ChapterPDF Available

Collagen-Based Hydrogels and Their Applications for Tissue Engineering and Regenerative Medicine

Authors:

Figures

Content may be subject to copyright.
Collagen-Based Hydrogels and Their
Applications for Tissue Engineering
and Regenerative Medicine
Sorina Dinescu, Madalina Albu Kaya, Leona Chitoiu, Simona Ignat,
Durmus Alpaslan Kaya, and Marieta Costache
Contents
1 Introduction ................................................................................... 2
2 Collagen-Based Hydrogels: Overview .. . . . . . .. . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . .. . . . . . . 3
2.1 Methods of Obtaining Collagen Hydrogels ............................................ 4
2.2 Properties of Collagen Hydrogels ...................................................... 6
2.3 Collagen-Natural Polymer for Tissue Engineering .................................... 7
3 Applications of Collagen-Based Substituents in Tissue Engineering and Regenerative
Medicine ....................................... ............................................... 8
3.1 Bone and Cartilage Repair .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . 8
3.2 Muscle Tissue Repair ................................................................... 11
3.3 Nerve Tissue Regeneration . . ........................................................... 12
3.4 Vascular Grafts .. . . . . . . . ................................................................ 13
3.5 Corneal Reconstruction ................................................................. 13
3.6 Other Biomedical Applications .. . ..................................................... 14
4 Original Collagen Hydrogels Designed and Tested for Adipose and Cartilage Tissue
Engineering ...................................... ............................................. 15
4.1 Collagen-Sericin Hydrogels .. . . . . .. . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . 15
4.2 Collagen-Polysaccharides Hydrogels .. ................................................ 16
5 Conclusions ................................................................................... 17
6 Future Scope .................................................................................. 17
References .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . 18
S. Dinescu · L. Chitoiu · S. Ignat · M. Costache (*)
Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
e-mail: marietacostache@gmail.com
M. Albu Kaya
Collagen Department, INCDTP Leather and Footwear Research Institute, Bucharest, Romania
D. A. Kaya
Department of Medicinal and Aromatic Plants, Mustafa-Kemal University, Hatay, Turkey
#Springer International Publishing AG, part of Springer Nature 2018
Md. I. H. Mondal (ed.), Cellulose-Based Superabsorbent Hydrogels,
Polymers and Polymeric Composites: A Reference Series,
https://doi.org/10.1007/978-3-319-76573-0_54-1
1
Abstract
A promising solution for soft tissue regeneration is tissue engineering, a multi-
disciplinary eld of research which involves the use of biomaterials, growth
factors, and stem cells in order to repair, replace, or regenerate tissues and organs
damaged by injury or disease. The success of tissue engineering (TE) depends on
the composition and microstructure of the used scaffolds. Ideally, scaffolds have to
be similar to natural tissues. Collagen is the major component of the extracellular
matrix of most soft tissues. The interactions between collagen and cells are vital
in the wound healing process and in adult tissue remodeling, collagen being able
to support differentiation and maintenance of cellular phenotype. As a natural
molecule, collagen possesses the major advantage of being biodegradable, bio-
compatible, easily available, and highly versatile and presents low antigenicity.
This chapter aims to present an overview on the structure, properties, and bio-
medical applications of collagen hydrogels. Moreover, it introduces the reader
to the latest research in the eld of tissue engineering related to collagen. It also
displays the results we obtained as a joint bioengineering group on collagen
hydrogels designed for soft (ATE) or cartilage tissue engineering (CTE) applica-
tions: type I collagen hydrogels improved with either silk sericin (CollSS) or
with pro-chondrogenic factors hyaluronic acid and chondroitin sulfate
(CollSSHACS). Results indicated in both cases the positive inuence of sericin
on the interaction between cells and the surface of the hydrogels. In the absence
of HA and CS, specic chondrogenic inducers, CollSS hydrogel is adapted for
soft tissue reconstruction, whether the addition of HA and CS transforms
CollSSHACS into a suitable hydrogel formula for semihard tissue repair via
modern strategies in tissue engineering and regenerative medicine.
Keywords
Collagen hydrogels · Biomaterial · Regenerative medicine · Tissue engineering ·
Sericin · Hyaluronic acid · Chondroitin sulfate
1 Introduction
Hydrogels are three-dimensional (3D) polymer networks that are able to maintain
a large amount of water and biological uids, whose physical and chemical cross-
links between polymer chains keep their structural integrity [1,2]. The polymer
chains can be natural, synthetic, or hybrid [3]. The natural polymers like collagen,
chitosan, elastin, alginates, hyaluronic acid, pullulan, and brin have been processed
from living organisms. Having a similar structure with the extracellular matrix,
natural polymers possess good biocompatibility properties with low cytotoxicity.
However, because of processing/extraction, they are not preserving mechanical
properties [4]. The synthetic polymers generally used in hydrogels preparation are
poly(ethylene glycol) (PEG), poly(acrylamide) (PAM), poly(vinyl alcohol) (PVA),
polyethylene oxide (PEO), poly(acrylic acid) (PAA), and poly(propylene fumarate-
co-ethylene glycol) P(PF-co-EG) [5,6], which possess controllable chemical and
2 S. Dinescu et al.
mechanical features but less biocompatibility properties. In the last 20 years, hybrid
hydrogels consisting in both natural and synthetic polymers have been developed in
order to provide a better quality of life, mechanical resistance, and water absorption
capacity [3].
The applications of hydrogels are multiple and in different research areas such
as pharmaceuticals, biomedical implants, drug delivery, tissue engineering, and
regenerative medicine [1]. The hydrogels were found to be ideal for clinical appli-
cations, in healing of wounds and burns and even necrotic tissue due to their ability
to transfer vapor and oxygen resulting from their high-water content applied to the
wound site [5].
Due to tissue-mimicking characteristics, the most recent research recommended
the use of hydrogels as scaffolds to provide a biomimetic 3D microenvironment for
the growth of cells [4].
Regenerative medicine is an emerging multidisciplinary eld of research which
involves the use of biomaterials, growth factors, and stem cells in order to regener-
ate, repair, or replace tissues and organs damaged by injury or disease. Currently,
tissue engineering applications are focused toward the use of implantable biohybrids
consisting of scaffolds combined with stem or precursor cells and appropriate
inducers, as a regeneration strategy.
The objective of this chapter is to describe the structure, properties, and
main applications of collagen hydrogels for practical applications in Biology and
Medicine and to present our main ndings in this eld of research.
2 Collagen-Based Hydrogels: Overview
Collagen is the main natural protein of most soft, lax, semirigid, and rigid connective
tissues (skin, bones, tendon, basal membranes, etc.), providing mainly structural
integrity for tissues [7]. Collagen is found in proportion of 80% at skin level,
reported to all dermal dry substances [8,9].
Collagen is a polymer which is characterized by high hydrophilicity, variable
ionic character, and diverse functionality and can be involved in a large number of
interaction systems with other micro- and macromolecular components [10,11].
Collagen is an amphoteric macromolecule with rigid triple helix conformation,
containing both polar groups and hydrophobic portions derived from hydrophobic
amino acids. Consequently, at pH values outside the isoelectric range, between the
molecules, brils, and chain fragments of the non-denatured type I brillar collagen,
all types of intramolecular electrostatic, dipole-dipole, hydrogen and hydrophobic
bonds can be established in the aqueous medium. This may be the reason why
aqueous systems of this type of collagen become gels at low concentrations com-
pared to denatured collagen (gelatin or collagen hydrolysates), which have the
molecules in the form of statistical coils.
The most used collagen extracts which are basic resources for obtaining medical
biomaterials are type I collagen gels and solutions. A gel is dened as a system with
intermediate properties between a uid and a solid. Thus, a gel can be a less viscous
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 3
uid or a very viscous solid. At isoelectric pH, the bers and brils are separated
from the collagen gel, making it dispersible. Collagen gels and solutions are poly-
dispersed colloidal systems, despite tropocollagen solutions which are mono-
dispersed, containing only molecules with triple helix conformation.
According to our experience, type I brillar collagen extracted from the calf
hide by acidic and basic treatments at temperatures between 18 C and 25 C, with
an average molecular weight of 3.13.3.10
5
g/mol, consists of native collagen
molecules, brils, and fragments of single chains.
The rheological study of non-denatured type I brillar collagen gels at pHs outside
the isoelectric range, acid (2.54), neutral (7.0), and alkaline (8.0), showed that they
have pseudoplastic behavior, more obvious at higher concentrations. Viscosity at zero
shear rate increases sharply with collagen concentration. The gels are easier to destroy
by the shear forces at lower concentrations but restore their structure if they are left at
rest long enough. This may suggest that intermolecular hydrogen bonds are carried
out, as with micromolecular amides, via water molecules.
The main forces that produce the coalescence of the type I brillar collagen gel
are electrostatic forces, the others contributing in a small extent to its consistency.
The collagen brils linked by hydrogen bonding and hydrophobic and electrostatic
interactions are not stable, being dissociated by variations in temperature, ionic
strength, pH, or collagenase [12]. In order to improve mechanical strength, thermal
stability, and biodegradation rate, covalent bonds are suggested for collagen gels.
Besides the gels, hydrogels are three-dimensionally hydrophilic polymeric net-
works obtained by cross-linking of gels. Cross-linking ensures the insolubility of
hydrogels in water because of the ionic interaction and hydrogen bonding, providing
mechanical strength as well as physical integrity to the polymeric hydrogels [1].
2.1 Methods of Obtaining Collagen Hydrogels
Collagen hydrogels are biomaterials obtained by cross-linking of corresponding
gels. Mechanical and biological properties are controllable and usually superior
compared to the gels from which they were obtained.
Natural in vivo cross-linking provides mechanical strength and proteolysis resis-
tance to collagen [13]. Used in vivo, collagen biomaterials are subject to enzymatic
degradation, which is why they are stabilized by in vitro cross-links, especially with
aldehydes [1416]. Cross-linking methods lead to creation of additional chemical
bond between collagen molecules and/or brils, increasing mechanical and chemical
strength and, consequently, reducing biodegradability.
In vitro collagen cross-linking methods can be classied in two categories:
physical and chemical. The main disadvantage of chemical cross-linking is the
potentially toxic effect of residual cross-linking agent or of complexes formed in
the process of in vivo degradation. In order to eliminate this disadvantage, physical
treatment is applied, incl uding heat-drying and exposure to UV and γradiation. Both
dehydrothermal and UV exposure at 254 nm wave length increase the collagen
contraction temperature and also the enzymatic degradation strength. However,
these physical treatments cause partial denaturation in collagen [13].
4 S. Dinescu et al.
Chemical cross-linking consists of collagen reactions with aldehydes,
diisocyanates, acyl azides, polyepoxides, and polyphenolic compounds, which
lead to the formation of ionic or covalent bonds between molecules or brils.
The most used cross-linking agents are aldehydes, especially formic and glutaric.
From all cross-linking agents, glutaraldehyde (GA) is the most used due to its high
efciency in stabilization of biomaterials made out of collagen. GA cross-linking
involves reactions of the free ε-amine groups of lysine or hydroxylysine from the
polypeptidic chains with the GA aldehyde groups [17]. Lysine and hydroxylysine
represent 3% from all collagen amino acids and, because of this, approximately 60%
of those amino acids residues react with GA under the conditions used to achieve
a certain degree of xation [18].
Although other cross-linking agents are preferred to reduce cytotoxicity, they
cannot equal GA in terms of stability [11]. The success of thousands bioprosthesis
implanted in last years has shown that GA cross-linking is clinically accepted,
despite reported cytotoxicity [16,19,20]. Biocompatibility of collagenic materials
cross-linked with GA can be increased by lowering its concentration or by combin-
ing it with others physical-chemical treatments [21]. As an alternative to aldehyde
treatment, resistant collagenic materials can be obtained using hexamethylene
diisocyanates (HDC) as cross-linking agents. HDC is solubilized in water with
surfactants or dissolved in 2-propanol. It forms bonds with two chemical ε-amine
groups passing through urea-like linkages, mainly used to obtain cross-linked
gelatin-based plasma substitutes.
Many studies have focused on the use of epoxide compounds (diglycidyl ethylene
glycol, polyglycidyl glycol, methylglycidyl ether) [22]. Epoxide compounds react
easily with amine groups of lysine, but they also pose the problem of
cytotoxicity [23].
Carbodiimide cross-linking, especially with 1-ethyl-3-(3-dimethylaminopropyl)
carbodiimide (EDC), offers the advantage of forming amide linkages between the
carboxyl and amine groups of the collagen molecules without becoming part of the
effective linkage. Thus, dysfunctional cross-linking agents are avoided [13].
Collagen gel and collagen gel cross-linked with glutaraldehyde and with
carbodiimide obtained in our laboratory are presented in Fig. 1.
Intermediary forming of acyl azide is an alternative method of those with
carbodiimide, which has as result amidic bonds introduction [24]. Carboxyl groups
are transformed into hydrazide. Then, the reaction with sodium nitrite takes place to
form the acyl azide. Otherwise, the diphenylphosphoryl azide modication can be
made. Acyl azide treatment increases collagen-glycosaminoglycan matrix resistance
for up to 3 months and inhibits calcication in vivo, while collagen treated with
glutaraldehyde is completely calcied after 15 days due to calcium xation at free
carboxyl groups [25].
Polyphenols have been used as vegetal tannins for tanning animal skins for a very
long time [26]. These are good cross-linking agents due to multiple interactions with
proteins, thus increasing the stability of collagen from hydrogels or spongious
matrices.
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 5
Genipin is a natural cross-linker which demonstrated lower cytotoxicity and
better biocompatibility compared with chemically synthetized cross-linking
agent [27].
Collagen properties can be controlled by using different type or amount of cross-
linkers named before and by varying cross-linking conditions. Cross-linking gener-
ally creates a covalently bonded gel that does not deform as a uid during injection,
but it rather fractures and does not recover, leaving a mass of particles.
2.2 Properties of Collagen Hydrogels
Hydrogels are three-dimensional networks made of hydrophilic polymers obtained
from gels by covalent bonds cross-linking or by ionic forces, which in aqueous
medium swell to an equilibrium value.
Cross-linking degree is one of the most important factors which affects hydrogel
swelling. It is dened as a ratio between the number of moles of cross-linker and the
number of moles of units that are repeated from the polymer molecule. The higher
the degree of cross-linking, the more dense structure and the less intense swelling
due to reduction in polymeric chain mobility. The degree of swelling is also affected
by the chemical structure of the polymer: hydrogels containing large amount of
hydrophilic groups are swelling more intense than those containing hydrophobic
groups.
Hydrogel swelling may also be affected by the swelling environment temperature
or ionic strength and pH, as well as by other factors [28].
The kinetics of hydrogel swelling can be controlled by diffusion when applying
Ficks law or by relaxation when the law is no longer respected. If the diffusion of
water in the hydrogel occurs faster than the relaxation of the polymeric chain, the
swelling kinetics is controlled by diffusion.
The mechanical properties of hydrogels are very important for pharmaceutical
applications and can be achieved by modifying their cross-linking degree. Increasing
cross-linking leads to more consistent but more fragile gels. There is an optimal
degree of cross-linking for which the hydrogel is elastic and resistant.
Fig. 1 Collagen gels: (a) un-cross-linked and cross-linked with (b) glutaraldehyde and
(c) EDC/NHS
6 S. Dinescu et al.
Collagen, alone or in combination with other molecules from the extracellular
matrix, plays an important role in the physiology and behavior of cells from
connective tissue. Keratinocytes and broblasts are important cells involved in
healing skin lesions. Therefore, the use of collagen hydrogels as a substrate for the
cell culture improves adherence, migration, growth, and cell differentiation under
normal and pathological conditions [4].
The interaction between collagen and blood platelets plays an important role in
the mechanism of hemostasis induction. Platelets adhere on the collagen surface;
their adhesion and aggregation activate clotting and initiate hemostasis. The reaction
depends on the cross-linking degree of collagen from hydrogels and on the positive
loads localized on the base groups from the side chain. If free collagen carboxylic
groups are blocked by chemical processes, more than 98% of hemostatic activity
disappears. Hemostasis is dependent also on the conformational structure of colla-
gen. Thus, denatured collagen (gelatin) does not activate platelet aggregation,
whereas non-denatured collagen in forms of brils can activate platelets, being
efcient in hemostasis induction.
Generally, the interactions between collagen and cells are important phenomena in
wound healing process and in adult tissue remodeling, collagen inducing differenti-
ation and maintaining cell phenotype [29,30]. Another important biological property
of collagen is biocompatibility, due to its low toxicity and poor immunogenicity.
Also, collagen is poorly antigenic. Low antigenicity can be due to the presence
of aromatic amino acids, especially tyrosine. In collagen, there is a low quantity of
aromatic amino acids, about three residues of tyrosine on a chain. To reduce or
eliminate collagen antigenicity, the N-terminal regions of the polypeptide chains
are removed during the collagen extraction process.
2.3 Collagen-Natural Polymer for Tissue Engineering
The most recent studies showed a continuing concern about obtaining biomaterials
which mimic extracellular matrix for further use in regenerative medicine. The
collagen, the main component of skin extracellular matrix, can be used as tissue
engineering scaffold in combination with other polymers or cross-linked [27].
Collagen-elastin cross-linked squaric acid created a 3D transparent hydrogel
highly resistant to enzymatic degradation to be used in medicine and tissue engi-
neering [31]. Other hydrogels based on collagen, hyaluronic acid (HA), and sericin
in different ratios showed that HA played the role of cross-linking agent which were
proved to be nontoxic and very consistent against enzymatic treatment [32]. Collagen
and chitosan in the presence of α,β-glycerophosphate formed hydrogels which
proved their ability to be biocompatible substrates with L929 cells, being promising
scaffolds for tissue engineering [33]. Drug delivery systems and hybrid hydrogels
based on collagen such as collagen-hydroxyapatite-alendronate hydrogel for
MC3T3-E1 osteoblastic cells [33], collagen-polyvinylpyrrolidone superabsorbent
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 7
hydrogels cross-linked by γ-irradiation [34], chitosan-collagen coated with
poly(L-lactic acid) by electrospinning [35] for tendon regeneration, and collagen-
poly(N-isopropyl acrylamide) with montmorillonite nanoparticles incorporated
hydrogel [36] were also developed.
Our experience in natural collagen hydrogels allowed the development of func-
tional hydrogel scaffolds biocompatible with different types of cells and designed
for different tissue engineering applications. These hydrogels are further presented
in Sect. 4.
3 Applications of Collagen-Based Substituents in Tissue
Engineering and Regenerative Medicine
Collagen is the key component of the extracellular matrix in most tissues and is
involved in the maintenance of tissue three-dimensional architecture, as well as in
the development of new tissues and organs. Consequently, one of the targets in tissue
engineering is to develop substitute biomaterials that can closely resemble the
extracellular matrix and, thus, to be integrated both structurally and functionally in
the newly formed tissue. Due to the high prevalence of collagen in all tissues and
organs, the engineered collagen-based substituents have wide applications, as further
described.
3.1 Bone and Cartilage Repair
Collagen is the main structural protein in the extracellular matrix of hard and
semihard tissues [37]. Consequently, orientation toward collagen-based biomaterials
for cartilage and bone tissue engineering came as a natural choice; however, because
collagen alone is not a suitable substituent for bone tissue engineering (BTE)
applications in terms of mechanical properties, the usage of a new combination of
materials in the pursuit of a more biomimetic scaffold was reported [38]. The team
created a nanosized hydroxyapatite (nHA) and collagen-based hydrogel as a support
for the proliferation and differentiation of human mesenchymal stem cells (hMSCs)
isolated from either adipose tissue (AT-MSCs) or bone marrow (BM-MSCs).
Although their hypothesis stated that osteogenic differentiation should be dependent
on nHA concentration, the results of the study showed that AT-MSCs display a
higher osteogenic potential compared to BM-MSCs, irrespective of nHA amount in
the constructs.
Similarly, in 2016, Gurumurthy et al. [39] reported the attempt to nd a more
suitable scaffold for guided bone regeneration. Since collagen alone is characterized
by mechanical weaknesses, composites with elastin-like polypeptide (ELP) were
analyzed as a support for human adipose-derived stem cells (hASCs) in osteogenic
differentiation processes. After 3 weeks of evolution, the ELP-collagen constructs
proved to be superior to pure collagen scaffolds in terms of tensile strength, elastic
modulus, osteogenic activity, and mineralization.
8 S. Dinescu et al.
A new technique in BTE [40] offers a deeper perspective in long-term repair
of bone defects. Using an iterative layering freeze-drying method of assembling
gradual amounts of collagen (Col) and hyaluronic acid (HA), the group has created
a biomimetic bone substitute which presents a top-bottom gradient of both
interconnected porosity and HA content. The similarities with the physiological
structure of the bone were easily observed, and the chemical composition resembled
the natural model, with 1
=
3organic and 2
=
3inorganic elements. The biocompatibility of
this Col/HA scaffold was assessed via the evolution of a seeded BM-MSCs culture.
Compared to a collagen scaffold, the Col/HA composite indicated better rates of
success in bone tissue regeneration, and thus, it could be considered an advanced
strategy in tissue engineering.
One of the main implementation problems in BTE is to ensure the blood supply to
the construct. According to a recent study [41], this issue may have found its solution
in cocultivation of cells. As well as bone grafting, bone tissue engineering is a feasible
approach toward minimizing extended bone defects in the elds of orthopedics and
oncology. In order to regenerate bone fragments, the team studied the advantages of
cocultivating endothelial cells together with an osteogenic cell lineage in the context
of a mouse calvarial defect model. Using a negative control represented by a pure
collagen scaffold, a positive control of a collagen scaffold enhanced with bone
morphogenetic protein 7 (BMP-7), and three collagen scaffolds seeded with human
osteoblasts (hOBs) only, CD34+ cells only, and a coculture of hOBs and CD34+ cells,
respectively, Hertweck et al. concluded through a series of radiographic, histological,
immunohistological, and statistical methods that the cocultivation of hOBs and CD34
+ cells or even the monoculture of CD34+ cells offers a better perspective in bone
regeneration than monocultures of osteoblasts.
More recently, Nguyen et al. [42] reported the analysis of a suitable biomaterial
for prevascularized bone fabrication prior to implantation in a patient. The group
determined the importance of specic environmental parameters in human umbilical
vein endothelial cells (HUVECs) and hMSCs coculture. Different scaffold types,
from alginate hydrogels for hMSCs differentiation into osteoblasts to collagen type I
hydrogels for HUVECs angiogenesis, were assessed. In addition, shear stress was
applied to this coculture system in a tubular perfusion system bioreactor in order to
advance the progression toward osteogenesis and prevascularization. Moreover, it
was observed that cells attached, proliferated, and differentiated with better rates due
to specic peptide sequences as native content of cell-binding sites in collagen
brils, leading to the conclusion that collagen would be better for future bone tissue
engineering applications.
In 2015, Lee et al. [43] studied the fabrication process of biomimetic three-
dimensional structures with preosteoblasts and hASCs. Their team engineered a
bioink that apart from being nontoxic, biocompatible, and printable, also has cell-
activating properties. By pre-cultivating cells in collagen, they enriched the tradi-
tional collagen/alginate composite with extracellular matrix. After being printed, the
resulted cell-embedded bioink undergone a cell viability and differentiation study:
preosteoblasts successfully started displaying osteogenic activity; whereas hASCs
followed a hepatogenic differentiation pathway, expressing liver-specic genes.
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 9
Cartilage is an avascular, aneural, and alymphatic tissue, with a very low number
of progenitor cells and a very slow turnover of the extracellular matrix. Therefore,
this tissue has a very limited auto-regeneration potential and represents one of the
most addressed tissues by tissue engineering and reconstruction procedures. In this
context, collagen-based biomaterials have been widely used for applications, such as
repair of articular cartilage lesions, replacement of intervertebral discs, knee menisci,
anatomical disorders of the temporomandibular joint, deformities of the midface and
ears, etc.
To address one of the challenges of cartilage engineering, several collagen-
mimetic hydrogels with chondrogenesis-inducing properties have been proposed.
For example, Parmar et al. suggested a novel biodegradable collagen hydrogel
with incorporated heparin-binding, integrin-binding, and hyaluronic acid-binding
sequences at the backbone of a streptococcal collagen-like 2 protein. Meta-
lloproteinase 7 (MMP7) and aggrecanase (ADAMTS4)- cleavable peptides were
later used in the process as cross-linkers. Their conclusion was that this new
collagen-based hydrogel can be used for cartilage regeneration therapies, since it
promoted cell viability, cell adhesion, and chondrogenesis [44].
One of the treatment courses adopted for focal cartilage lesions involves matrix-
induced chondrocytes implantation (MACI). As this method requires autologous
chondrocytes prone to dedifferentiation during in vitro expansion, Fensky et al.
proposed the implantation of predifferentiated MSCs seeded in a type I collagen
hydrogel. The MSCs were cultured in collagen hydrogels, and predifferentiation was
initiated by adding TGF-β1 in the culture media. After 10 days, the hydrogels were
switched to a TGF-β1-free media for another 11 days. In both histochemical assay and
chondrogenic markers, gene expression conrmed the evolution of chondrogenesis.
Therefore, the collagen hydrogel could represent a reliable method in delivering
predifferentiated cells to the needed implantation site [45]. Additionally, Chen
et al. [46] concluded that a type I collagen hydrogel seeded with mesenchymal
stem cells isolated from the Whartons jelly of human umbilical cord (UC-MSC)
could be a suitable 3D model for cartilage healing.
The suitability of collagen hydrogels in CTE was also conrmed in a study
focused on materials immunomodulatory properties [47]. The group investigated
the immunogenicity of neonatal rabbit chondrocytes seeded in a collagen type I
hydrogel. Even though the immunogenicity markers showed a tendency to increase
over the rst 14 days of in vitro culture, after 28 days, the immunogenic effects were
lower. The collagen hydrogel promoted the synthesis of extracellular matrix over the
28 days in culture. This led to the accumulation of extracellular matrix, and it was
suggested that it isolated the seeded cells from the host immune cells and from the
hydrogel itself, providing some immunogenicity-reducing effects. In addition,
in 2016 Yang et al. [48] published a study that compared the evolution of
chondrogenesis and the secretion of immunoregulatory factors of MSCs in 2D and
3D culture. The cells expanded in the collagen hydrogels (3D) showed higher
chondrogenic markers at gene and protein levels of expression than the ones
on plastic (2D), simultaneously with an enhanced secretion of immunoregulatory
factors. This could suggest that the collagen hydrogel promoted the chondrogenic
differentiation of MSCs.
10 S. Dinescu et al.
3.2 Muscle Tissue Repair
Collagen constitutes 12% of the muscular tissue, where it serves as a major
component of the endomysium. Several studies have been performed in order to
reconstruct muscle tissue assisted by collagen substituents, as further described.
Recently a study was conducted regarding the usage of vascularized tissue scaffolds
in large volumetric muscle defects [49]. Working on a rat biceps femoral model, the
team developed a series of vascularized collagen hydrogels with adipose-derived
microvessels. After seeding these scaffolds with myoblasts and implanting them in
the existent limb injury, Li et al. observed the revascularization and muscle regen-
eration processes, as well as the differentiation of myoblasts in myotubes. However,
as compared to a vascularized scaffold only, which showed a similar regeneration
rate after a 2-week evolution, the team concluded that while the vascular network of
the construct may support the regeneration of volumetric muscle defects, other
factors have an important role as well.
With respect to the cardiac muscle, studies have been developed to test
the efciency of hybrid hydrogels based on collagen in myocardial regeneration.
Injectable bioactive hydrogels were developed from thiolated collagen (Col-SH) by
adding multiple acrylate containing oligo(acryloyl carbonate)-b-poly(ethylene gly-
col)-b-oligo(acryloyl carbonate) (OAC-PEG-OAC) copolymers for improving the
materials mechanical properties [50]. The cellular lineage selected for this study
consisted of BM-MSCs, due to their rapid spreading rate and extensive network-
forming capacity. After the implantation of BM-MSC-encapsulating hybrid hydro-
gel in a rat infarction model, Xu et al. observed a signicant improvement in cardiac
function in the case of the hydrogels as compared to the PBS control. Moreover, the
echocardiography analysis showed an increase in the ejection fraction at 28 days,
while the histological assessments demonstrated a decrease in the infarct size and an
increase in ventricular wall thickness.
More recently, von Marion et al. [51] published a study regarding a different
method of addressing cardiac stem cell therapy by culturing cells directly in the
biomaterial for better survival, proliferation, and differentiation rates. Cardiomyocyte
progenitor cells (CMPCs) were seeded in a unidirectional constrained and a stress-
free collagen hydrogel. The three-dimensional environment highly supported cellular
growth and maturation, and an enhancement was observed in specic markers
(Nkx2.5, myocardin). The proof that the CMPCs were actively involved in the matrix
secretion and remodeling processes was given by levels of collagen type I, collagen
type III, elastin, bronectin, and matrix remodeling enzymes. Furthermore, when
constrained, CMPCs became mechanosensitive, adopted a rod-shaped morphology
and responded to mechanical stimuli in addition to developing sarcomeres and
contractility.
In 2017, Ketabat et al. [52] identied collagen and alginate injectable conductive
hydrogel systems as promising candidates for cardiac muscle regeneration in terms
of cardiomyocyte viability and syringeability. Other tissue engineering applications
of such hydrogels rely on their specic property that allow delivery to the requested
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 11
site through a catheter. Collagen is therefore used for its self-assembly characteristic,
resulting brous forms upon warming up to 37 C, while alginate is recommended
for its non-thrombogenic nature.
3.3 Nerve Tissue Regeneration
Currently, nerve injuries or diseases that affect the nervous system are found more and
more often and seriously affect patientslives. These nerve injuries could be ef-
ciently addressed by modern strategies in tissue engineering, namely, by designed
scaffolds that would represent the necessary support for nerves to regenerate. A few
of these strategies are further discussed.
In 2015, a study regarding a novel combination of PuraMatrix 3D nanobrous
hydrogel and a honeycomb collagen sponge in creating a scaffold suitable for
neuronal regeneration after complete transection of the spinal cord in adult rats
was published. After the implantation of the construct in the 5 mm spinal gap, its
evolution was recorded during 24 days to 4 months, as compared to the PBS-injected
control group. The histological and western blot analyses have indicated the migra-
tion of both neurons and astrocytes toward the implant site, as well as their
differentiation and maturation stages, therefore suggesting functional recovery,
spinal repair, and neuronal regeneration [53].
Using carbon nanotubes and collagen in generating three-dimensional micro-
environmental conditions for MSCs in neural tissue regeneration, Lee et al. [54]
highlighted once again the impact of collagen scaffolds on bioengineering. Studying
the differentiation process of MSCs in neural lineages, the team has selected collagen
as the ideal support for MSCs growth and expression of neural phenotypes, while the
addition of small amounts of carbon nanotubes (0.11 wt%) has improved cellular
proliferation rate. Moreover, CNTs were associated with improvements in both
expression of neuronal markers and secretion of neurotrophic factors, especially
nerve growth factor and brain-derived neurotrophic factor.
Similarly, Park et al. [55] introduced the concept of 3D collagen scaffolds
for human umbilical cord blood (UCB) cells in order to promote the secretion
of therapeutic factors, such as neurotrophic factors. Collagen proved to be an
advantageous biomaterial, not only being a suitable 3D microenvironment but also
stimulating the secretion of neurotrophins, nerve growth factor, brain-derived
neurotrophic factor, and ciliary neurotrophic factor. Moreover, the 3D scaffold has
also been described as an efcient reservoir of neurotrophic factors by storing and
slowly delivering them toward the target cells. To conrm their hypothesis, Park
et al. have facilitated indirect interaction of UCB environment with human neural
precursor cells, observing afterward neurite outgrowth. Future implications of such
scaffolds include therefore neural repair and regeneration processes.
12 S. Dinescu et al.
3.4 Vascular Grafts
Aiming to study vascular biology in health and disease, Roberts et al. published
in 2016 a study concerning the process of generating microvessels by seeding
endothelial cells in a microuidic channel embedded with a native type I collagen
hydrogel. Their three-dimensional model serves as a solution for the inadequate
planar cell culture and uses injection molding methods to generate engineered blood
vessels. The study of seeded HUVECs has already offered an insight in pericyte
migration, platelets adhesion to activated endothelium, uid shear stress modulation
of endothelial activity, and von Willebrand factor (VWF) secretion and assembly.
Moreover, by adding parenchymal cells in the collagen matrix, the researchers have
created a tissue microenvironment which allows them to further study processes
related to the intestinal lining, lung alveoli, kidney tubule, and many others [56].
The developing process of an injectable allogenic collagen-phenolic hydroxyl
(collagen-Ph) hydrogels in mice that supports de novo generation of a three-
dimensional vascular network was recently documented [57]. Specically, they
injected in subcutaneous murine models a prepolymer solution of collagen-Ph,
human blood-derived endothelial colony-forming cells (ECFCs), bone marrow-
derived mesenchymal stem cells (MSCs), horseradish peroxidase, and hydrogen
peroxide and analyzed the course of tissue forming. After 7 days, the ECFCs have
generated extensive vascular networks and functional anastomoses with the existing
vasculature of the host tissue; the collagen has improved MSCs differentiation, while
phenolic hydroxyl has favored an increase in the number of adipocytes present in the
targeted tissue [57].
3.5 Corneal Reconstruction
The cornea may suffer from diseases, injury, infections, or inammation, and these
can lead to the need of a corneal replacement. To avoid total replacement by surgery,
modern approaches in tissue engineering identied the opportunity to use adapted
hydrogels to assist corneal regeneration. Latest research in this eld is further
presented.
In 2016, Rafat et al. [58] assessed the suitability of collagen-based hydrogels
as biomaterials in corneal regeneration. Their team engineered a construct with
a transparent core and an adjustable periphery characterized by faster degradability
rates. The material was used in in vitro cultivation of human epithelial cell popul-
ations and transplanted in an in vivo mice model. After a 3 monthstime evolution,
the collagen construct was declared suitable for corneal transparency restauration,
maintaining the overall corneal shape and integrity.
Previously, other studies [59] presented an alternative to the cross-linkage
method of generating transparent collagen hydrogels. By substituting the
usage of N-(3-dimethylaminopropil)-N0-ethylcarbodiimide (EDC) with a sterically
bulky carbodiimide, N-cyclohexyl-N0-(2-morpholinethyl)carbodiimide metho-p-
toluensulfonate (CMC), the group obtained corneal replacements with longer, easier
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 13
to control gelation time, as well as superior resistance to collagenase. Liu et al. [60]
started using collagen in developing corneal substitutes since 2008. Their hydrogels
were synthesized as interpenetrating polymeric networks (IPNs) and were constituted
of 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide, N-hydroxysuccinimide cross-
linked porcine atelocollagen, and poly(ethylene glycol) diacrylate cross-linked
2-methacryloyloxyethyl phosphorylcholine. Compared to each individual com-
pound, the hydrogel presented greater mechanical strength, as well as enzymatic
stability and low UV degradation rates. Due to collagen, the corneal substitute
appeared to be cell-friendly, promoting nerve ingrowth and regeneration. Moreover,
its optical properties resemble the human model. When implanted in pigs, the
allografts indicated in vivo compatibility, the experiment being declared a success.
After previously postulating porcine collagen hydrogels as corneal substitutes,
another study [61] assessed the potential use of recombinant human type I and type
III collagen in such applications in order to minimize the possible immune reactions
toward animal origin implants. By cross-linking the collagen brils with 1-ethyl-3-
(3-dimethyl aminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS),
the team created a corneal substitute with light transmission properties comparable to
superior to that of a human model. Not only that these substitutes had adequate
tensile strength and elasticity for surgical procedures, but type III collagen proved
to be superior in terms of mechanical characteristics. During a 12-month post-
implantation assessment in pigs, the hydrogels maintained their optical clarity,
promoted regeneration of corneal cells, nerves, and tear lm and proved to be
eligible for further clinical applications.
McLaughlin et al. [62] addressed the problem of tissue regeneration by extracel-
lular mimics in the context of cornea, a three cellular layered structure in a hydrated
extracellular matrix, delimited by a nonkeratinizing epithelium and an inner endo-
thelium. Proposing a corneal substitute made out of carbodiimide cross-linked
porcine type I collagen, they have demonstrated that simplicity in fabrication
might be the key to future biomaterial applications. During a 12-month integration
and development of the collagen implant in the host tissue, it was observed that the
substitute supported the regeneration of corneal cells, nerves, and the tear lm.
Moreover, it had been described as maintaining its optical clarity.
3.6 Other Biomedical Applications
Collagen scaffolds can also be used as apoptotic sites when conjugated with
cyclopamine, according to a study of Jain et al. published in 2014. The team
analyzed the migration of the cells constituting a glioblastoma, an aggressive brain
tumor which invades adjacent cerebral regions along white matter tracts and blood
vessels. After engineering aligned polycaprolactone-based nanobers as an invasion
site, they mediated the transfer of the tumor toward the extracortical cyclopamine
drug-conjugated, collagen-based hydrogel. Cyclopamine was preferred because it
only affects cells which are dependent on the sonic hedgehog pathway of survival,
i.e., cancer stem cells. Tumor loads were signicantly higher in the case of nanober
14 S. Dinescu et al.
implants compared to smooth lm or empty conduit implants. Moreover, tumor
loads outside of the collagen-based hydrogel were signicantly lower in the case of
nanober implants [63].
In 2013, Rao et al. developed an unprecedented study of a series of three-
dimensional collagen hyaluronan composite hydrogel in order to achieve an
accurate model of glioblastoma tumor cell migration. Their work focused on using
biomaterials to understand tumor cell biology rather than promote tissue engineer-
ing. Collagen scaffolds were selected due to the materials association with cell
migration, collagen types I, III, and IV being found in glial lamina externa and
vascular basement membranes, while collagen types I and III as well as hyaluronan
are the main constituents of the native brain extracellular matrix. The team observed
that glioblastoma migration occurs in inverse function of hyaluronan concentration;
therefore hyaluronan has a delaying effect on cell movement [64].
4 Original Collagen Hydrogels Designed and Tested
for Adipose and Cartilage Tissue Engineering
Based on the positive results reported by other groups and on the versatility
of collagen-based hydrogels for different tissue engineering applications, our
group has studied two different composites designed for cartilage and soft tissue
reconstruction.
4.1 Collagen-Sericin Hydrogels
Hydrogels based on type I brillar collagen (Coll) with different ratio of silk sericin
(SS) presented a superporous and absorbent structure (Fig. 2). The pore size varied
between 2 and 90 μm, the structure being denser with increasing amount of sericin.
The absorbent capacity showed values between 3000% and over 4000% at acidic,
alkaline, and neutral pH and classied hydrogel as being very absorbent. Even the
high amount of sericin showed more compact structures. The mechanical properties
and biodegradation rates demonstrated that the best scaffold was Coll:SS =5:2
ratios [65]. The physicochemical results were conrmed by biological ones which
showed that preadipocytes colonizing the scaffold containing 40% sericin, being a
good candidate for future applications in soft tissue engineering [66].
This validated composition of CollSS hydrogel was developed for adipose tissue
engineering (ATE) and revealed good biocompatibility and supported differentiation
of human adipose-derived stem cells (hASCs) [67]. Silk sericin (SS) is a natural
sticky protein isolated from B. mori silk bers, which proved to be responsible for
proliferation and attachment of several mammalian cell lines [68] as well as for the
activation of collagen production, both in vitro and in vivo [69].
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 15
4.2 Collagen-Polysaccharides Hydrogels
Furthermore, in order to direct this biocompatible composite (CollSS) toward
cartilage tissue engineering (CTE), two pro-chondrogenic molecules, components
of cartilage extracellular matrix hyaluronic acid (HA) and chondroitin sulfate (CS)
were added in the composition, resulting in a pro-chondrogenic collagen hydrogel
(CollSSHACS). Several ratios between HA and CS were tested to identify the
most efcient one for CTE: CollSS-10%HA, CollSS-5%HA, CollSS-10%CS, and
CollSS-5%CS. HA increased the thermal stability of scaffolds; the absorbent prop-
erties showed a higher capacity to uptake water (42004300%) than the control
samples (CollSS) and the pore size between 20 and 150 μm, being larger for CollSS
samples which contain HA and smaller for samples which contain CS [70].
Cytocompatibility studies with hASCs and their differentiation toward the carti-
lage lineage revealed the collagen hydrogel improved with 10% HA and 5% CS
to be the most equilibrated formula to support cell proliferation [70] and hASCs
chondrogenesis during in vitro studies. Briey, 3D cell-scaffold culture was
achieved by seeding hASCs in CollSSHACS and then exposed to chondrogenic
induction cocktail for 28 days. The in vitro differentiation process was monitored at
7, 14, and 28 days post-induction. Scanning electron microscopy (SEM) revealed
that cells distributed in the volume of the hydrogel through the network of pores and
adhered to the CollSSHACS better than to control Coll, probably due to the sticky
properties of sericin. Gene expression studies revealed upregulated chondrogenic
markers such as SRY (sex determining region Y)-box 9 (Sox9) and cartilage
oligomeric matrix protein (COMP). These results were also conrmed at protein
level by confocal microscopy.
Fig. 2 SEM image of
collagen: sericin (5:2)
hydrogel, x 200
16 S. Dinescu et al.
To conclude our experience with collagen hydrogels, we have evaluated
hASCs/CollSS bioconstruct for ATE [67] and hASCs/CollSSHACS for CTE.
Results indicated in both cases the positive inuence of sericin on the interaction
between hASCs and the surface of the hydrogels and on the differentiation pro-
cesses. Evaluation by immunohistochemistry and microscopy showed the formation
of cellular aggregates in CollSSHACS, which is specic for the rst steps of
chondrogenesis. Nevertheless, in the absence of HA and CS specic chondrogenic
inducers, the conditions offered by CollSS hydrogel favor hASCs differentiation
toward the adipogenic lineage, thus conrming CollSS to be appropriate for soft
tissue engineering applications.
5 Conclusions
Collagen-based hydrogels incorporating other natural polymers (derived from extra-
cellular matrix tissue) showed very good biocompatibility with different types of
cells, but request controllable biodegradability and improved mechanical properties
using suitable cross-linker agents (preferably natural). The hydrogels consisting in
collagen and synthetic polymer have controllable physical-chemical properties but
poor biocompatibility and request surface functionalization with natural polymers in
order to improve their biocompatibility. Biopolymers such as hyaluronic acid, chon-
droitin sulfate, and sericin in combination with collagen formed 3D hydrogels with
high biocompatibility and potential to support cells differentiation. Furthermore,
CollSS could prove useful for soft tissue engineering, while CollSSHACS could
be a good candidate for cartilage reconstruction.
6 Future Scope
For future studies, more attention should be focused on smart biomaterials such as
drug delivery systems with antimicrobial properties based on collagen hydrogels.
The drug delivery systems request complex structure so that the drug can be released
from hydrogels in a controlled way to avoid the risk of not being effective from the
pharmaceutical point of view. A high amount of drug could be toxic for cells the
hydrogel will not be biocompatible, while a small amount of drug would not
inuence the biocompatibility nor be effective for the proposed aim.
Other challenge in tissue engineering is the risk of infection at the tissular level.
For this reason, we focused to establish a balance between antimicrobial and
biocompatibility properties, using natural ingredients such as essential oils and
zeolite [71]. Previous results showed that it is possible to open such a barrierbut
extensive research needs to be performed in the future.
Another opportunity to further improve the collagen hydrogel would be to bring
together the benets of collagen with the benets of autologous platelet-rich plasma
(PRP). Rich in growth factors that positively inuence cell growth, proliferation,
adhesion, and other cellular processes, PRP would ensure better biocompatibility,
lower inammatory potential, and better acceptance rate at the implant site of
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 17
collagen. One recent study [72] approached this idea and used it to create a collagen-
based construct dedicated for skin regeneration. Researchers developed a type I
collagen, fractionated platelet-rich plasma (PRP)-supplemented scaffold, able to
recruit dermal stem cells from adjacent tissue, fully disclaiming the need of cell
seeding. As compared to a collagen fetal bovine serum control, the new biomaterial
proved to be better when implanted into rat wound models in terms of epitheliali-
zation and neovascularization. Not only the number of recruited stem cells was
higher when PRP was used, but also the dermis-like tissue formation proceeded at
a faster rate, leading to hair growth and sweat gland production [72].
Acknowledgments The authors would like to acknowledge the funding sources that supported
this work: grant 65PCCDI/2018 (REGMED), Project 3- dedicated to regeneration of soft tissues and
to national project Bridge Grant PNIII-P2-2.1-BG-2016660 0458 (123BG/2016), as well as the
National Executive Agency for Research Funding.
References
1. Varaprasad K, Raghavendra GM, Jayaramudu T, Yallapu MM, Sadiku R (2017) A mini review
on hydrogels classication and recent developments in miscellaneous applications. Mater Sci
Eng C Mater Biol Appl 79:958971
2. Drury JL, Mooney DJ (2003) Hydrogels for tissue engineering: scaffold design variables and
applications. Biomaterials 24:43374351
3. Gyles DA, Castro LD, Carrera Silva JO Jr, Ribeiro-Costa RM (2017) A review of the designs
and prominent biomedical advances of natural and synthetic hydrogel formulations. Eur Polym
J 88:373392
4. Naahidi S, Jafari M, Logan M, Wang Y, Yuan Y, Bae H, Dixon B, Chen P (2017)
Biocompatibility of hydrogel-based scaffolds for tissue engineering applications. Biotechnol
Adv 35(5):530544
5. Vedadghavami A, Minooei F, Mohammadi MH, Khetani S, Rezaei A, Mashayekhan S,
Sanati-Nezhad A (2017) Manufacturing of hydrogel biomaterials with controlled mechanical
properties for tissue engineering applications. Acta Biomater 62:42
6. Saldin LT, Cramer MC, Velankar S, White LJ, Badylak SF (2017) Extracellular matrix
hydrogels from decellularized tissues: structure and function. Acta Biomater 49:115
7. Ramachandran GN (1967) Structure of collagen at the molecular level. In: Ramachandran GN
(ed) Treatise on Collagen. Academic, London, pp 747748
8. Prockop DJ, Kivirikko KI (1995) Collagens: molecular biology, diseases, and potential for
therapy. Annu Rev Biochem 64:403434
9. Uitto J, Pulkkinen L, Chu ML (1999) Collagen. In: Freedberg IM (ed) Dermatology in general
medicine. McGraw-Hill, New York, pp 303314
10. Mecham R (2012) Overview of extracellular matrix. Curr Protoc Cell Biol 57:10.1.110.1.16
11. Eyre DR (1980) Collagen: molecular diversity in the bodys protein scaffold. Science
207(4437):13151322
12. Tian Z, Liu W, Li G (2016) The microstructure and stability of collagen hydrogel cross-linked
by glutaraldehyde. Polym Degrad Stab 130:264270
13. Friess W (1998) Collagen biomaterial for drug delivery. Eur J Pharm Biopharm 45:113136
14. Khor E (1997) Methods for the treatment of collagenous tissues for bioprostheses. Biomaterials
18:95105
15. Jayakrishnan A, Jameela SR (1996) Glutaraldehyde as a xative in bioprosthetic and drug
delivery matrices. Biomaterials 17:471484
18 S. Dinescu et al.
16. Dunn MG, Avasarala PN, Zawadsky JP (1993) Optimization of extruded collagen bers for
ACL reconstruction. J Biomed Mater Res 27:15451552
17. Bigi A, Cojazzi G, Panzavolta S, Rubini K, Roveri N (2001) Mechanical and thermal properties
of gelatin lms at different degrees of glutaraldehyde crosslinking. Biomaterials 22:763768
18. Nimni ME, Cheung DT, Strates B, Kodama M, Sheikh K (1988) Bioprosthesis derived
from cross-linked and chemically modied collagen tissues. In: Collagen and biomechanics,
vol 2. CRC Press, Boca Raton, pp 202206
19. Olde Damink LHH, Dijkstra PJ, van Luyn MJA, van Wachem PB, Nieuwenhuis P, Feijen J
(1985) Glutaraldehyde as a crosslinking agent for collagen-based biomaterials. J Mater Sci
Mater Med 6:460472
20. Speer DP, Chvapil M, Eskelson CD, Ulreich J (1980) Biological effects of residual glutaralde-
hyde in glutaraldehyde-tanned collagen biomaterials. J Biomed Mater Res 14:753764
21. Goissis G, Marcantonio E Jr, Marcantonio RAC, Lia RCC, Cancia DCJ, De Carvalho WM
(1999) Biocompatibility studies of anionic collagen membranes with different degree of
glutaraldehyde cross-linking. Biomaterials 20:2734
22. Tu R, Lu CL, Thzagarajan K, Wang E, Nguyen H, Shen S, Hata C, Quijano RC (1993) Kinetic
study of collagen xation with polyepoxy xatives. J Biomed Mater Res 27:39
23. Nishi C, Nakajima N, Ikada Y (1995) In vitro evaluation of cytotoxicity of diepoxy compounds
used for biomedical modication. J Biomed Mater Res 29:829834
24. Petide H, Rault I, Huc A, Menasche PH, Herbage D (1990) Use of the acyl azide method for
crosslinking collagen-rich tissues such as pericardium. J Biomed Mater Res 24:179187
25. Anselme K, Petite H, Herbage D (1992) Inhibition of calcication in vivo by acyl azide
crosslinking of a collagen-glycosaminoglycan sponge. Matrix 12:264273
26. Liu T, Shi L, Gu Z, Dan W, Dan N (2017) A novel combined polyphenol-aldehyde crosslinking
of collagen lm- applications in biomedical materials. Int J Biol Macromol 101:889895
27. Yang X, Guo L, Fan Y, Zhang X (2013) Preparation and characterization of macromolecule
cross-linked collagen hydrogels for chondrocyte delivery. Int J Biol Macromol 61:487493
28. Peppas NA, Bures P, Leobandung W, Ichikawa H (2000) Hydrogels in pharmaceutical formu-
lations. Eur J Pharm Biopharm 50:2746
29. Sassi ML, Eriksen H, Risteli L, Niemi S, Mansell J, Gowen M, Risteli J (2000) Immunochem-
ical characterization of assay for carboxyterminal telopeptide of human type I collagen: loss of
antigenicity by treatment with cathepsin. Bone 26:367373
30. Kleimann HK, Klebe RJ, Martin GR (1981) Role of collagenous matrices in the adhesion and
growth of cells. J Cell Biol 88:473485
31. Skopinska-Wisniewska J, Kuderko J, Bajek A, Maj M, Sionkowska A, Ziegler-Borowska M
(2016) Collagen/elastin hydrogels cross-linked by squaric acid. Mater Sci Eng C Mater Biol
Appl 60:100108
32. Vulpe R, Le Cerf D, Dulong V, Popa M, Peptu C, Verestiuc L, Picton L (2016) Rheological
study of in-situ crosslinkable hydrogels based on hyaluronanic acid, collagen and sericin. Mater
Sci Eng C Mater Biol Appl 69:388397
33. Ma Z, He Z, Han F, Zhong Z, Chen L, Li B (2016) Preparation of
collagen/hydroxyapatite/alendronate hybrid hydrogels as potential scaffolds for bone
regeneration. Colloids Surf B Biointerfaces 143:8187
34. Demeter M, Virgolici M, Vancea C, Scarisoreanu A, Albu Kaya MG, Meltzer V (2017)
Network structure studies on γirradiated collagenPVP superabsorbent hydrogels. Radiat
Phys Chem 131:5159
35. Deepthi S, Nivedhitha Sundaram M, Deepti Kadavan J, Jayakumar R (2016) Layered chitosan-
collagen hydrogel/aligned PLLA nanober construct for exor tendon regeneration. Carbohydr
Polym 153:492500
36. Nistor MT, Vasile C, Chiriac AP (2015) Hybrid collagen-based hydrogels with embedded
montmorillonite nanoparticles. Mater Sci Eng C Mater Biol Appl 53:212221
37. Burgeson RE, Nimni ME (1992) Molecular structure and tissue distribution. Clin Orthop Relat
Res 282:250272
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 19
38. Hayrapetyan A, Bongio M, Leeuwenburgh SC, Jansen JA, van den Beuken JJ (2016) Effect of
nano-HA/collagen composite hydrogels on osteogenic behaviour of mesenchymal stromal cells.
Stem Cell Rev 12(3):352364
39. Gurumurty B, Bierdeman PC, Janorkar AV (2016) Composition of elastin like polypeptide-
collagen composite scaffold inuences in vitro osteogenic activity of human adipose derived
stem cells. Dent Mater 32(10):12701280
40. Chen L, Wu Z, Zhou Y, Li L, Wang Y, Wang Z, Chen Y, Zhang P (2017) Biomimetic porous
collagen/hydroxyapatite scaffold for bone tissue engineering. J Appl Polym Sci 134(37):45271
41. Hertweck J, Ritz U, Götz H, Schottel PC, Rommens PM, Hofmann A (2018) CD34+ cells 756
seeded in collagen scaffolds promote bone formation in a mouse calvarial defect model. 757
J Biomed Mater Res B Appl Biomater 106(4):15051516
42. Nguyen BB, Moriarty RA, Kamalitdinov T, Etheridge JM, Fisher JP (2017) Collagen hydrogel
scaffold promotes mesenchymal stem cell and endothelial cell coculture for bone tissue
engineering. J Biomed Mater Res A 105(4):11231131
43. Lee HJ, Kim YB, Ahn SH, Lee JS, Jang CH, Yoon H, Chun W, Kim GH (2015) A new approach
for fabricating collagen/ECM-based bioinks using preosteoblasts and human adipose stem cells.
Adv Healthc Mater 244(9):13591368
44. Parmar PA, Skaalure SC, Chow LW, St-Pierre JP, Stoichevska V, Peng YY, Werkmeister JA,
Ramshaw JA, Stevens MM (2015) Temporally degradable collagen-mimetic hydrogels tuned to
chondrogenesis of human mesenchymal stem cells. Biomaterials 99:5671
45. Fensky F, Reichert JC, Traube A, Rackwitz L, Siebenlist S, Nöth U (2014) Chondrogenic
predifferentiation of human mesenchymal stem cells in collagen type I hydrogels. Biomed Tech
(Berl) 59(5):375383
46. Chen X, Zhang F, He X, Xu Y, Yang Z, Chen L, Zhou S, Yang Y, Zhou Z, Sheng W, Zeng Y
(2013) Chondrogenic differentiation of umbilical cord-derived mesenchymal stem cells in type I
collagen-hydrogel for cartilage engineering. Injury 44(4):540549
47. Yuan T, Zhang L, Li K, Fan H, Fan Y, Liang J, Zhang X (2014) Collagen hydrogel as an
immunomodulatory scaffold in cartilage tissue engineering. J Biomed Mater Res B Appl
Biomater 102(2):337344
48. Yang J, Chen X, Yuan T, Yang X, Fan Y, Zhang X (2017) Regulation of the secretion of
immunoregulatory factors of mesenchymal stem cells (MSCs) by collagen-based scaffolds
during chondrogenesis. Mater Sci Eng C Mater Biol Appl 70.(Pt 2:983991
49. Li MT, Ruehle MA, Stevens HY, Servies N, Willett NJ, Karthikeyakannan S, Warren GL,
Guldberg RE, Krishnan L (2017) Skeletal myoblast-seeded vascularized tissue scaffolds in
the treatment of a large volumetric muscle defect in the rat biceps femoris muscle. Tissue Eng
Part A 23:989
50. Xu G, Wang X, Deng C, Teng X, Suuronen EJ, Shen Z, Zhong Z (2015) Injectable biodegrad-
able hybrid hydrogels based on thiolated collagen and oligo(acryloyl carbonate)-poly(ethylene
glycol)-oligo(acryloyl carbonate) copolymer for functional cardiac regeneration. Acta Biomater
15:5564
51. van Marion MH, Bax NA, van Turnhout MC, Mauretti A, van der Schaft DW, Goumans MJ,
Bouten CV (2015) Behavior of CMPCs in unidirectional constrained and stress-free
3D hydrogels. J Mol Cell Cardiol 87:7991
52. Ketabat F, Karkhaneh A, Mehdinavaz Aghdam R, Hossein Ahmadi Tafti S (2017) Injectable
conductive collagen/alginate/polypyrrole hydrogels as a biocompatible system for biomedical
applications. J Biomater Sci Polym Ed 28(8):794805
53. Kaneko A, Matsushita A, Sankai Y (2015) A 3D nanobrous hydrogel and collagen sponge
scaffold promotes locomotor functional recovery, spinal repair, and neuronal regeneration after
complete transection of the spinal cord in adult rats. Biomed Mater 10(1):015008
54. Lee JH, Lee JY, Yang SH, Lee EJ, Kim HW (2014) Carbon nanotube-collagen
three-dimensional culture of mesenchymal stem cells promotes expression of neural phenotypes
and secretion of neurotrophic factors. Acta Biomater 10(10):44254436
20 S. Dinescu et al.
55. Park JW, Kang YD, Kim JS, Lee JH, Kim HW (2014) 3D microenvironment of collagen
hydrogel enhances the release of neurotrophic factors from human umbilical cord blood cells
and stimulates the neurite outgrowth of human neural precursor cells. Biochem Biophys Res
Commun 447(3):400406
56. Roberts MA, Kotha SS, Phong KT, Zheng Y (2016) Micropatterning and assembly of
3D microvessels. J Vis Exp 115:e54457, 110
57. Kuo KC, Lin RZ, Tien HW, Wu PY, Li YC, Melero-Martin JM, Chen YC (2015) Bioengineer-
ing vascularized tissue constructs using an injectable cell-laden enzymatically crosslinked
collagen hydrogel derived from dermal extracellular matrix. Acta Biomater 27:151166
58. Rafat M, Xeroudaki M, Koulikovska M, Sherrell P, Groth F, Fagerholm P, Lagali N (2016)
Composite core-and-skirt collagen hydrogels with differential degradation for corneal thera-
peutic applications. Biomaterials 83:142155
59. Ahn JI, Kuffova L, Merrett K, Mitra D, Forrester JV, Li F, Grifth M (2013) Crosslinked
collagen hydrogels as corneal implants: effects of sterically bulky vs. non-bulky carbodiimides
as crosslinkers. Acta Biomater 9(8):77967805
60. Liu W, Deng C, McLaughlin CR, Fagerholm P, Lagali NS, Heyne B, Scaiano JC, Watsky MA,
Kato Y, Munger R, Shinozaki N, Li F, Grifth M (2009) Collagen-phosphorylcholine
interpenetrating network hydrogels as corneal substitutes. Biomaterials 30(8):15511559
61. Liu W, Merrett K, Grifth M, Fagerholm P, Dravida S, Heyne B, Scaiano JC, Watsky MA,
Shinozaki N, Lagali N, Munger R, Li F (2008) Recombinant human collagen for tissue
engineered corneal substitutes. Biomaterials 29(9):11471158
62. McLaughlin CR, Fagerholm P, Muzakare L, Lagali N, Forrester JV, Kuffova L, Rafat MA,
Liu Y, Shinozaki N, Vascotto SG, Munger R, Grifth M (2008) Regeneration of corneal cells
and nerves in an implanted collagen corneal substitute. Cornea 27(5):580589
63. Jain A, Betancur M, Patel GD, Valmikinathan CM, Mukhatyar VJ, Vakharia A, Pai SB,
Brahma B, MacDonald TJ, Bellamkonda RV (2014) Guiding intracortical brain tumour cells
to an extracortical cytotoxic hydrogel using aligned polymeric nanobers. Nat Mater
13(3):308316
64. Rao SS, Dejesus J, Short AR, Otero JJ, Sarkar A, Winter JO (2013) Glioblastoma behaviors
in three-dimensional collagen-hyaluronan composite hydrogels. ACS Appl Mater Interfaces
5(19):92769284
65. Lungu A, Albu MG, Stancu IC, Florea NM, Vasile E, Iovu H (2013) Superporous collagen-
sericin scaffolds. J Appl Polym Sci 127(3):22692279
66. Mitran V, Albu MG, Vasile E, Cimpean A, Costache M (2015) Dose-related effects of sericin
on preadipocyte behavior within collagen/sericin hybrid scaffolds. Prog Nat Sci Mater Int
25(2):122130
67. Dinescu S, Galateanu B, Albu M, Cimpean A, Dinischiotu A, Costache M (2013) Sericin
enhances the bioperformance of collagen-based matrices preseeded with hADSCs. Int J Mol Sci
14(1):18701889
68. Tsubouchi K, Igarashi Y, Takasu Y, Yamada H (2005) Sericin enhances attachment of cultured
human skin broblasts. Biosci Biotechnol Biochem 69:403405
69. Aramwit P, Kanokpanont S, Nakpheng T, Srichana T (2010) The effects of sericin from various
extraction methods on cell viability and collagen production. Int J Mol Sci 11:22002211
70. Dinescu S, Gălăt
eanu B, Albu M, Lungu A, Radu E, Hermenean A, Costache M (2013)
Biocompatibility assessment of novel collagen-sericin scaffolds improved with hyaluronic
acid and chondroitin sulfate for cartilage regeneration. Biomed Res Int 2013(111):article
ID 598056
71. Kaya DA, Albu MG, Vuluga Z, Duran N, Albu L, Mert A. Collagen biomaterials with zeolite
and essential oils for treatment of skin infections and method for their preparation. National
Patent Application, OSIM no A 01269/29.11.2011
72. Houdek MT, Wyles CC, Stalboerger PG, Terzic A, Behfar A, Moran SL (2016) Collagen
and fractionated platelet-rich plasma scaffold for dermal regeneration. Plast Reconstr Surg
137(5):14981506
Collagen-Based Hydrogels and Their Applications for Tissue Engineering and... 21
... However, it also has some disadvantages, such as poor mechanical strength, low elasticity, poor dimensional and thermal stability. Therefore, to overcome the limitations, different crosslinking methods with other substances are used [27]. ...
... Synthetic polymers such as poly(2-hydroxyethyl methacrylate) (PHEMA), polyethylene glycol (PEG) or polylactic acid (PLA) possess good mechanical strength, but lack bioactivity and in some cases biocompatibility due to inflammatory responses in vivo. Hydrogels consisting of natural polymers, such as collagen [8], hyaluronic acid [9], dextran [10], chitosan [11], or decellularized tissue (ECM) [12,13], are desirable due to their biocompatibility and biodegradability, but often lack sufficient strength to function directly as tissue substitutes. ...
Article
Full-text available
Background There is a great clinical need and it remains a challenge to develop artificial soft tissue constructs that can mimic the biomechanical properties and bioactivity of natural tissue. This is partly due to the lack of suitable biomaterials. Hydrogels made from human placenta offer high bioactivity and represent a potential solution to create animal-free 3D bioprinting systems that are both sustainable and acceptable, as placenta is widely considered medical waste. A combination with silk and gelatin polymers can bridge the biomechanical limitations of human placenta chorion extracellular matrix hydrogels (hpcECM) while maintaining their excellent bioactivity. Method In this study, silk fibroin (SF) and tyramine-substituted gelatin (G-TA) were enzymatically crosslinked with human placental extracellular matrix (hpcECM) to produce silk-gelatin-ECM composite hydrogels (SGE) with tunable mechanical properties, preserved elasticity, and bioactive functions. The SGE composite hydrogels were characterized in terms of gelation kinetics, protein folding, and bioactivity. The cyto- and biocompatibility of the SGE composite was determined by in vitro cell culture and subcutaneous implantation in a rat model, respectively. The most cell-supportive SGE formulation was then used for 3-dimensional (3D) bioprinting that induced chemical crosslinking during extrusion. Conclusion Addition of G-TA improved the mechanical properties of the SGE composite hydrogels and inhibited crystallization and subsequent stiffening of SF for up to one month. SGE hydrogels exhibit improved and tunable biomechanical properties and high bioactivity for encapsulated cells. In addition, its use as a bioink for 3D bioprinting with free reversible embedding of suspended hydrogels (FRESH) has been validated, opening the possibility to fabricate highly complex scaffolds for artificial soft tissue constructs with natural biomechanics in future. Graphical Abstract
... Collagen type 1 is especially abundant and is a key component of bone, cartilage, cornea, tendons, and ligaments [102][103][104]. As an important constituent of the extracellular matrix (ECM), collagen type 1 is widely used as a scaffold material to provide mechanical support and biological cues for cell proliferation and differentiation [105,106]. For example, to develop alternatives to autologous bone grafts in posterolateral spinal fusion, mesenchymal stem cells (MSC) have been cultured in type 1 collagen gels with hydroxyapatite particles, resulting in a favorable degree of spinal fusion in adult rabbits [107]. ...
Article
Full-text available
The periosteum is a thin layer of connective tissue covering bone. It is an essential component for bone development and fracture healing. There has been considerable research exploring the application of the periosteum in bone regeneration since the 19th century. An increasing number of studies are focusing on periosteal progenitor cells found within the periosteum and the use of hydrogels as scaffold materials for periosteum engineering and guided bone development. Here, we provide an overview of the research investigating the use of the periosteum for bone repair, with consideration given to the anatomy and function of the periosteum, the importance of the cambium layer, the culture of periosteal progenitor cells, periosteum-induced ossification, periosteal perfusion, periosteum engineering, scaffold vascularization, and hydrogel-based synthetic periostea. Keywords: periosteum; ossification; hydrogels; cambium layer; progenitor cells; synthetic periosteum; 3-D printing
Article
Pluripotent stem cell‐derived skin organoids (PSOs) recently emerged as a developmental skin model that is self‐organized into multiple components, such as hair follicles. Despite their impressive cellular and anatomical complexity, PSOs are currently generated in the absence of 3D extracellular matrix (ECM) signals and have several major limitations, including an inverted anatomy (e.g., epidermis inside/dermis outside). In this work, we established a method to generate PSOs effectively in a chemically‐defined 3D ECM environment. After examining various dermal ECM molecules, we found that PSOs generated in collagen type I supplemented with laminin 511 exhibit increased growth compared to conventional free‐floating condition, but fail to induce complete skin differentiation due in part to necrosis. We addressed this problem by generating the PSOs in a 3D bioprinted spindle‐shaped hydrogel device, which constrains organoid growth longitudinally. This new culture system significantly reduced organoid necrosis, and led to a two‐fold increase in keratinocyte differentiation and an eight‐fold increase in hair follicle formation. Finally, we adapted the system as a microfluidic device to create asymmetrical gradients of differentiation factors, and improved spatial organization of dermal and epidermal cells. Our study highlights the pivotal role of ECM and morphogen gradients in promoting and spatially‐controlling skin differentiation in the PSO framework. This article is protected by copyright. All rights reserved
Article
Collagen is usually dissolved in the acid solution and neutralized to mix cells for three-dimensional (3D) cell culture. During this process, the acid collagen solution must be precisely adjusted into the pH neutral on the ice platform; after mixing cells, the collagen/cells solution need more than 30 min to gelate at 37 ℃; moreover, the cells embedded in the collagen/cells hydrogel have to be harvested through collagen enzymolysis with enzyme such as collagenase if necessary. Herein, we developed the rapid sol-gel reversible thermosensitive collagen (RRTC) for 3D cell culture. The pH neutral collagen solution could directly mix cells at room temperature; the RRTC/cells solution could gelate in 60 s or less at 37 ℃; especially, the cells embedded in the RRTC/cells hydrogel could be harvested through hydrogel phase transformation from gel to sol. The L929 cells, human umbilical vein endothelial cells (HUVEC), Sp2/0 cells, murine aortic vascular smooth muscle cells (MOVAS), rat intestinal epithelial cells (IEC-6) and human chondrocytes were 3D cultured via the RRTC and grew well. The cells could proliferate by about 10 times in 14 days and preserve the high cell viability of over 90 %. The experiments of mouse skin regeneration demonstrated the RRTC solution could in situ quickly gelate on the skin wound for the skin regeneration. Therefore, this novel collagen biomaterial could be used as an efficient tool to fabricate 3D cell culture and have more clinical applications.
Article
In this study, we extract type I collagen from fish scales and employ an electrostatic self-assembly technique to crosslink it with negatively charged graphene. By incorporating 0%, 1%, 5%, and 10% weight of graphene with collagen, we significantly enhance the mechanical strength, conductivity, and 3D porous structure of the scaffolds. The incorporation of graphene increases the Young’s modulus of the scaffolds threefold compared to pure collagen scaffolds. Impedance measurements reveal values of 4 kΩ, 2.5 kΩ, and 1 kΩ for scaffolds containing 1%, 5%, and 10% weight of graphene with collagen, respectively. The scaffolds demonstrate cell viability above 90%, and the osteogenic differentiation potential, as determined by ALP assay, confirms successful osteogenesis. Moreover, the eco-friendly synthesis route establishes the hybrid 3D graphene-collagen nanocomposite scaffold as a stable material with excellent biocompatible properties in a biological medium.
Article
Full-text available
The development of biomaterial inks suitable for biofabrication and mimicking the physicochemical properties of the extracellular matrix is essential for bioprinting technology in tissue engineering applications. The use of animal-derived proteinous materials, such as jellyfish collagen, or fish scale gelatin, has become an important pillar in biomaterial ink design to increase the bioactivity of hydrogels. However, besides the extraction of proteinous structures, the use of structurally intact fish scales as an additive could increase biocompatibility and bioactivity of hydrogels due to its organic (collagen and gelatin) and inorganic (hydroxyapatite) contents, while simultaneously enhancing mechanical strength in 3D printing applications. To test this hypothesis, we present here a composite biomaterial ink composed of fish scale (FS) and alginate dialdehyde (ADA)-gelatin (GEL) for three-dimensional (3D) bioprinting applications. We fabricate 3D cell-laden hydrogels using mouse pre-osteoblast MC3T3-E1 cells. We evaluate the physicochemical and mechanical properties of FS incorporated ADA-GEL biomaterial inks as well as the bioactivity and cytocompatibility of cell-laden hydrogels. Due to the distinctive collagen orientation of the FS, the compressive strength of the hydrogels significantly increases with increasing FS particle content. Addition of FS also provides a tool to tune hydrogel stiffness. FS particles were homogeneously incorporated into the hydrogels. Particle-matrix integration was confirmed via scanning electron microscopy. FS incorporation in the ADA-GEL matrix increased the osteogenic differentiation of MC3T3-E1 cells in comparison to pristine ADA-GEL, as FS incorporation led to increased ALP activity and osteocalcin secretion of MC3T3-E1 cells. Due to the significantly increased stiffness and supported osteoinductivity of the hydrogels, FS structure as a natural collagen and hydroxyapatite source contributed to the biomaterial ink properties for bone engineering applications. Our findings indicate that ADA-GEL/FS represents a new biomaterial ink formulation with great potential for 3D bioprinting applications, and FS is confirmed as promising additive for bone tissue engineering applications.
Chapter
Chitin is a bio-based and biodegradable polymer since it is the second most abundant natural polysaccharide on earth after cellulose. It is composed of N-acetyl-d-glucosamine, and it is found in different organisms such as in the exoskeleton of crustacean, insects, annelids, mollusks, plants, and some microorganisms. On the other hand, Chitosan is generally obtained by the controlled deacetylation of chitin in the presence of alkali, composed of randomly distributed β-(1–4) linked d-glucosamine (deacetylated unit) and N-acetyl-d-glucosamine (acetylated unit). Chitin and chitosan are different based on the degree of deacetylation. These polysaccharides possess different physicochemical and biological properties which has been modified and/or improved thanks to the development of chitin and chitosan nanocomposites. There are various methods for the development of chitin and chitosan-based nanocomposites, including solvent casting, spray coating, layer-by-layer, and extrusion. Also, it has been combined with different nano compounds to obtain these nano composites. This chapter summarizes some of the most important technologies involved in synthesizing chitin and chitosan nanocomposites, as well as the study of its physicochemical and biological properties, its production costs and ending in the analysis and discussion of the applications that these materials have in various fields.
Chapter
Regenerative medicine has sought new alternatives of materials capable of being used in humans for different purposes, with low or no cytotoxicity, ability to adapt to the body and mainly biodegradation. In this sense, polymeric nanocomposites based on biodegradable polymers have shown a high level of acceptance for this application. Among the most promising biodegradable polymers are polylactic acid (PLA), polyvinyl alcohol (PVA), polyhydroxybutyrate (PHB), polyhydroxyalkanoates (PHAs); which have served as a support or matrix, nevertheless, they lack molecular signals to respond to stimuli from the receiving organism, due to this, different biopolymers have been used such as chitosan, chitin, cellulose, collagen, alginate and combinations of them, to improve the biological response and generate a synergy effect. Biodegradable nanocomposites have been a sensitive topic due to the implications involved in the incorporation and subsequent release of by-products of the degradation of the polymeric material, therefore, it is of great interest to study the possible mechanisms of biodegradation in in vitro and in vivo environments and the response at the cellular level due to the presence of metabolites from the biodegradation of the support matrix, on the immune response within the organism.
Article
Full-text available
Bone tissue engineering (BTE) holds promise for managing the clinical problem of large bone defects. However, clinical adoption of BTE is limited due to limited vascularization of constructs, which could be circumvented by pre-cultivation of osteogenic and endothelial derived cells in natural-based polymer scaffolds. However, until now not many studies compared the effect of mono- and cocultures pre-seeded in collagen before implantation. We utilized a mouse calvarial defect model and compared five groups of collagen scaffolds: a negative control of a collagen scaffold alone, a positive control treated with BMP-7, monocultures of either human osteoblasts (hOBs) or CD34+ cells, and a coculture of hOB and CD34+ cells. Each pre-seeded collagen scaffold was implanted in mice. After 6 weeks mice were sacrificed and their skulls prepared for volumetric and histologic analysis. We found that a monoculture of CD34+ cells and a coculture of hOB and CD34+ cells pre-cultured in the collagen scaffold increased bone regeneration to a similar extend. In these groups, greater amounts of new bone were found compared with hOB monocultures. Interestingly, monoculture of CD34+ cells demonstrated better fracture healing than monoculture of hOBs, emphasizing the possible role of angiogenesis. Our results are promising regarding a cellular based collagen BTE construct, but more work is needed to understand the complex interaction between the osteogenic and endothelial cells. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 2017.
Article
Full-text available
Statement of significance: Hydrogels biomaterials are essential constituting components of engineered tissues with the applications in regenerative medicine and drug delivery. The mechanical properties of hydrogels play crucial roles in regulating the interactions between cells and extracellular matrix and directing the cells phenotype and genotype. Despite significant advances in developing methods and techniques with the ability of tuning the biomechanical properties of hydrogels, there are still challenges regarding the synthesis of hydrogels with complex mechanical profiles as well as limitations in vascularization and patterning of complex structures of natural tissues which barricade the production of sophisticated organs. Therefore, in addition to a review on advanced methods and techniques for measuring a variety of different biomechanical characteristics of hydrogels, the new techniques for enhancing the biomechanics of hydrogels are presented. It is expected that this review will profit future works for regulating the biomechanical properties of hydrogel biomaterials to satisfy the demands of a variety of different human tissues.
Article
Full-text available
Recently, understanding of the extracellular matrix (ECM) has expanded rapidly due to the accessibility of cellular and molecular techniques and the growing potential and value for hydrogels in tissue engineering. The fabrication of hydrogel-based cellular scaffolds for the generation of bioengineered tissues has been based on knowledge of the composition and structure of ECM. Attempts at recreating ECM have used either naturally-derived ECM components or synthetic polymers with structural integrity derived from hydrogels. Due to their increasing use, their biocompatibility has been questioned since the use of these biomaterials needs to be effective and safe. It is not surprising then that the evaluation of biocompatibility of these types of biomaterials for regenerative and tissue engineering applications has been expanded from being primarily investigated in a laboratory setting to being applied in the multi-billion dollar medicinal industry. This review will aid in the improvement of design of non-invasive, smart hydrogels that can be utilized for tissue engineering and other biomedical applications. In this review, the biocompatibility of hydrogels and design criteria for fabricating effective scaffolds are examined. Examples of natural and synthetic hydrogels, their biocompatibility and use in tissue engineering are discussed. The merits and clinical complications of hydrogel scaffold use are also reviewed. The article concludes with a future outlook of the field of biocompatibility within the context of hydrogel-based scaffolds.
Article
Bone defect and osteochondral injury frequently occur due to diseases or traumatism and bring a crucial challenge in orthopedics. The hybrid scaffold has shown promise as a potential strategy for the treatment of such defects. In this study, a novel biomimetic porous collagen (Col)/hydroxyapatite (HA) scaffold was fabricated through assembling layers of Col containing gradual amount of HA under the assistance of “iterative layering” freeze-drying process. The scaffold presents a double gradient of highly interconnected porosity and HA content from top to bottom, mimicking the inherent physiological structure of bone. Owing to the biomimetic structure and component, significant increase of cell proliferation, alkaline phosphatase activity, and osteogenic differentiation in vitro was observed, illustrating potential application of the excellent Col/HA scaffold as a promising strategy for bone tissue engineering. © 2017 Wiley Periodicals, Inc. J. Appl. Polym. Sci. 2017, 134, 45271.
Article
Hydrogels are composed of three-dimensional smart and/or hungry networks, which do not dissolve in water but swell considerably in an aqueous medium, demonstrating an extraordinary ability to absorb water into the reticulated structure. Such inherent feature is a subject of considerable scientific research interest which leads to a dominating path in extending their potential in hi-tech applications. Over the past decades, significant progress has been made in the field of hydrogels. Further, explorations are continuously being made in all directions at an accelerated pace for their extensive usage. In view of this, the present review discusses the subject on the miscellaneous hydrogels with regard to their raw materials, methods of fabrication and applications. In addition, this article summarizes the classification of hydrogels, based on their cross-linking and physical states. Lately, a brief outlook on the future prospects of hydrogels is also presented.
Article
High velocity impact injuries can often result in loss of large skeletal muscle mass, creating defects devoid of matrix, cells, and vasculature. Functional regeneration within these regions of large volumetric muscle loss (VML) continues to be a significant clinical challenge. Large cell-seeded, space-filling tissue engineered constructs that may augment regeneration require adequate vascularization to maintain cell viability. However, the long-term effect of improved vascularization and the effect of addition of myoblasts to vascularized constructs have not been determined in large VMLs. Here, our objective was to create a new VML model, consisting of a full-thickness, single muscle defect, in the rat biceps femoris muscle, and evaluate the ability of myoblast seeded vascularized collagen hydrogel constructs to augment VML regeneration. Adipose derived microvessels were cultured with or without myoblasts to form vascular networks within collagen constructs. In the animal model, the VML injury was created in the left hind limb, and treated with the harvested autograft itself, constructs with microvessels only (MVF), constructs with microvessels and myoblasts (MVF+Myoblasts), or left empty. We evaluated the formation of vascular networks in vitro by light microscopy, and the capacity of vascularized constructs to augment early revascularization and muscle regeneration in the VML using perfusion angiography and creatine kinase activity respectively. Myoblasts (Pax7+) were able to differentiate into myotubes (sarcomeric myosin MF20+) in vitro. The MVF+Myoblasts group showed longer and more branched microvascular networks than the MVF group in vitro, but showed similar overall defect site vascular volumes at 2 weeks post-implantation by ?CT angiography. However, a larger number of small diameter vessels were observed in the vascularized construct treated groups. Yet, both vascularized implant groups showed primarily fibrotic tissue with adipose infiltration, poor maintenance of tissue volume within the VML and little muscle regeneration. These data suggest that while vascularization may play an important supportive role, other factors besides adequate vascularity may determine the fate of regenerating volumetric muscle defects.
Article
Recently, Injectable Conducting Hydrogel (ICH) systems have gained much attention for tissue engineering and regenerative medicine. These systems can promote the regeneration of tissues responding to electrical responses. In this study, a novel ICH system was introduced. To achieve this system, firstly, a soluble non-toxic polypyrrole (PPy) synthesized by grafting pyrrole on alginate (Alg) backbone (Alg-graft-PPy), and then, ICH systems were prepared by the given ratios of Alg-graft-PPy, Alg, and collagen (Col). Three different amounts of Col (0.5, 1, and 1.5 mg/ml) were added to the system including Alg-graft-PPy: Alg wt. % with the ratio of 20:80 and 30:70. FTIR spectroscopy, electrical conductivity, viscosity, syringeability, gelation time, and MTT assay were performed in order to characterize the produced hydrogels. Due to the rheological behavior of 20:80 (Alg-graft-PPy: Alg wt. %), it was recognized more suitable to inject. Also this system associated with 0.5 mg/ml Col introduced as the best sample with respect to its viscosity and injectability. This ICH system has shown high conductivity in addition to a good level of cell viability and syringeability. With respect to properties of the produced ICH system, it can be applied for bone, nerve, muscle and cardiac cells, which respond to electrical impulses.
Article
The generation of functional, vascularized tissues is a key challenge for the field of tissue engineering. Before clinical implantations of such tissue engineered bone constructs can succeed, tactics to promote neovascularization need to be strengthened. We have previously demonstrated that the tubular perfusion system (TPS) bioreactor is an effective culturing method to augment osteogenic differentiation and maintain viability of human mesenchymal stem cells (hMSC). Here, we devised a strategy to address the need for a functional microvasculature by designing an in vitro coculture system that simultaneously cultures osteogenic differentiating hMSCs with endothelial cells (ECs). We utilized the TPS bioreactor as a dynamic coculture environment, which we hypothesize will encourage prevascularization of endothelial cells and early formation of bone tissue and could aid in anastomosis of the graft with the host vasculature after patient implantation. To evaluate the effect of different natural scaffolds for this coculture system, the cells were encapsulated in alginate and/or collagen hydrogel scaffolds. We discovered the necessity of cell-to-cell proximity between the two cell types as well as preference for the natural cell binding capabilities of hydrogels like collagen. We discovered increased osteogenic and angiogenic potential as seen by amplified gene and protein expression of ALP, BMP-2, VEGF, and PECAM. The TPS bioreactor further augmented these expressions, indicating a synergistic effect between coculture and applied shear stress. The development of this dynamic coculture platform for the prevascularization of engineered bone, emphasizing the importance of the construct microenvironments and will advance the clinical use of tissue engineered constructs. This article is protected by copyright. All rights reserved.
Article
Extracellular matrix (ECM) bioscaffolds prepared from decellularized tissues have been used to facilitate constructive and functional tissue remodeling in a variety of clinical applications. The discovery that these ECM materials could be solubilized and subsequently manipulated to form hydrogels expanded their potential in vitro and in vivo utility; i.e. as culture substrates comparable to collagen or Matrigel, and as injectable materials that fill irregularly-shaped defects. The mechanisms by which ECM hydrogels direct cell behavior and influence remodeling outcomes are only partially understood, but likely include structural and biological signals retained from the native source tissue. The present review describes the utility, formation, and physical and biological characterization of ECM hydrogels. Two examples of clinical application are presented to demonstrate in vivo utility of ECM hydrogels in different organ systems. Finally, new research directions and clinical translation of ECM hydrogels are discussed. Statement of Significance More than 70 papers have been published on extracellular matrix (ECM) hydrogels created from source tissue in almost every organ system. The present manuscript represents a review of ECM hydrogels and attempts to identify structure-function relationships that influence the tissue remodeling outcomes and gaps in the understanding thereof. There is a Phase 1 clinical trial now in progress for an ECM hydrogel.