin (GE) along with ECM proteins and growth fac-
combinatorial microenvironments is an important step to tors. ECM proteins including fibronectin, laminin and
simultaneous provides various cues to the stem cells[139]. osteocalcin in different compositions along with BMP-2 and
Multiwell based combinatorial microenvironments have BMP-5 were used for microgel fabrication. The technique
been well explored for screening the fate of stem utilizes 1000 fold less materials and cells compared to con-
cells[140–142]. In these techniques, stem cell encapsulated ventional multiwell based assays[152]. Figure 9a shows the
ECM rich hydrogels are directly added to the regular multi- schematic of the microarray fabrication process. Cell laden
well plates[140]. In other approaches, polymeric solutions are GE hydrogel with different combination of ECM proteins
deposited into the multiwell plates to generate the scaffold are robotically spotted in the form of microgels. The depos-
arrays[141,142]. Despite of wide applications, these multiwell ited microgels are then crosslinked by ultraviolet light
based platforms face several limitations due to high reagent exposure to develop cell-laden constructs. Figure 9b shows
cost which leads to the less versatile technology[139]. the different combinations used for the microgel fabrication.
Therefore, development of cost-efficient platforms can Florescent imaging Figures 9c and 9d revels the ECM
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 11

Figure 9. Schematic of the microgel based microarray fabrication. (a) GE hydrogel containing hMSCs with different combination of ECM proteins are spotted robot-
ically. The microgel spots are crosslimked by UV light exposure. (b) shows the various combination of ECM proteins and the media formulations used to conduct
the experiments. (c) shows the florescence images of the ECM proteins used in the fabrication of microgel array. (d) shows the florescence images to depict the via-
bility of the cells with colour-diagram of percent cell viability. The images are reproduced from reference[152].

protein combinations used for the microgel fabrication and mesenchymal stem cells. No direct study was performed for
the viability of the encapsulated cells in the microgels bone tissue engineering; however these devices were utilized
respectively. for stem cells expansion and growth for tissue engineering
Different applications of microgels for bone tissue engin- applications[154,155]. For instance, Cytodex 3 is commercially
eering also have been summarized in Table 2. available micro-carrier from GE healthcare. These micro-
carriers are dextran matrix coated with collagen for cell
attachment. It was observed that 3000 micro-carriers per ml
5. Microgel: commercial aspect suspension with 5X cell density to micro-carrier showed
The current research on microgel is majorly focused on optimum hMSCs expansion. The maximum yield was
novel fabrication methods and applications in bone tissue obtained at 10 days of culture having 20-fold increased cell
engineering. There are very few microgel devices available per micro-carrier[156]. Microgel based devices have been
commercially for bone tissue engineering applications. studied in vivo on small animal models for bone regener-
Microgels have been available as micro-carriers, which can ation applications[157,158]. For growth factor/drug delivery,
be used for expansion culture of cells in vitro[153]. These none of microgel device is been available commercially or
micro-carriers were explored for large scale production of been extended to clinical phase till date. Similarly, the
bioactive compounds and bioreactor cultures of human
12 L. K. MEENA ET AL.

Table 2. Various applications of microgels for bone tissue engineering.


Device type Composition Application
Microgels Heparin, hyaluronan Controlled release of BMP-2 for bone regeneration[57]
Microgels reinforced hydrogels Glycidyl methacrylate, heparin, hyaluronan, Sustained delivery of BMP-2 for bone regeneration[78]
poly(N-isopropylacrylamide-co-acrylic acid), PLGA, BioglassV
R
Microgels coated scaffold Initial rapid followed by sustained release of vancomycin for
prevent the infection in bone healing[67]
Microgels polyvinyl alcohol-borax Controlled release of vancomycin for prevent the infection in
bone healing[95]
Microgels based scaffold Collagen, poly(L-Lactide) Controlled release of dexamethsone for osteoblasts differenti-
ation of MSCs[77]
Microgels Poly(N-isopropylacrylamide-co-acrylic acid) Controlled release of dexamethsone and vancomycin[101]
Microgels Alginate, RGD peptide Scaffold for MSCs culture and delivery[115]
Microgels Gelatin, methacrylate, thiolated hyaluronic acid Encapsulation and delivery of hMSCs[117]

microgel loaded hybrid scaffolds also not explored clinically Looking forward, recent advancement in microgel based
to date. technologies could enrich translational bone tissue engineer-
The literature suggests that still there is no such device ing field and advance the current bone rejuven-
available exclusively for bone tissue engineering applications. ation strategies.
However, considering the numbers of in vitro and in vivo
studies significant growth and translation of devices can be
anticipated. Acknowledgements
The author acknowledges the University Grant Commission (Govt of
India) for a senior research fellowship.
6. Conclusions and future perspectives
Bone replacement is becoming the most widely transplant- Disclosure statement
ation in the world, utilizing various types of innovative
materials and scaffolds. In recent years, microgels have been No potential conflict of interest was reported by the authors.
explored as potential scaffolds for bone tissue engineering.
The soft and elastic nature of microgels imparts the inject- Funding
ability of scaffolds eliminate the dependency of open surgical
procedures, which greatly expand its applicability in bone The RV acknowledges the GSBTM [GSBTM/MD/Projects/SSA/1431/
2014-15] for providing the financial support.
tissue engineering. Its structural and physic-chemical fea-
tures provide the adhesive and encapsulation properties,
which allow them to utilize for osteoconductive molecules References
or progenitor cells delivery. Moreover, small size can widen
[1] Campana, V.; Milano, G.; Pagano, E.; Barba, M.; Cicione, C.;
the fabrication parameters to make hybrid scaffolds. Taken Salonna, G.; Lattanzi, W.; Logroscino, G. Bone Substitutes in
all together these third generation devices have not merely Orthopaedic Surgery: From Basic Science to Clinical Practice.
scaffolding the bone tissue, rather dynamically participate in J. Mater. Sci: Mater. Med. 2014, 25, 2445–2461. doi:10.1007/
and enhance the bone regeneration. Which have signifi- s10856-014-5240-2.
cantly advanced the bone transplantation strategies. [2] Liu, X.; Ma, P. X. Polymeric Scaffolds for Bone Tissue
Engineering. Ann. Biomed. Eng. 2004, 32, 477–486. doi:
However, this field is continuously growing, having limited
doi.org/10.1023/B:ABME.000.
commercially developed devices, indicates further require- [3] Seal, B. L.; Otero, T. C.; Panitch, A. Polymeric Biomaterials
ment of research and development for clinical efficacy and for Tissue and Organ Regeneration. Mater. Sci. Eng. R Rep.
commercial scale-up. 2001, 34, 147–230. doi:10.1016/S0927-796X(01)00035-3.
In addition, these scaffolds have enormous potential to [4] Bolgen, N.; Yang, Y.; Korkusuz, P.; Guzel, E.; El Haj, A. J.;
Piskin, E. Three-Dimensional Ingrowth of Bone Cells within
explore in several ways to design and develop for specific
Biodegradable Cryogel Scaffolds in Bioreactors at Different
bone tissue engineering applications as a future perspective. Regimes. Tissue. Eng. Part A. 2008, 14, 1743–1750. doi:
In particular, bone regeneration is a multifaceted process 10.1089/ten.tea.2007.0277.
comprising involvement of numerous molecules and com- [5] Ensrud, K. E. Epidemiology of Fracture Risk with Advancing
plex cell-matrix interactions orchestrated at a time. Age. J. Gerontol. A Biol. Sci. Med. Sci. 2013, 68, 1236–1242.
Recapitulating this multipart process is essential for achiev- doi:10.1093/gerona/glt092.
[6] GlobalData. Medipoint: Bone Grafts and Substitutes – Global
ing accurate bone restoration. The biological prospect could Analysis and Market Forecasts. 2014.
include the involvement of multiple growth factor and cells [7] Kolk, A.; Handschel, J.; Drescher, W.; Rothamel, D.; Kloss, F.;
in to elicit multi-scale signaling pathways. Furthermore, a Blessmann, M.; Heiland, M.; Wolff, K. D.; Smeets, R. Current
multilayered microgel would be designed to incorporate Trends and Future Perspectives of Bone Substitute Materials –
more than one molecule/growth factor in a single device from Space Holders to Innovative Biomaterials.
J. Craniomaxillofac. Surg. 2012, 40, 706–718. doi:10.1016/
having sequential release. This would be helpful to stimulate
j.jcms.2012.01.002.
the different stages of bone regeneration. A desired device [8] Giannoudis, P. V.; Einhorn, T. A.; Marsh, D. Fracture
could also be envisioned as stimuli responsive device for Healing: The Diamond Concept. Injury 2007, 38, S3–S6. doi:
release of signaling molecules based on requirement. 10.1016/S0020-1383(08)70003-2.
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 13

[9] Giannoudis, P. V.; Einhorn, T. A.; Schmidmaier, G.; Marsh, D. Scaffolds for Bone Tissue Engineering. J. Biomed. Nanotechnol.
The Diamond Concept–Open Questions. Injury 2008, 39, 2013, 9, 719–731. doi:10.1166/jbn.2013.1574.
S5–S8. doi:10.1016/S0020-1383(08)70010-X. [26] Schneider, R. K.; Puellen, A.; Kramann, R.; Raupach, K.;
[10] Hutmacher, D. W.; Schantz, J. T.; Lam, C. X.; Tan, K. C.; Lim, Bornemann, J.; Knuechel, R.; Perez-Bouza, A.; Neuss, S. The
T. C. State of the Art and Future Directions of Scaffold-Based Osteogenic Differentiation of Adult Bone Marrow and
Bone Engineering from a Biomaterials Perspective. J. Tissue Perinatal Umbilical Mesenchymal Stem Cells and Matrix
Eng. Regen. Med. 2007, 1, 245–260. doi:10.1002/term.24. Remodelling in Three-Dimensional Collagen Scaffolds.
[11] Woodruff, M. A.; Lange, C.; Reichert, J.; Berner, A.; Chen, F.; Biomaterials 2010, 31, 467–480. doi:10.1016/j.biomaterials.
Fratzl, P.; Schantz, J.-T.; Hutmacher, D. W. Bone Tissue 2009.09.059.
Engineering: From Bench to Bedside. Mater. Today 2012, 15, [27] Suarez-Gonzalez, D.; Barnhart, K.; Saito, E.; Vanderby, R.; Jr.;
430–435. doi:10.1016/S1369-7021(12)70194-3. Hollister, S. J.; Murphy, W. L. Controlled Nucleation of
[12] Glombitza, M.; Steinhausen, E. Treatment of Chronic Hydroxyapatite on Alginate Scaffolds for Stem Cell-Based
Osteomyelitis of the Lower Limb with a New Injectable, Bone Tissue Engineering. J. Biomed. Mater. Res. A 2010, 95,
Vancomycin-Loaded, Calcium Sulfate/Hydroxyapatite 222–234. doi:10.1002/jbm.a.32833.
Composite. Orthop. Proc. 2016, 98-B, 39. doi:10.1302/1358- [28] Zhou, H.; Xu, H. H. The Fast Release of Stem Cells from
992x.98bsupp_23.ebjis2016-039. Alginate-Fibrin Microbeads in Injectable Scaffolds for Bone
[13] Schwartz, C.; Bordei, R. Biphasic Phospho-Calcium Ceramics Tissue Engineering. Biomaterials 2011, 32, 7503–7513. doi:
Used as Bone Substitutes Are Efficient in the Management of 10.1016/j.biomaterials.2011.06.045.
Severe Acetabular Bone Loss in Revision Total Hip [29] Wang, L.; Stegemann, J. P. Thermogelling Chitosan and
Arthroplasties. Eur. J. Orthop. Surg. Traumatol. 2005, 15, Collagen Composite Hydrogels Initiated with Beta-
191–196. doi:10.1007/s00590-005-0244-8. Glycerophosphate for Bone Tissue Engineering. Biomaterials
[14] Scheer, J. H.; Adolfsson, L. E. Tricalcium Phosphate Bone 2010, 31, 3976–3985. doi:10.1016/j.biomaterials.2010.01.131.
Substitute in Corrective Osteotomy of the Distal Radius. [30] Thein-Han, W.; Xu, H. H. Collagen-Calcium Phosphate
Injury 2009, 40, 262–267. doi:10.1016/j.injury.2008.08.013. Cement Scaffolds Seeded with Umbilical Cord Stem Cells for
[15] Saikia, K. C.; Bhattacharya, T. D.; Bhuyan, S. K.; Talukdar, Bone Tissue Engineering. Tissue Eng. Part A 2011, 17,
D. J.; Saikia, S. P.; Jitesh, P. Calcium Phosphate Ceramics as 2943–2954. doi:10.1089/ten.tea.2010.0674.
Bone Graft Substitutes in Filling Bone Tumor Defects. Indian [31] Lopez-Noriega, A.; Quinlan, E.; Celikkin, N.; O’Brien, F. J.
J. Orthop. 2008, 42, 169–172. doi:10.4103/0019-5413.39588. Incorporation of Polymeric Microparticles into Collagen-
[16] Heise, U.; Osborn, J. F.; Duwe, F. Hydroxyapatite Ceramic as Hydroxyapatite Scaffolds for the Delivery of a Pro-Osteogenic
a Bone Substitute. Int. Orthop. 1990, 14, 329–338. doi:10.1007/ Peptide for Bone Tissue Engineering. APL Materials 2015, 3,
BF00178768. 014910. doi:10.1063/1.4902833.
[17] Whitehouse, M. R.; Blom, A. W. The Use of Ceramics as Bone [32] P, G. J.; Frank, L.; Sonia, P.; O. B. F, J. Collagen-
Substitutes in Revision Hip Arthroplasty. Materials 2009, 2, Hydroxyapatite Composite Scaffolds as an Alternative to
1895–1907. doi:10.3390/ma2041895. Autogenous Bone Grafts. Orthop. Proc. 2012, 94-B, 8. doi:
[18] Lindfors, N. C.; Hyvonen, P.; Nyyssonen, M.; Kirjavainen, M.; 10.1302/1358-992x.94bsupp_xxxviii.coa2011-008.
Kankare, J.; Gullichsen, E.; Salo, J. Bioactive Glass S53p4 as [33] Villa, M. M.; Wang, L.; Huang, J.; Rowe, D. W.; Wei, M. Bone
Bone Graft Substitute in Treatment of Osteomyelitis. Bone Tissue Engineering with a Collagen–Hydroxyapatite Scaffold
2010, 47, 212–218. doi:10.1016/j.bone.2010.05.030. and Culture Expanded Bone Marrow Stromal Cells. J. Biomed.
[19] Wheeler, D. L.; Eschbach, E. J.; Hoellrich, R. G.; Montfort, Mater. Res. B. 2015, 103, 243–253. doi:10.1002/jbm.b.33225.
M. J.; Chamberland, D. L. Assessment of Resorbable Bioactive [34] Sarikaya, B.; Aydin, H. M. Collagen/Beta-Tricalcium
Material for Grafting of Critical-Size Cancellous Defects. Phosphate Based Synthetic Bone Grafts via Dehydrothermal
J. Orthop. Res. 2000, 18, 140–148. doi:10.1002/jor.1100180120. Processing. Biomed. Res. Int. 2015, 2015, 1–9. doi:10.1155/
[20] Zwingenberger, S.; Nich, C.; Valladares, R. D.; Yao, Z.; 2015/576532.
Stiehler, M.; Goodman, S. B. Recommendations and [35] Baheiraei, N.; Nourani, M. R.; Mortazavi, S. M. J.; Movahedin,
Considerations for the Use of Biologics in Orthopedic Surgery. M.; Eyni, H.; Bagheri, F.; Norahan, M. H. Development of a
BioDrugs 2012, 26, 245–256. doi:10.2165/11631680-000000000- Bioactive Porous Collagen/B-Tricalcium Phosphate Bone Graft
00000. Assisting Rapid Vascularization for Bone Tissue Engineering
[21] Buchanan, F.; Gallagher, L.; Jack, V.; Dunne, N. Short-Fibre Applications. J. Biomed. Mater. Res. A 2018, 106, 73–85. doi:
Reinforcement of Calcium Phosphate Bone Cement. Proc. Inst. doi:10.1002/jbm.a.36207.
Mech. Eng. H. 2007, 221, 203–211. doi:10.1243/ [36] Diogo, G. S.; Gaspar, V. M.; Serra, I. R.; Fradique, R.; Correia,
09544119JEIM235. I. J. Manufacture of B-Tcp/Alginate Scaffolds through a
[22] Xu, H. H. K.; Wang, P.; Wang, L.; Bao, C.; Chen, Q.; Weir, Fab@Home Model for Application in Bone Tissue
M. D.; Chow, L. C.; Zhao, L.; Zhou, X.; Reynolds, M. A. Engineering. Biofabrication 2014, 6, 025001. doi:10.1088/1758-
Calcium Phosphate Cements for Bone Engineering and Their 5082/6/2/025001.
Biological Properties. Bone Res. 2017, 5, 17056. doi:10.1038/ [37] Fang, X.; Lei, L.; Jiang, T.; Chen, Y.; Kang, Y. Injectable
boneres.2017.56. Thermosensitive Alginate/B-Tricalcium Phosphate/Aspirin
[23] Verron, E.; Pissonnier, M. L.; Lesoeur, J.; Schnitzler, V.; Fellah, Hydrogels for Bone Augmentation. J. Biomed. Mater. Res. B
B. H.; Pascal-Moussellard, H.; Pilet, P.; Gauthier, O.; Bouler, 2018, 106, 1739–1751. doi:10.1002/jbm.b.33982.
J. M. Vertebroplasty Using Bisphosphonate-Loaded Calcium [38] Miguel, C.; Jorge, R.; In^es, P.; Barbara, G.; Manuel, P.; Claus,
Phosphate Cement in a Standardized Vertebral Body Bone M.; J€urgen, G.; Andrea, E.; Elke, V. Fabrication of Individual
Defect in an Osteoporotic Sheep Model. Acta Biomater. 2014, Alginate-tcp scaffolds for Bone Tissue Engineering by Means
10, 4887–4895. doi:10.1016/j.actbio.2014.07.012. of Powder Printing. Biofabrication 2015, 7, 015004. doi:
[24] Hiraoka, Y.; Kimura, Y.; Ueda, H.; Tabata, Y. Fabrication and 10.1088/1758-5090/7/1/015004.
Biocompatibility of Collagen Sponge Reinforced with [39] Kanasan, N.; Adzila, S.; Suid, M. S.; Gurubaran, P. Preparation
Poly(Glycolic Acid) Fiber. Tissue Eng. 2003, 9, 1101–1112. doi: and Characterization of Hydroxyapatite/Sodium Alginate
10.1089/10763270360728017. Biocomposites for Bone Implant Application. AIP Conf. Proc.
[25] Aravamudhan, A.; Ramos, D. M.; Nip, J.; Harmon, M. D.; 2016, 1756, 020006. doi:10.1063/1.4958749.
James, R.; Deng, M.; Laurencin, C. T.; Yu, X.; Kumbar, S. G. [40] Lin, H.-R.; Yeh, Y.-J. Porous Alginate/Hydroxyapatite
Cellulose and Collagen Derived Micro-Nano Structured Composite Scaffolds for Bone Tissue Engineering: Preparation,
14 L. K. MEENA ET AL.

Characterization, and in Vitro Studies. J. Biomed. Mater. Res. Microgels for Injectable Tissue Constructs. Biomacromolecules
2004, 71B, 52–65. doi:10.1002/jbm.b.30065. 2014, 15, 283–290. doi:10.1021/bm401533y.
[41] Einhorn, T. A.; Gerstenfeld, L. C. Fracture Healing: [60] Moraes, C.; Simon, A. B.; Putnam, A. J.; Takayama, S.
Mechanisms and Interventions. Nat. Rev. Rheumatol. 2015, 11, Aqueous Two-Phase Printing of Cell-Containing Contractile
45–54. doi:10.1038/nrrheum.2014.164. Collagen Microgels. Biomaterials 2013, 34, 9623–9631. doi:
[42] Huang, Y. H.; Polimeni, G.; Qahash, M.; Wikesjo, U. M. Bone 10.1016/j.biomaterials.2013.08.046.
Morphogenetic Proteins and Osseointegration: Current [61] Siltanen, C.; Yaghoobi, M.; Haque, A.; You, J.; Lowen, J.;
Knowledge – Future Possibilities. Periodontol 2000 2008, 47, Soleimani, M.; Revzin, A. Microfluidic Fabrication of Bioactive
206–223. doi:10.1111/j.1600-0757.2007.00240.x. Microgels for Rapid Formation and Enhanced Differentiation
[43] Williams, D. F. On the Mechanisms of Biocompatibility. of Stem Cell Spheroids. Acta Biomater. 2016, 34, 125–132. doi:
Biomaterials 2008, 29, 2941–2953. doi:10.1016/ 10.1016/j.actbio.2016.01.012.
j.biomaterials.2008.04.023. [62] Das, A.; Barker, D. A.; Wang, T.; Lau, C. M.; Lin, Y.;
[44] Lichte, P.; Pape, H. C.; Pufe, T.; Kobbe, P.; Fischer, H. Botchwey, E. A. Delivery of Bioactive Lipids from Composite
Scaffolds for Bone Healing: Concepts, Materials and Evidence. Microgel-Microsphere Injectable Scaffolds Enhances Stem Cell
Injury 2011, 42, 569–573. doi:10.1016/j.injury.2011.03.033. Recruitment and Skeletal Repair. PLoS One. 2014, 9, 1–11.
[45] Rouwkema, J.; Rivron, N. C.; van Blitterswijk, C. A. doi:10.1371/journal.pone.0101276.
Vascularization in Tissue Engineering. Trends Biotechnol. [63] Saunders, B. R.; Vincent, B. Microgel Particles as Model
2008, 26, 434–441. doi:10.1016/j.tibtech.2008.04.009. Colloids: Theory, Properties and Applications. Adv.
[46] Hutmacher, D. W. Scaffolds in Tissue Engineering Bone and Colloid Interface Sci 1999, 80, 1–25. doi:10.1016/S0001-
Cartilage. Biomaterials 2000, 21, 2529–2543. doi:10.1016/ 8686(98)00071-2.
S0142-9612(00)00121-6. [64] Reufer, M.; Diaz-Leyva, P.; Lynch, I.; Scheffold, F.
[47] Olszta, M. J.; Cheng, X.; Jee, S. S.; Kumar, R.; Kim, Y.-Y.; Temperature-Sensitive Poly(N-Isopropyl-Acrylamide) Microgel
Kaufman, M. J.; Douglas, E. P.; Gower, L. B. Bone Structure Particles: A Light Scattering Study. Eur. Phys. J. E Soft Matter.
and Formation: A New Perspective. Mater. Sci. Eng. R Rep. 2009, 28, 165–171. doi:10.1140/epje/i2008-10387-2.
2007, 58, 77–116. doi:10.1016/j.mser.2007.05.001. [65] Riederer, M. S.; Requist, B. D.; Payne, K. A.; Way, J. D.;
[48] Staudinger, H.; Husemann, E. Uber € Hochpolymere Krebs, M. D. Injectable and Microporous Scaffold of Densely-
Verbindungen, 116. Mitteil.: Uber€ Das Begrenzt Quellbare Packed, Growth Factor-Encapsulating Chitosan Microgels.
Poly-Styrol. Ber. Dtsch. Chem. Ges. A/B. 1935, 68, 1618–1634. Carbohydr. Polym. 2016, 152, 792–801. doi:10.1016/
doi:10.1002/cber.19350680841. j.carbpol.2016.07.052.
[49] Baker, W. O. Microgel, a New Macromolecule. Ind. Eng. [66] Kim, P. H.; Yim, H. G.; Choi, Y. J.; Kang, B. J.; Kim, J.; Kwon,
S. M.; Kim, B. S.; Hwang, N. S.; Cho, J. Y. Injectable
Chem. 1949, 41, 511–520. doi:10.1021/ie50471a016.
[50] Dickinson, E. Microgels — an Alternative Colloidal Ingredient Multifunctional Microgel Encapsulating Outgrowth
Endothelial Cells and Growth Factors for Enhanced
for Stabilization of Food Emulsions. Trends Food Sci. Technol.
Neovascularization. J. Control. Release. 2014, 187, 1–13. doi:
2015, 43, 178–188. doi:10.1016/j.tifs.2015.02.006.
10.1016/j.jconrel.2014.05.010.
[51] Omari, A.; Tabary, R.; Rousseau, D.; Calderon, F. L.; Monteil,
[67] Olalde, B.; Garmendia, N.; Saez-Martinez, V.; Argarate, N.;
J.; Chauveteau, G. Soft Water-Soluble Microgel Dispersions:
Nooeaid, P.; Morin, F.; Boccaccini, A. R. Multifunctional
Structure and Rheology. J. Colloid Interface Sci. 2006, 302,
Bioactive Glass Scaffolds Coated with Layers of Poly(D,L-
537–546. doi:10.1016/j.jcis.2006.07.006.
Lactide-Co-Glycolide) and Poly(N-Isopropylacrylamide-Co-
[52] Crowther, H. M.; Vincent, B. Swelling Behavior of Poly- N-
Acrylic Acid) Microgels Loaded with Vancomycin. Mater. Sci.
Isopropylacrylamide Microgel Particles in Alcoholic Solutions.
Eng. C Mater. Biol. Appl. 2013, 33, 3760–3767. doi:10.1016/
Colloid. Polym. Sci. 1998, 276, 46–51. doi:10.1007/
j.msec.2013.05.002.
s003960050207. [68] Foster, G. A.; Headen, D. M.; Gonzalez-Garcia, C.; Salmeron-
[53] Dai, Z.; Ngai, T. Microgel Particles: The Structure-Property Sanchez, M.; Shirwan, H.; Garcia, A. J. Protease-Degradable
Relationships and Their Biomedical Applications. J. Polym. Sci. Microgels for Protein Delivery for Vascularization.
Part A: Polym. Chem. 2013, 51, 2995–3003. doi:10.1002/ Biomaterials 2017, 113, 170–175. doi:10.1016/
pola.26698. j.biomaterials.2016.10.044.
[54] Hoare, T.; Pelton, R. Dimensionless Plot Analysis: A New [69] Panda, P.; Ali, S.; Lo, E.; Chung, B. G.; Hatton, T. A.;
Way to Analyze Functionalized Microgels. J. Colloid Interface Khademhosseini, A.; Doyle, P. S. Stop-Flow Lithography to
Sci. 2006, 303, 109–116. doi:10.1016/j.jcis.2006.07.047. Generate Cell-Laden Microgel Particles. Lab Chip. 2008, 8,
[55] Malmsten, M.; Bysell, H.; Hansson, P. Biomacromolecules in 1056–1061. doi:10.1039/b804234a.
Microgels — Opportunities and Challenges for Drug Delivery. [70] Hamidi, M.; Azadi, A.; Rafiei, P. Hydrogel Nanoparticles in
Curr. Opin. Colloid Interface Sci. 2010, 15, 435–444. doi: Drug Delivery. Adv. Drug Del. Rev. 2008, 60, 1638–1649. doi:
10.1016/j.cocis.2010.05.016. 10.1016/j.addr.2008.08.002.
[56] Bai, X.; Gao, M.; Syed, S.; Zhuang, J.; Xu, X.; Zhang, X.-Q. [71] Peppas, N. A.; Bures, P.; Leobandung, W.; Ichikawa, H.
Bioactive Hydrogels for Bone Regeneration. Bioact. Mater. Hydrogels in Pharmaceutical Formulations. Eur. J.
2018, 3, 401–417. doi:10.1016/j.bioactmat.2018.05.006. Pharm. Biopharm. 2000, 50, 27–46. doi:10.1016/S0939-
[57] Zhao, J.; Luo, C.; Chen, Y.; Wu, D.; Shen, C.; Han, W.; Tu, 6411(00)00090-4.
M.; Zeng, R. Preparation, Structure and Bmp-2 Controlled [72] Freemont, T. J.; Saunders, B. R. Ph-Responsive Microgel
Release of Heparin-Conjugated Hyaluronan Microgels. Dispersions for Repairing Damaged Load-Bearing Soft Tissue.
Carbohydr. Polym. 2011, 86, 806–811. doi:10.1016/ Soft Matter. 2008, 4, 919–924. doi:10.1039/b718441g.
j.carbpol.2011.05.026. [73] Li, F.; Truong, V. X.; Thissen, H.; Frith, J. E.; Forsythe, J. S.
[58] Zuo, Q.; Lu, J.; Hong, A.; Zhong, D.; Xie, S.; Liu, Q.; Huang, Microfluidic Encapsulation of Human Mesenchymal Stem
Y.; Shi, Y.; He, L.; Xue, W. Preparation and Characterization Cells for Articular Cartilage Tissue Regeneration. ACS Appl.
of Pem-Coated Alginate Microgels for Controlled Release of Mater. Interfaces 2017, 9, 8589–8601. doi:10.1021/
Protein. Biomed. Mater. 2012, 7, 1–12. doi:10.1088/1748-6041/ acsami.7b00728.
7/3/035012. [74] Xie, J.; Ma, B.; Michael, P. L. Fabrication of Novel 3d
[59] Cha, C.; Oh, J.; Kim, K.; Qiu, Y.; Joh, M.; Shin, S. R.; Wang, Nanofiber Scaffolds with Anisotropic Property and Regular
X.; Camci-Unal, G.; Wan, K. T.; Liao, R.; Khademhosseini, A. Pores and Their Potential Applications. Adv. Healthc. Mater.
Microfluidics-Assisted Fabrication of Gelatin-Silica Core-Shell 2012, 1, 674–678. doi:10.1002/adhm.201200100.
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 15

[75] Thibault, R. A.; Mikos, A. G.; Kasper, F. K. Scaffold/ [91] Oh, J. K.; Lee, D. I.; Park, J. M. Biopolymer-Based Microgels/
Extracellular Matrix Hybrid Constructs for Bone-Tissue Nanogels for Drug Delivery Applications. Prog. Polym. Sci.
Engineering. Adv. Healthc. Mater. 2013, 2, 13–24. doi:10.1002/ 2009, 34, 1261–1282. doi:10.1016/j.progpolymsci.2009.08.001.
adhm.201200209. [92] Yang, X.; Kim, J. C. Novel Ph-Sensitive Microgels Prepared
[76] Jhala, D.; Rather, H.; Vasita, R. Polycaprolactone-Chitosan Using Salt Bridge. Int. J. Pharm. 2010, 388, 58–63. doi:
Nanofibers Influence Cell Morphology to Induce Early 10.1016/j.ijpharm.2009.12.035.
Osteogenic Differentiation. Biomater. Sci. 2016, 4, 1584–1595. [93] Yoon, J. J.; Chung, H. J.; Lee, H. J.; Park, T. G. Heparin-
doi:10.1039/C6BM00492J. Immobilized Biodegradable Scaffolds for Local and Sustained
[77] Nanda, H. S.; Nakamoto, T.; Chen, S.; Cai, R.; Kawazoe, N.; Release of Angiogenic Growth Factor. J. Biomed. Mater. Res. A
Chen, G. Collagen Microgel-Assisted Dexamethasone Release 2006, 79, 934–942. doi:10.1002/jbm.a.30843.
from Plla-Collagen Hybrid Scaffolds of Controlled Pore [94] Pike, D. B.; Cai, S.; Pomraning, K. R.; Firpo, M. A.; Fisher,
Structure for Osteogenic Differentiation of Mesenchymal Stem R. J.; Shu, X. Z.; Prestwich, G. D.; Peattie, R. A. Heparin-
Cells. J. Biomater. Sci. Polym. Ed. 2014, 25, 1374–1386. doi: Regulated Release of Growth Factors in Vitro and Angiogenic
Response In Vivo to Implanted Hyaluronan Hydrogels
10.1080/09205063.2014.938980.
Containing VEGF and bFGF. Biomaterials 2006, 27,
[78] Yuan, X.; Li, R.; Rao, Z.; Li, H.; Rong, J.; Tu, M.; Zeng, R.;
5242–5251. doi:10.1016/j.biomaterials.2006.05.018.
Zhao, J. Synthesis and Characterization of a Dually
[95] Lv, J.; Li, X.; Yin, H.; Wang, L.; Pei, Y.; Lv, X. Controlled
Crosslinked Heparin-Conjugated Ha Hydrogel for Bmp-2
Release of Vancomycin Hydrochloride from a Composite
Sustained Delivery. Int. J. Polym. Mater. Polym. Biomater.
Structure of Polymeric Films and Porous Fibers on Implants.
2016, 65, 928–937. doi:10.1080/00914037.2016.1180609. Chem. Eng. J. 2017, 325, 601–610. doi:10.1016/
[79] Murray, M. M.; Mankin, H. J.; Glowacki, J. The j.cej.2017.05.118.
Musculoskeletal System. In The Physiologic Basis of Surgery; [96] Meena, L. K.; Raval, P.; Kedaria, D.; Vasita, R. Study of Locust
O’Leary, J. P., Ed.; Lippincott, Williams & Wilkins: Bean Gum Reinforced Cyst-Chitosan and Oxidized Dextran
Philadelphia, PA, 2002; pp 577–595. Based Semi-Ipn Cryogel Dressing for Hemostatic Application.
[80] Tayalia, P.; Mooney, D. J. Controlled Growth Factor Delivery Bioact. Mater. 2018, 3, 370–384. doi:10.1016/
for Tissue Engineering. Adv. Mater. 2009, 21, 3269–3285. doi: j.bioactmat.2017.11.005.
10.1002/adma.200900241. [97] Sagbas, S.; Sahiner, N. Modifiable Natural Gum Based
[81] Xia, P.; Zhang, K.; Gong, Y.; Li, G.; Yan, S.; Yin, J. Injectable Microgel Capsules as Sustainable Drug Delivery Systems.
Stem Cell Laden Open Porous Microgels That Favor Carbohydr. Polym. 2018, 200, 128–136. doi:10.1016/
Adipogenesis: In Vitro and in Vivo Evaluation. ACS Appl. j.carbpol.2018.07.085.
Mater. Interfaces 2017, 9, 34751–34761. doi:10.1021/ [98] Johnson, E. N.; Burns, T. C.; Hayda, R. A.; Hospenthal, D. R.;
acsami.7b13065. Murray, C. K. Infectious Complications of Open Type Iii
[82] Hou, Y.; Xie, W.; Achazi, K.; Cuellar-Camacho, J. L.; Melzig, Tibial Fractures among Combat Casualties. Clin. Infect. Dis.
M. F.; Chen, W.; Haag, R. Injectable Degradable Pva 2007, 45, 409–415. doi:10.1086/520029.
Microgels Prepared by Microfluidic Technology for Controlled [99] de Lissovoy, G.; Fraeman, K.; Hutchins, V.; Murphy, D.; Song,
Osteogenic Differentiation of Mesenchymal Stem Cells. Acta D.; Vaughn, B. B. Surgical Site Infection: Incidence and
Biomater. 2018, 77, 28–37. doi:10.1016/j.actbio.2018.07.003. Impact on Hospital Utilization and Treatment Costs. Am. J.
[83] Griffin, D. R.; Weaver, W. M.; Scumpia, P. O.; Di Carlo, D.; Infect. Control 2009, 37, 387–397. doi:10.1016/
Segura, T. Accelerated Wound Healing by Injectable j.ajic.2008.12.010.
Microporous Gel Scaffolds Assembled from Annealed Building [100] Cross, W. W.; Swiontkowski, M. F. Treatment Principles in
Blocks. Nat. Mater. 2015, 14, 737–744. doi:10.1038/nmat4294. the Management of Open Fractures. Indian J. Orthop. 2008,
[84] Goustin, A. S.; Leof, E. B.; Shipley, G. D.; Moses, H. L. 42, 377–386. doi:10.4103/0019-5413.43373.
Growth Factors and Cancer. Cancer Res. 1986, 46, 1015–1029. [101] Saez-Martinez, V.; Argarate, N.; Morin, F.; Garagorri, N.
doi: Published March 1986. Comparative Study of Dexamethasone and Vancomycin
[85] Varkey, M.; Gittens, S. A.; Uludag, H. Growth Factor Delivery Release Behavior from Stimuli-Sensitive Microgel Aqueous
for Bone Tissue Repair: An Update. Expert Opin. Drug Deliv. Dispersions. J. Drug Deliv. Sci. Technol. 2012, 22, 313–316.
2004, 1, 19–36. doi:10.1517/17425247.1.1.19. doi:10.1016/S1773-2247(12)50053-5.
[86] Rasi Ghaemi, S.; Delalat, B.; Ceto, X.; Harding, F. J.; Tuke, J.; [102] Vogel, J. P.; Szalay, K.; Geiger, F.; Kramer, M.; Richter, W.;
Voelcker, N. H. Synergistic Influence of Collagen I and Bmp 2 Kasten, P. Platelet-Rich Plasma Improves Expansion of
Human Mesenchymal Stem Cells and Retains Differentiation
Drives Osteogenic Differentiation of Mesenchymal Stem Cells:
Capacity and in Vivo Bone Formation in Calcium Phosphate
A Cell Microarray Analysis. Acta Biomater. 2016, 34, 41–52.
Ceramics. Platelets 2006, 17, 462–469. doi:10.1080/
doi:10.1016/j.actbio.2015.07.027.
09537100600758867.
[87] Baker, M.; Carr, F. Pre-Clinical Considerations in the
[103] Smith, L. A.; Liu, X.; Hu, J.; Wang, P.; Ma, P. X. Enhancing
Assessment of Immunogenicity for Protein Therapeutics. Curr
Osteogenic Differentiation of Mouse Embryonic Stem Cells by
Drug Saf. 2010, 5, 308–313. doi:10.2174/157488610792246000.
Nanofibers. Tissue Eng. Part A. 2009, 15, 1855–1864. doi:
[88] Taghavi, C. E.; Lee, K. B.; He, W.; Keorochana, G.; Murray,
10.1089/ten.tea.2008.0227.
S. S.; Brochmann, E. J.; Uludag, H.; Behnam, K.; Wang, J. C. [104] Evans, N. D.; Swain, R. J.; Gentleman, E.; Gentleman, M. M.;
Bone Morphogenetic Protein Binding Peptide Mechanism and Stevens, M. M. Gene-expression Analysis Reveals That
Enhancement of Osteogenic Protein-1 Induced Bone Healing. Embryonic Stem Cells Cultured under Osteogenic Conditions
Spine 2010, 35, 2049–2056. doi:10.1097/BRS.0b013e3 Produce Mineral Non-specifically Compared to Marrow
181cc0220. Stromal Cells or Osteoblasts. Eur. Cell. Mater. 2012, 24,
[89] Lad, S. P.; Bagley, J. H.; Karikari, I. O.; Babu, R.; Ugiliweneza, 211–223. doi:10.22203/eCM.v024a15
B.; Kong, M.; Isaacs, R. E.; Bagley, C. A.; Gottfried, O. N.; [105] Fiedler, J.; Etzel, N.; Brenner, R. E. To Go or Not to Go:
Patil, C. G.; et al. Cancer after Spinal Fusion: The Role of Migration of Human Mesenchymal Progenitor Cells
Bone Morphogenetic Protein. Neurosurgery 2013, 73, 440–449. Stimulated by Isoforms of PDGF. J. Cell. Biochem. 2004, 93,
doi:10.1227/NEU.0000000000000018. 990–998. doi:10.1002/jcb.20219.
[90] Pelton, R. Temperature-Sensitive Aqueous Microgels. Adv. [106] Cenni, E.; Ciapetti, G.; Granchi, D.; Fotia, C.; Perut, F.; Giunti,
Colloid Interface Sci. 2000, 85, 1–33. doi:10.1016/S0001- A.; Baldini, N. Endothelial Cells Incubated with Platelet-Rich
8686(99)00023-8. Plasma Express PDGF-B and ICAM-1 and Induce Bone
16 L. K. MEENA ET AL.

Marrow Stromal Cell Migration. J. Orthop. Res. 2009, 27, [123] Boskey, A. L. Bone Composition: Relationship to Bone
1493–1498. doi:10.1002/jor.20896. Fragility and Antiosteoporotic Drug Effects. Bonekey Rep.
[107] Ren, L.; Ma, D.; Liu, B.; Li, J.; Chen, J.; Yang, D.; Gao, P. 2013, 2, 447. doi:10.1038/bonekey.2013.181.
Preparation of Three-Dimensional Vascularized MSC Cell [124] Rey, C.; Combes, C.; Drouet, C.; Glimcher, M. J. Bone
Sheet Constructs for Tissue Regeneration. Biomed. Res. Int. Mineral: Update on Chemical Composition and Structure.
2014, 2014, 1. doi:10.1155/2014/301279. Osteoporos. Int. 2009, 20, 1013–1021. doi:10.1007/s00198-009-
[108] Yu, H.; VandeVord, P. J.; Mao, L.; Matthew, H. W.; Wooley, 0860-y.
P. H.; Yang, S. Y. Improved Tissue-Engineered Bone [125] Birkholz, M. N.; Agrawal, G.; Bergmann, C.; Schroder, R.;
Regeneration by Endothelial Cell Mediated Vascularization. Lechner, S. J.; Pich, A.; Fischer, H. Calcium Phosphate/
Biomaterials 2009, 30, 508–517. doi:10.1016/ Microgel Composites for 3D Powderbed Printing of Ceramic
j.biomaterials.2008.09.047. Materials. Biomed. Tech. Biomed. Eng. 2016, 61, 267–279. doi:
[109] Asahara, T.; Masuda, H.; Takahashi, T.; Kalka, C.; Pastore, C.; 10.1515/bmt-2014-0141.
Silver, M.; Kearne, M.; Magner, M.; Isner, J. M. Bone Marrow [126] Dai, Z.; Shu, Y.; Wan, C.; Wu, C. Effects of Culture Substrate
Origin of Endothelial Progenitor Cells Responsible for Made of Poly(N-Isopropylacrylamide-Co-Acrylic Acid)
Postnatal Vasculogenesis in Physiological and Pathological Microgels on Osteogenic Differentiation of Mesenchymal Stem
Neovascularization. Circ. Res 1999, 85, 221–228. doi:10.1161/ Cells. Molecules 2016, 21, 1192. doi:10.3390/
01.RES.85.3.221. molecules21091192.
[110] Duttenhoefer, F.; Lara de Freitas, R.; Meury, T.; Loibl, M.; [127] Van Vlierberghe, S.; Dubruel, P.; Schacht, E. Biopolymer-
Benneker, L. M.; Richards, R. G.; Alini, M.; Verrier, S. 3D Based Hydrogels as Scaffolds for Tissue Engineering
Scaffolds Co-Seeded with Human Endothelial Progenitor and Applications: A Review. Biomacromolecules 2011, 12,
Mesenchymal Stem Cells: Evidence of Prevascularisation 1387–1408. doi:10.1021/bm200083n.
within 7 Days. Eur. Cell. Mater. 2013, 26, 49–64. discussion [128] Ni, P.; Ding, Q.; Fan, M.; Liao, J.; Qian, Z.; Luo, J.; Li, X.;
64-5. Luo, F.; Yang, Z.; Wei, Y. Injectable Thermosensitive PEG-
[111] van Ramshorst, J.; Rodrigo, S. F.; Schalij, M. J.; Beeres, S. L.; PCL-PEG Hydrogel/Acellular Bone Matrix Composite for
Bax, J. J.; Atsma, D. E. Bone Marrow Cell Injection for Bone Regeneration in Cranial Defects. Biomaterials 2014, 35,
Chronic Myocardial Ischemia: The past and the Future. J. 236–248. doi:10.1016/j.biomaterials.2013.10.016.
Cardiovasc. Trans. Res. 2011, 4, 182–191. doi:10.1007/s12265- [129] Hoare, T. R.; Kohane, D. S. Hydrogels in Drug Delivery:
010-9249-8. Progress and Challenges. Polymer 2008, 49, 1993–2007. doi:
[112] Slaughter, B. V.; Khurshid, S. S.; Fisher, O. Z.; 10.1016/j.polymer.2008.01.027.
Khademhosseini, A.; Peppas, N. A. Hydrogels in Regenerative [130] Arun Kumar, R.; Sivashanmugam, A.; Deepthi, S.; Iseki, S.;
Medicine. Adv. Mater. 2009, 21, 3307–3329. doi:10.1002/ Chennazhi, K. P.; Nair, S. V.; Jayakumar, R. Injectable Chitin-
adma.200802106. Poly(Epsilon-Caprolactone)/Nanohydroxyapatite Composite
[113] Nicodemus, G. D.; Bryant, S. J. Cell Encapsulation in Microgels Prepared by Simple Regeneration Technique for
Biodegradable Hydrogels for Tissue Engineering Applications. Bone Tissue Engineering. ACS Appl. Mater. Interfaces 2015, 7,
Tissue Eng. Part. B Rev. 2008, 14, 149–165. doi:10.1089/ 9399–9409. doi:10.1021/acsami.5b02685.
ten.teb.2007.0332. [131] Schuldiner, M.; Yanuka, O.; Itskovitz-Eldor, J.; Melton, D. A.;
[114] Jiang, W.; Li, M.; Chen, Z.; Leong, K. W. Cell-Laden Benvenisty, N. Effects of Eight Growth Factors on the
Microfluidic Microgels for Tissue Regeneration. Lab Chip Differentiation of Cells Derived from Human Embryonic Stem
2016, 16, 4482–4506. doi:10.1039/C6LC01193D. Cells. Proc. Natl. Acad. Sci. USA. 2000, 97, 11307–11312. doi:
[115] Schmidt, J. J.; Jeong, J.; Kong, H. The Interplay Between Cell 10.1073/pnas.97.21.11307.
Adhesion Cues and Curvature of Cell Adherent Alginate [132] Engler, A. J.; Sen, S.; Sweeney, H. L.; Discher, D. E. Matrix
Microgels in Multipotent Stem Cell Culture. Tissue Eng. Part Elasticity Directs Stem Cell Lineage Specification. Cell 2006,
A 2011, 17, 2687–2694. doi:10.1089/ten.tea.2010.0685. 126, 677–689. doi:10.1016/j.cell.2006.06.044.
[116] Tsuda, Y.; Morimoto, Y.; Takeuchi, S. Monodisperse Cell- [133] Discher, D. E. Matrix Elasticity Directs Stem Cell Lineage—
Encapsulating Peptide Microgel Beads for 3D Cell Culture. Soluble Factors That Limit Osteogenesis. Bone 2009, 44,
Langmuir 2010, 26, 2645–2649. doi:10.1021/la902827y. S205–S206. doi:10.1016/j.bone.2009.03.025.
[117] Guermani, E.; Shaki, H.; Mohanty, S.; Mehrali, M.; Arpanaei, [134] Dalby, M. J.; Gadegaard, N.; Tare, R.; Andar, A.; Riehle,
A.; Gaharwar, A. K.; Dolatshahi-Pirouz, A. Engineering M. O.; Herzyk, P.; Wilkinson, C. D.; Oreffo, R. O. The
Complex Tissue-Like Microgel Arrays for Evaluating Stem Control of Human Mesenchymal Cell Differentiation Using
Cell Differentiation. Sci. Rep. 2016, 6, 30445. doi:10.1038/ Nanoscale Symmetry and Disorder. Nat. Mater. 2007, 6,
srep30445. 997–1003. doi:10.1038/nmat2013.
[118] Hall, B. K.; Miyake, T. Divide, Accumulate, Differentiate: Cell [135] Markert, L. D. A.; Lovmand, J.; Foss, M.; Lauridsen, R. H.;
Condensation in Skeletal Development Revisited. Int. J. Dev. Lovmand, M.; F€ uchtbauer, E.-M.; F€ uchtbauer, A.; Wertz, K.;
Biol. 1995, 39, 881–893. Besenbacher, F.; Pedersen, F. S.; Duch, M. Identification of
[119] Kale, S.; Biermann, S.; Edwards, C.; Tarnowski, C.; Morris, M.; Distinct Topographical Surface Microstructures Favoring
Long, M. W. Three-Dimensional Cellular Development Is Either Undifferentiated Expansion or Differentiation of
Essential for Ex Vivo Formation of Human Bone. Nat. Murine Embryonic Stem Cells. Stem Cells Dev. 2009, 18,
Biotechnol. 2000, 18, 954–958. doi:10.1038/79439. 1331–1342. doi:10.1089/scd.2009.0114.
[120] Yamaguchi, Y.; Ohno, J.; Sato, A.; Kido, H.; Fukushima, T. [136] Lund, A. W.; Yener, B.; Stegemann, J. P.; Plopper, G. E. The
Mesenchymal Stem Cell Spheroids Exhibit Enhanced in-Vitro Natural and Engineered 3D Microenvironment as a Regulatory
and in-Vivo Osteoregenerative Potential. BMC Biotechnol. Cue During Stem Cell Fate Determination. Tissue Eng. Part B
2014, 14, 105. doi:10.1186/s12896-014-0105-9. Rev. 2009, 15, 371–380. doi:10.1089/ten.teb.2009.0270.
[121] Chan, H. F.; Zhang, Y.; Ho, Y.-P.; Chiu, Y.-L.; Jung, Y.; [137] Tian, X. F.; Heng, B. C.; Ge, Z.; Lu, K.; Rufaihah, A. J.; Fan,
Leong, K. W. Rapid Formation of Multicellular Spheroids in V. T.; Yeo, J. F.; Cao, T. Comparison of Osteogenesis of
Double-Emulsion Droplets with Controllable Human Embryonic Stem Cells within 2D and 3D Culture
Microenvironment. Sci. Rep. 2013, 3, 3462. doi:10.1038/ Systems. Scand. J. Clin. Lab. Invest. 2008, 68, 58–67. doi:
srep03462. 10.1080/00365510701466416.
[122] Boskey, A. L. Mineralization of Bones and Teeth. Elements [138] Jabbari, E. Role of Substrate Microstructure on Osteogenic
2007, 3, 385–391. doi:10.2113/GSELEMENTS.3.6.385. Differentiation of Mesenchymal Stem Cells. Conf. Proc. IEEE
INTERNATIONAL JOURNAL OF POLYMERIC MATERIALS AND POLYMERIC BIOMATERIALS 17

Eng. Med. Biol. Soc. 2010, 2010, 3543–3545. doi:10.1109/ Conditions for Stem, Primary and Rare Cell Populations. Nat.
IEMBS.2010.5627489. Protoc. 2012, 7, 703–717. doi:10.1038/nprot.2012.017.
[139] Mei, Y. Microarrayed Materials for Stem Cells. Mater. Today [150] Soen, Y.; Mori, A.; Palmer, T. D.; Brown, P. O. Exploring the
2012, 15, 444–452. doi:10.1016/S1369-7021(12)70196-7. Regulation of Human Neural Precursor Cell Differentiation
[140] Yang, F.; Cho, S.-W.; Son, S. M.; Hudson, S. P.; Bogatyrev, S.; Using Arrays of Signaling Microenvironments. Mol. Syst. Biol.
Keung, L.; Kohane, D. S.; Langer, R.; Anderson, D. G. 2006, 2, 37. doi:10.1038/msb4100076.
Combinatorial Extracellular Matrices for Human Embryonic [151] Chen, X.; Bai, S.; Li, B.; Liu, H.; Wu, G.; Liu, S.; Zhao, Y.
Stem Cell Differentiation in 3D. Biomacromolecules 2010, 11, Fabrication of Gelatin Methacrylate/Nanohydroxyapatite
1909–1914. doi:10.1021/bm100357t. Microgel Arrays for Periodontal Tissue Regeneration. Int J
[141] Yang, Y.; Bolikal, D.; Becker, M. L.; Kohn, J.; Zeiger, D. N.; Nanomed. 2016, 11, 4707–4718. doi:10.2147/IJN.S111701.
Simon, C. G. Combinatorial Polymer Scaffold Libraries for [152] Dolatshahi-Pirouz, A.; Nikkhah, M.; Gaharwar, A. K.; Hashmi,
Screening Cell-Biomaterial Interactions in 3D. Adv. Mater. B.; Guermani, E.; Aliabadi, H.; Camci-Unal, G.; Ferrante, T.;
2008, 20, 2037–2043. doi:doi:10.1002/adma.200702088. Foss, M.; Ingber, D. E.; et al. A Combinatorial Cell-Laden Gel
[142] Simon, C. G.; Jr.; Lin-Gibson, S. Combinatorial and High- Microarray for Inducing Osteogenic Differentiation of Human
Throughput Screening of Biomaterials. Adv. Mater. 2011, 23, Mesenchymal Stem Cells. Sci. Rep. 2014, 4, 3896. doi:10.1038/
369–387. doi:10.1002/adma.201001763. srep03896.
[143] Revzin, A.; Rajagopalan, P.; Tilles, A. W.; Berthiaume, F.; [153] Rafiq, Q. A.; Coopman, K.; Nienow, A. W.; Hewitt, C. J.
Yarmush, M. L.; Toner, M. Designing a Hepatocellular Systematic Microcarrier Screening and Agitated Culture
Conditions Improves Human Mesenchymal Stem Cell Yield in
Microenvironment with Protein Microarraying and
Bioreactors. Biotechnol. J. 2016, 11, 473–486. doi:10.1002/
Poly(Ethylene Glycol) Photolithography. Langmuir 2004, 20,
biot.201400862.
2999–3005. doi:10.1021/la035827w.
[154] dos Santos, F.; Andrade, P. Z.; Eibes, G.; da Silva, C. L.;
[144] Falsey, J. R.; Renil, M.; Park, S.; Li, S.; Lam, K. S. Peptide and
Cabral, J. M. Ex Vivo Expansion of Human Mesenchymal
Small Molecule Microarray for High Throughput Cell
Stem Cells on Microcarriers. Methods Mol. Biol. 2011, 698,
Adhesion and Functional Assays. Bioconjug. Chem. 2001, 12,
189–198. doi:10.1007/978-1-60761-999-4_15.
346–353. doi:10.1021/bc000141q. [155] Rafiq, Q. A.; Brosnan, K. M.; Coopman, K.; Nienow, A. W.;
[145] Hook, A. L.; Chang, C. Y.; Yang, J.; Luckett, J.; Cockayne, A.; Hewitt, C. J. Culture of Human Mesenchymal Stem Cells on
Atkinson, S.; Mei, Y.; Bayston, R.; Irvine, D. J.; Langer, R.; Microcarriers in a 5 L Stirred-Tank Bioreactor. Biotechnol.
et al. Combinatorial Discovery of Polymers Resistant to Lett. 2013, 35, 1233–1245. doi:10.1007/s10529-013-1211-9.
Bacterial Attachment. Nat. Biotechnol. 2012, 30, 868–875. doi: [156] Hewitt, C. J.; Lee, K.; Nienow, A. W.; Thomas, R. J.; Smith,
10.1038/nbt.2316. M.; Thomas, C. R. Expansion of Human Mesenchymal Stem
[146] Flaim, C. J.; Chien, S.; Bhatia, S. N. An Extracellular Matrix Cells on Microcarriers. Biotechnol. Lett. 2011, 33, 2325–2335.
Microarray for Probing Cellular Differentiation. Nat. Meth. doi:10.1007/s10529-011-0695-4.
2005, 2, 119–125. doi:10.1038/nmeth736. [157] Das, A.; Barker, D. A.; Wang, T.; Lau, C. M.; Lin, Y.;
[147] Flaim, C. J.; Teng, D.; Chien, S.; Bhatia, S. N. Combinatorial Botchwey, E. A. Delivery of Bioactive Lipids from Composite
Signaling Microenvironments for Studying Stem Cell Fate. Microgel-Microsphere Injectable Scaffolds Enhances Stem Cell
Stem Cells Dev. 2008, 17, 29–39. doi:10.1089/scd.2007.0085. Recruitment and Skeletal Repair. PLoS ONE 2014, 9, e101276.
[148] Klim, J. R.; Li, L.; Wrighton, P. J.; Piekarczyk, M. S.; Kiessling, doi:10.1371/journal.pone.0101276.
L. L. A Defined Glycosaminoglycan-Binding Substratum for [158] Das, A.; Tanner, S.; Barker, D. A.; Green, D.; Botchwey, E. A.
Human Pluripotent Stem Cells. Nat. Methods 2010, 7, Delivery of S1p Receptor-Targeted Drugs via Biodegradable
989–994. doi:10.1038/nmeth.1532. Polymer Scaffolds Enhances Bone Regeneration in a Critical
[149] Brafman, D. A.; Chien, S.; Willert, K. Arrayed Cellular Size Cranial Defect. J. Biomed. Mater. Res. A 2014, 102,
Microenvironments for Identifying Culture and Differentiation 1210–1218. doi:10.1002/jbm.a.34779.

You might also like