ArticlePDF AvailableLiterature Review

Evaluation and Application of Silk Fibroin Based Biomaterials to Promote Cartilage Regeneration in Osteoarthritis Therapy

MDPI
Biomedicines
Authors:

Abstract and Figures

Osteoarthritis (OA) is a common joint disease characterized by cartilage damage and degeneration. Traditional treatments such as NSAIDs and joint replacement surgery only relieve pain and do not achieve complete cartilage regeneration. Silk fibroin (SF) biomaterials are novel materials that have been widely studied and applied to cartilage regeneration. By mimicking the fibrous structure and biological activity of collagen, SF biomaterials can promote the proliferation and differentiation of chondrocytes and contribute to the formation of new cartilage tissue. In addition, SF biomaterials have good biocompatibility and biodegradability and can be gradually absorbed and metabolized by the human body. Studies in recent years have shown that SF biomaterials have great potential in treating OA and show good clinical efficacy. Therefore, SF biomaterials are expected to be an effective treatment option for promoting cartilage regeneration and repair in patients with OA. This article provides an overview of the biological characteristics of SF, its role in bone and cartilage injuries, and its prospects in clinical applications to provide new perspectives and references for the field of bone and cartilage repair.
This content is subject to copyright.
Citation: Su, X.; Wei, L.; Xu, Z.; Qin,
L.; Yang, J.; Zou, Y.; Zhao, C.; Chen,
L.; Hu, N. Evaluation and
Application of Silk Fibroin Based
Biomaterials to Promote Cartilage
Regeneration in Osteoarthritis
Therapy. Biomedicines 2023,11, 2244.
https://doi.org/10.3390/
biomedicines11082244
Academic Editor: Elisa Belluzzi
Received: 6 June 2023
Revised: 27 July 2023
Accepted: 29 July 2023
Published: 10 August 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
biomedicines
Review
Evaluation and Application of Silk Fibroin Based Biomaterials
to Promote Cartilage Regeneration in Osteoarthritis Therapy
Xudong Su 1, 2, , Li Wei 1, 2, , Zhenghao Xu 1,2, Leilei Qin 1,2, Jianye Yang 1,2, Yinshuang Zou 1,2, Chen Zhao 1,2,
Li Chen 1, 2, *,‡ and Ning Hu 1 ,2 ,*,
1Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University,
Chongqing 400016, China
2Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
*Correspondence: chenlicq@163.com (L.C.); huncqjoint@yeah.net (N.H.)
These authors contributed equally to this work.
These authors also contributed equally to this work.
Abstract:
Osteoarthritis (OA) is a common joint disease characterized by cartilage damage and
degeneration. Traditional treatments such as NSAIDs and joint replacement surgery only relieve
pain and do not achieve complete cartilage regeneration. Silk fibroin (SF) biomaterials are novel
materials that have been widely studied and applied to cartilage regeneration. By mimicking the
fibrous structure and biological activity of collagen, SF biomaterials can promote the proliferation and
differentiation of chondrocytes and contribute to the formation of new cartilage tissue. In addition,
SF biomaterials have good biocompatibility and biodegradability and can be gradually absorbed and
metabolized by the human body. Studies in recent years have shown that SF biomaterials have great
potential in treating OA and show good clinical efficacy. Therefore, SF biomaterials are expected to
be an effective treatment option for promoting cartilage regeneration and repair in patients with OA.
This article provides an overview of the biological characteristics of SF, its role in bone and cartilage
injuries, and its prospects in clinical applications to provide new perspectives and references for the
field of bone and cartilage repair.
Keywords: silk fibroin; osteoarthritis; bone/cartilage repair; treatment
1. Introduction
Due to the gradual increase in the proportion of the aging population, obesity, and joint
injuries worldwide, the risks of bone and cartilage diseases have significantly increased,
seriously affecting people’s quality of life and physical health [
1
]. About 20~33% of the
world’s population suffers from bone and joint diseases, and their incidence in adults is
30~40% [
2
], among which the incidence of osteoporosis [
3
], bone fractures [
4
], cartilage
wear [
5
], and arthritis [
6
] gradually increases with age. Osteoarthritis (OA) is a progressive
degenerative joint disease and the main cause of disability in adults, characterized by
cartilage and subchondral bone degeneration [
7
]. Generally, cartilage and subchondral
bone degeneration are due to increased metalloproteases and inflammatory cytokines; the
excessive mechanical load and pathological factors cause bone and cartilage destruction,
resulting in an imbalance in the dynamic equilibrium of repair and leading to OA. Thus,
repairing damaged cartilage is a fundamental element in the treatment of OA [8,9].
Biomedicines 2023,11, 2244. https://doi.org/10.3390/biomedicines11082244 https://www.mdpi.com/journal/biomedicines
Biomedicines 2023,11, 2244 2 of 21
Although clinical treatments for bone/cartilage-related diseases caused by OA include
pain management, physical therapy, joint injection, and surgical treatment, they only im-
prove the patients’ pain symptoms and do not achieve the goal of cartilage
repair [10,11]
.
Autologous chondrocyte transplantation and allogeneic/autologous cartilage transplanta-
tion have also been applied in clinical practice, but there are still limitations, such as donor
shortage, rejection reactions, and infections. Secondary surgery, limited collection sources,
and complications often make tissue transplantation ineffective as a clinical treatment,
especially for joint cartilage damage and key-sized bone defects [
12
15
]. Autologous and
allogeneic transplantation are common clinical techniques for replacing damaged tissues,
but they are limited by various factors, such as a lack of tissue that can be removed from
healthy areas, and a lack of suitable donors. Allogeneic transplant material from donor
tissue can cause immune responses, and, in cases of extensive injury and large surface
areas, it is difficult to obtain appropriate materials on time, leading to low success rates [
16
].
Tissue engineering (TE) relies on the use of a variety of biocompatible materials to restore,
maintain, and improve tissue function to regenerate injured tissues and organs. These
materials can be seeded with cells and contain various supportive components. In recent
years, TE has gained increasing attention as an alternative method for producing patient-
specific tissues for repair and replacement applications [
17
20
], but various biomaterials
have inherent limitations, so finding an excellent biomaterial has become the focus of
research in recent decades [21].
Silk fibroin (SF) is a common natural material with excellent mechanical properties,
low rejection reaction, tunable biodegradability, and good stability in the field of biomedi-
cal engineering, especially in tissue engineering [
22
24
]. SF is an important extracellular
matrix protein, widely present in bone and cartilage tissues, and has important biological
functions [25]
. SF plays an important role in tissue engineering. Due to its good biocompat-
ibility and biodegradability, SF can be used to build artificial tissues and organs. It can act
as a scaffold or matrix material to promote cell attachment, proliferation, and differentiation
in vivo
and support new tissues. Secondly, SF also promotes wound healing. It has good
biocompatibility and bioactivity to promote the regeneration and repair of traumatized
tissue. SF can promote angiogenesis, accelerate the wound-healing process, and reduce
inflammation. Therefore, SF has a wide range of applications in treating trauma, burns, and
ulcers. In addition, SF is used in the development of drug delivery systems. Due to its good
degradability and drug-modification properties, SF can be used as a carrier to control the
release of drugs. This property makes SF promising, improving traditional drug-delivery
systems, and developing novel drug delivery technologies. In conclusion, SF has a wide
range of roles in biomedical applications. It can be used in tissue engineering, wound repair,
and the development of drug delivery systems, providing valuable tools and materials for
medical research and clinical practice. [
26
28
]. Therefore, SF-based biomaterials have been
used as potential bio-polymer applications in bone/cartilage repair in tissue engineering
(Figure 1) [
29
31
]. This paper introduces the biological characteristics of SF, its role in
bone/cartilage injury, and its clinical applications.
Biomedicines 2023,11, 2244 3 of 21
Figure 1.
SF-based biomaterials in cartilage/osteochondral repair. (
A
) Sequential release of E7/KGN
from silk nanosphere matrix in osteochondral defect repair. Reprinted/adapted with permission
from Ref. [
32
], copyright 2020, Elsevier. (
B
) Mechanically strong silica-silk bioaerogel for bone regener-
ation. Reprinted/adapted with permission from Ref. [
33
], copyright 2019, American Chemical Society.
(
C
) Fabrication of silk/calcium silicate/sodium alginate composite scaffolds. Reprinted/adapted
with permission from Ref. [34], copyright 2018, Elsevier.
2. Biological Properties of SF
Silk is currently an SF raw material that can be mass-produced and has been applied
in clinical practice. As a natural fiber material, it mainly comprises two proteins: SF (SF)
and silk sericin (SS). SF accounts for about 75%, and silk sericin accounts for about 25%.
Silk has good biocompatibility and biodegradability [
35
,
36
]. To obtain pure SF, silk needs to
undergo processes such as degumming, washing, and drying [
37
]. SF is a large molecular
protein composed of 18 amino acids, of which glycine, alanine, and serine account for more
than 80% of the total amino acid content [
38
,
39
]. It is widely distributed in the extracellular
matrix and has various biological functions (Figure 2) [37,40].
Figure 2. SF structure and SF-based materials.
Biomedicines 2023,11, 2244 4 of 21
2.1. Structure of SF
The molecular structure of SF has certain characteristics, consisting of a heavy (H)
chain of 390 kDa and a light (L) chain of 26 kDa connected by disulfide bonds, as well as
a glycoprotein (P25/30 kDa) secreted into the posterior silk gland [
41
43
]. The H chain
accounts for most of the SF, with an amino acid composition of Gly (46%), Ala (30%),
Ser (12%), Tyr (5.3%), and Val (1.8%) [
44
]. Another 4 kDa peptide encoded by the P25
gene is mainly associated with the H-L complex via hydrophobic interactions [
45
]. The
genes encoding the three peptides are located on different chromosomes but appear to be
coordinately regulated in the posterior silk gland [
46
,
47
]. In addition, interactions between
the H chain and the L chain or P25 are crucial for the secretion of SF [
48
,
49
]. The main
body of the SF chain consists of alternating crystalline and non-crystalline regions [
50
,
51
].
The crystalline region is dominated by the GAGAG sequence (Gly-Ala-Gly-Ala-Ala-Gly-
Ser) with short side chains. The arrangement of amino acid residues in the amorphous
region is complex and contains many amino acid residues with long side chains, such as
tyrosine, lysine, and arginine. These residues are relatively hydrophilic, obstructing the
regular assembly and crystallization of the chain segment, resulting in an irregularly coiled
molecular conformation. The mechanical properties of SF can be regulated by changing
the size, number, orientation, and arrangement of the crystalline (silk) and amorphous
regions [
52
,
53
]. The primary crystal structures of SF are Silk I and Silk II, and the water-
soluble and unstable Silk I can be transformed into Silk II with a
β
-fold structure that is
insoluble in water under certain conditions [54,55].
2.2. Properties of SF
The stability and degradation of SF depend on factors such as temperature, pH, oxida-
tion, enzyme action, light, and humidity. Reasonable control of these factors can prolong
the stability of SF. The biological properties of SF are closely related to its structure [
53
,
56
].
Research has shown that SF can bind to receptors on cell membranes, thereby regulat-
ing biological processes such as cell proliferation, differentiation, and migration [
40
]. In
addition, SF can also regulate the synthesis and degradation of the extracellular matrix,
promoting the reconstruction and repair of the extracellular matrix [
57
,
58
]. In bone and
cartilage tissues, SF is an important component that can regulate biological processes such
as cell proliferation, differentiation, and migration [
59
]. The degradation properties of
biomaterials directly affect the speed and quality of cartilage/osteochondral repair. As a
kind of protein material, the degradation rate of SF-based biomaterials is mainly affected
by proteases. Most proteolytic enzymes tend to degrade non-crystalline SF. This suggests
that SF-based biomaterials with controlled degradability can be prepared by changing the
content of crystalline structures [
31
]. The stability and degradation of SF depend on factors
such as temperature, pH, oxidation, enzyme action, light, and humidity. Reasonable control
of these factors can prolong the stability of SF. Due to its biocompatibility, adjustable degra-
dation, unique biomedical and mechanical properties, ease of processing, and abundant
supply, SF can be processed into gels [
60
], films [
61
], nanofibers [
62
], nanoparticles [
63
],
and other materials that can be widely applied in drug delivery [
64
], tissue repair [
65
], and
other fields.
2.3. Preparation Method of SF-Based Biomaterials
At present, the preparation methods of SF-based biomaterials mainly include 3D bio-
printing, electrospinning, and freeze-drying. The main types of 3D bioprinting equipment
include inkjet printing, extrusion printing, and laser-assisted printing [
66
,
67
]. Bio-inks are
very important in 3D printing [
68
]. In general, when using 3D bioprinting to prepare SF-
based biomaterials, it is necessary to modify SF bio-inks to enhance the biological activity of
the SF, and the mechanical strength of SF-based biomaterials can be overcome by modifying
SF-based bio-inks to meet the needs of cartilage/osteochondral repair [
69
,
70
]. Electrospin-
ning can mix multiple matrices and combine the properties of the matrix to suit the needs
of cartilage tissue engineering [
71
]. In addition, electrospinning can maintain the elasticity
Biomedicines 2023,11, 2244 5 of 21
of SF, which is essential for cartilage/osteochondral cartilage tissue
engineering [72]
. In
the process of preparing biomaterials via freeze-drying, technology freezes the solvent and
then sublimates, with little effect on the solute, so freeze-drying also facilitates the carrying
of drugs and growth factors for SF-based biomaterials [73].
2.4. The Main Types of SF-Based Biomaterials
The main types of SF-based biomaterials are hydrogels, scaffolds, and microcarriers.
The native EMC-like microenvironment of hydrogels is suitable for loading cells to promote
cartilage/osteochondral repair [
74
]. At the same time, the mechanical properties, shape,
and swelling properties of hydrogels will change with the changes in temperature, pH
value, and ion concentration, which can achieve the intelligent release of chondrogenic
and osteogenic drugs and closely fit the interface of cartilage defects to improve the
integration effect [
75
]. Scaffolds promote cartilage/osteochondral repair and regeneration
by providing a specific microenvironment [
76
]. Compared with hydrogels, scaffolds have a
fixed shape and higher mechanical strength, which can be used as an adjunct to cell therapy
to promote cell attachment, growth, and differentiation [
77
]. Microcarriers generally refer
to small spherical scaffolds suitable for cell culture, growth, and transport, as they not
only promote cell growth and maintain the cell differentiation phenotype but also enable
tissue regeneration through direct injection into the target site, enabling microcarriers to
accelerate cartilage/osteochondral cartilage repair [
78
]. At the same time, microcarriers can
be loaded with growth factors to promote cartilage/osteoblast adhesion and growth [79].
3. Osteoarthritic Articular Cartilage Model
Articular cartilage plays an important role in joint movement and is very finely and
scientifically structured to suit different functional needs [
80
]. Articular cartilage mainly
comprises a hyaline cartilage layer and a calcified cartilage layer [
81
]. In addition, the
subchondral bone is located directly beneath the articular cartilage, and the hyaline car-
tilage, calcified cartilage, and subchondral bone together form the cartilage complex [
82
].
Hyaline cartilage is composed mainly of oval chondrocytes, an extracellular matrix (con-
taining large amounts of proteoglycan), and type III collagen [
83
]. Articular cartilage is
not innervated or vascularized, and its nutrients must be obtained from the joint fluid [
84
].
The core proteins that aggregate proteoglycans have covalently bound and strongly neg-
atively charged glycosaminoglycan side chains that interact with the surrounding fluid
environment through collagen and proteoglycan non-covalent linkages, giving articular
cartilage its unique biomechanical properties [
85
]. The cells in the calcified cartilage zone
are hypertrophic chondrocytes [
86
], and, because calcified cartilage is more dense and
mineralized, its modulus of elasticity is at the megapascal level, between the kilopascal
level of hyaline cartilage and the quarter-pascal level of bone tissue, which is equivalent
to being a mechanical transition zone that prevents cartilage tissue from being damaged
when subjected to excessive loading, and also effectively disperses the concentrated stress
on cartilage tissue under shear [
87
]. In addition, the presence of a physiological demarca-
tion line (tidal line) between the hyaline cartilage layer and the mineralized edge of the
calcified layer indicates that the articular cartilage has significantly developed and that the
growth-plate cartilage is fixed to the epiphysis, sometimes through a thin layer of calcified
cartilage and tidal markings, while the hypertrophic edge does not form tidal markings
and undergoes continuous vascular infiltration and endochondral ossification (EO) until
the bone matures, and the growth plate is completely resorbed and replaced by bone [88].
The pathological changes of OA initially occur in the hyaline cartilage layer, which in
turn causes changes in the calcified cartilage layer [
89
,
90
]. The onset of OA initially causes
alterations in the spatial structure of the proteoglycan and collagen fibers of the hyaline
cartilage layer, with swelling of the collagen fibers and an increase in free water [
91
,
92
]. The
calcified cartilage layer, an important structure for conducting stress with the tide line under
repeated compression, undergoes structural pathological changes, especially in the weight-
bearing area, where microfractures appear [
93
]. The persistent production of microfractures
Biomedicines 2023,11, 2244 6 of 21
causes the invasion of neovascular tissue and mineralization of the tideline, exacerbating
the degenerative process of the hyaline cartilage [
94
,
95
]. As OA progresses, the loss of
proteoglycans (PGs) from the cartilage tissue intensifies, and the collagen fibers continue to
swell, causing the calcified layer to thin, micro-fissures to increase, and new capillaries to
invade the calcified layer [
96
,
97
]. When cartilage tissue loses the protection of the calcified
layer, a vicious cycle is formed, and the OA process is accelerated (Figure 3) [98].
Figure 3.
Schematic diagram of articular cartilage. Reprinted/adapted with permission from Ref. [
31
],
copyright 2020, Ivyspring.
4. The Role of SF in Bone/Cartilage Damage
SF plays a key role in chondrocyte differentiation. SF is a protein found primarily
in collagen fibers that helps form and maintain the structure and function of cartilage
tissue. During chondrocyte differentiation, SF forms a complete extracellular matrix envi-
ronment by interacting with other extracellular matrix components such as collagen and
glycosaminoglycans. This environment provides the necessary support and structure for
chondrocytes to grow and differentiate. In addition, SF is also involved in key pathways
that regulate cell signaling and cell function. It can affect cell proliferation, differentiation,
and migration by binding to cell surface receptors (Figure 4). SF can also interact with
intracellular signaling pathways, such as TGF-
β
and BMP, to regulate the direction and
speed of cell differentiation [
99
,
100
]. Recent studies have shown that SF plays an important
role in the repair and regeneration of bone/chondrogenic tissue (Table 1) [101,102].
Figure 4. The role of SF in bone/cartilage damage.
Biomedicines 2023,11, 2244 7 of 21
Table 1. Application of SF in bone tissue regeneration.
Application Areas Mechanism of Action Application Results References
Bone regeneration
and repair
Promotes osteoblast proliferation and
differentiation, bone matrix production,
and epiphyseal migration
Promotes the speed of fracture healing,
enhances fracture stability, and
promotes bone defect repair
[31,69]
Cartilage regeneration
and repair
Promotes proliferation and differentiation
of chondroblasts and synthesis of collagen
and cartilage matrix
Promotes healing of cartilage defects
and improves cartilage tissue structure
and functional recovery
[99,100]
Bone implant repair
Provides an extracellular matrix scaffold to
improve the biocompatibility and adhesion
of bone implants
Enhances the bonding of the bone
implant to the surrounding tissue and
promotes stability and growth of the
bone implant
[101]
Oral periodontal
restoration
Promotes the growth of dental bone
attachment tissue and soft tissue repair
Improves the effectiveness of
periodontitis treatment and promotes
oral wound healing
[102]
Other applications Various tissue engineering repairs,
angiogenesis, immunomodulation, etc.
SF has potential for a wide range of
applications in tissue engineering and
regenerative medicine
[44]
4.1. The Role of SF in Bone Tissue
Bone tissue is one of the hardest tissues in the body, and damage and lesions often lead
to fractures, osteoporosis, and other diseases [
103
,
104
]. Recent studies have shown that SFs
play an important role in bone tissue [
105
108
]. As a natural polymer material with good
elasticity, tensile strength, biocompatibility, and biodegradability, SF can provide sufficient
space for the growth and differentiation of bone cells, thus promoting the generation of
new bone tissue [
109
113
]. SF has been extensively studied in bone TE because of its high
toughness, mechanical strength, and proven biocompatibility. Meinel et al. combined bone
tissue engineering, gene therapy based on human mesenchymal stem cells (MSCs), and
SF biomaterials to investigate the effect of viral transfection on MSC osteogenic properties
in vitro
, and showed that RSF scaffolds promote osteogenic differentiation of human
mesenchymal stem cells (HMSCs)
in vitro
[
113
]. Meanwhile, Meinel et al. implanted porous
SF-based scaffolds into cranial defects in mice, demonstrating bone tissue regeneration
using silk-based implants with engineered bone, and expanding the selection of protein-
based bone implant materials through mechanical stability and durability [
114
]. RSF
scaffolds can be combined with other biomaterials, such as collagen or calcium phosphate-
based inorganic components, to enhance osteogenic properties [
115
]. To increase the success
of bone regeneration, accelerated angiogenesis is required [
116
]. Farokhi et al. used bio-
mixed SF/calcium phosphate/PLGA nanocomposite scaffolds as a vascular endothelial
growth factor (VEGF) to explore the efficacy of the delivery system, which also showed
good effects [117].
Bioactive molecules and active cells are a hot topic in tissue engineering research,
with various molecules and cells providing additional regulatory cues to guide cell dif-
ferentiation and functional bone regeneration [
118
]. Growth factors, drugs, and different
stem cells have been introduced into filament-based scaffolds to promote bone formation
(Figure 5) [119].
4.1.1. Bioactive Factor-Based Biomaterials
Bioactive factor-based biomaterials can release bioactive factors and promote bone
tissue repair by regulating cell proliferation and differentiation [
120
]. Drug-loaded beads
and colloidal crystals can be used to achieve a controlled release of bioactive factors through
microstructural alterations to improve bone repair [
121
,
122
]. Shen et al. developed an
SF/nano-hydroxyapatite (nHAp)-based scaffold with sequential and sustained release of
SDF-1 and BMP-2 in SF/nHAp scaffolds with synergistic effects on bone regeneration [
123
].
Biomedicines 2023,11, 2244 8 of 21
Exosome-encapsulated silk frames have also been shown to promote the recovery of bone
defects in vivo [124].
Figure 5. The role of silk fibroin in cartilage tissue engineering.
4.1.2. Biodegradable Polymer-Based Biomaterials
Biodegradable polymeric biomaterials are biomaterials with good biocompatibility and
tunability, which can be modified in terms of composition, structure, and physicochemical
properties to achieve a variety of different functions, such as cell adhesion, biodegrada-
tion, and drug retardation [
125
127
]. Currently, common biodegradable polymer-based
biomaterials include polylactic acid and polycaprolactone [
128
,
129
]. Diaz-Gomez et al.
prepared composite scaffolds using various combinations of PCL, SF, and nanohydrox-
yapatite (nHA), confirming the synergistic effect of silk and nHA on the extent of bone
repair [
130
]. As a biodegradable polymer film, the silk in protein/chitosan composite film
can be used not only as a metal implant coating for bone injury repair but also as a tissue
engineering scaffold for skin, cornea, adipose, and other soft tissue injury repair, while the
silk in protein/chitosan film not only provided a comparable environment for the growth
and proliferation of rat bone marrow-derived mesenchymal stem cells but also promoted
their osteogenic and lipogenic differentiation [
131
]. Biodegradable polymeric composites
of SF are widely used in tracheal tissue engineering [
132
], wound dressings [
133
], and
materials for biomedical applications [134].
4.1.3. Calcium- and Phosphorus-Based Biomaterials
Calcium and phosphorus biomaterials are biomaterials that can form,
in vivo
, similar
to bone tissue, and can promote bone tissue repair, for example, by promoting the prolif-
eration and differentiation of bone cells [
135
]. For example, hydroxyapatite and calcium
tri-calcium phosphate can be used as carriers of bone growth factors to promote bone tissue
repair through the slow release of bone growth factors [
136
,
137
]. At the same time, the
significant anti-stress properties of SF in concert with
β
-tricalcium phosphate provided a
good bionic environment for bone marrow MSCs [
138
]. The SF/calcium phosphate material
has good biocompatibility and mechanical properties as a bone repair scaffold, providing a
good growth space for osteoblast differentiation. In addition, the injectability exhibited by
the hydrogel and its use as a drug carrier allows the hydrogel to fit closely to the interface
of the cartilage defect, thus improving the integration effect [139].
4.2. The Role of SF in Cartilage Tissue
Cartilage tissue is an elastic connective tissue whose main role is to cushion and
support bone [
140
,
141
]. Injuries and lesions to cartilage tissue often lead to diseases such
as OA and cartilage lesions [
142
]. SF is associated with the extracellular matrix widely
distributed in cartilage tissue, and its main role is to maintain the structure and function
Biomedicines 2023,11, 2244 9 of 21
of the cartilage tissue [
143
]. SFs can promote the proliferation and differentiation of
chondrocytes and the generation of new cartilage tissue [
144
]. In addition, SFs can regulate
the apoptosis and survival of chondrocytes, and promote the repair and regeneration of
cartilage tissue [145].
4.2.1. Application of SFs in Cartilage Tissue Engineering
Cartilage tissue engineering is a method of repairing and reconstructing cartilage
tissue using biomaterials such as cells, matrix materials, and growth factors [
146
]. SF is a
natural protein that has many similarities with the protein structure of human skin and
has good biocompatibility and biodegradability [
28
]. In cartilage tissue engineering, SF
can be used to construct biocompatible, biodegradable scaffold materials, and can likewise
be used as surface modifiers to enhance scaffold surface cell adhesion and proliferation
properties (Figure 6) [
147
]. Studies have shown that SFs can promote the proliferation
and differentiation of chondrocytes, thereby promoting the growth and repair of cartilage
tissue [
101
]. In addition, SFs can improve the strength and stability of cartilage tissue, thus
increasing the success rate of cartilage tissue engineering [
148
]. Additionally, silk in proteins
has good water retention and can provide a good environment under growth conditions
to aid chondrocytes in nascent tissue growth and repair [
149
]. Saha et al. evaluated the
role of mulberry and non-mulberry laminar filament biomaterials in cartilage or bone
induction using human bone marrow stromal cells (hBMSC)
in vivo
and
in vitro
, showing
good bone induction [
150
]. In cartilage tissue engineering, combining SF with other
biomaterials, such as gelatin and alginate, supports the fabrication of scaffold materials
with good biocompatibility and biodegradable properties that can provide skeletal support
for the generation of new tissue, and can degrade into harmless metabolites with reduced
adverse effects on the human body [
151
,
152
]. Li et al. developed a silk-composite hydrogel
of SF and carboxymethyl chitosan (CMCS), which supported the adhesion, proliferation,
glycosaminoglycan synthesis, and chondrogenic phenotype of rabbit articular chondrocytes,
and the subcutaneous implantation of the hydrogel in mice showed no infection or local
inflammatory response, indicating good biocompatibility
in vivo
[
153
]. Liu et al. used
electrostatic spinning to prepare a fibrillar SF/poly L-lactic acid (PLLA) scaffold, showing
good adhesion, biocompatibility, and cytocompatibility [
154
]. Overall, SF, as an important
matrix material, has good prospects for application in cartilage tissue engineering. With a
better understanding of silk, we will be better able to apply this natural protein to promote
cartilage tissue repair and regeneration.
4.2.2. SF in the Treatment of Patients with OA
OA is a serious disease derived from the degeneration of cartilage tissue and, if nec-
essary, requires surgical intervention. Tissue engineering using stem cell graft scaffolds
is an attractive approach and a challenge for orthopedic surgery [
137
,
138
]. SFs can also
reduce pain and inflammatory responses in patients with arthritis, thereby relieving the
symptoms of arthritis. Wang et al. prepared silk/BDDE hydrogel balls using oil/water
(o/w) emulsification and evaluated their biocompatibility and biodegradability
in vivo
.
The silk/BDDE hydrogel ball was demonstrated to be biocompatible and can be used as
a lubricant for the treatment of OA, as well as for pain relief and the sustained release
of drugs for future OA treatment [
155
]. In clinical trials, Sharafat-Vaziri et al. evaluated
an autologous chondrocyte and collagen/silk heart protein scaffold consisting of newly
engineered tissues to repair osteochondral defects, showing great coverage and integra-
tion of the grafts in patients without effusion, edema, and reduced cartilage formation
signals [
156
]. Jaipaew et al. prepared SF/hyaluronic acid (HA) scaffolds, with different
SF/HA (w/w) ratios via freeze-drying, which were suitable for OA surgery [
157
]. Thus,
SF-based biomaterials have great promise in OA surgery.
Biomedicines 2023,11, 2244 10 of 21
Figure 6. In vivo
implantation of SF-based biomaterials. (
A
) Schematic diagram illustrating the
3D fabrication of a scaffold made via bioprinting for
in vivo
implantation. Reprinted/adapted
with permission from Ref. [
151
], copyright 2017, John Wiley and Sons. (
B
) Silk-GMA hydrogel
transplantation loaded with chondrocytes. Reprinted/adapted with permission from Ref. [
140
],
copyright 2020, Elsevier.
4.2.3. Application of SFs in Drug Delivery
SF is a biomaterial that has been extensively studied in tissue engineering and drug
delivery (Figure 7). As a carrier material, SF has high permeability, and its microporous
structure can promote the penetration of drugs and improve the delivery efficiency of
drugs. At the same time, SF can be degraded and absorbed by the human body, avoiding
the risk of secondary surgery. Additionally, SF can protect the drug from degradation
and inactivation during delivery and improve the stability of the drug. Moreover, SF
has low immunogenicity and histocompatibility, reducing rejection of the drug delivery
system [
158
]. At present, SF can be used as a carrier for drugs in OA drug delivery,
enveloping drugs in nanoparticles or microspheres to increase the efficiency and stability of
drug delivery. Additionally, it can be combined with growth factors or cytokines through
controlled release, promote the proliferation and differentiation of chondrocytes, and
promote the repair and regeneration of joint cartilage. At the same time, it can combine
with stem cells or osteoblasts to form a tissue-engineered scaffold for the repair and
regeneration of joint cartilage [
159
161
]. In addition, SFs can be used in combination
with other biomaterials to further enhance their drug delivery [
162
]. Ratanavaraporn
loaded previously developed gelatin/SF microspheres with curcumin, and the gelatin/SF
microspheres encapsulated with curcumin delayed cell destruction in joint and synovial
tissue, which showed prolonged anti-inflammatory effects compared to rapidly degrading
gelatin microspheres [
163
]. Red-modified silk nanoparticles were fabricated by Sharma
et al. and loaded with gentamicin as a deposition material on titanium surfaces, which
showed better killing of S. aureus on the titanium surfaces [
164
]. Hassani Besheli et al.
constructed a sustained drug delivery system by loading vancomycin (VANCO) in silk
nanoparticles to treat severe osteomyelitis [
165
]. Thus, in OA drug delivery, SF can play an
important role (Table 2).
Biomedicines 2023,11, 2244 11 of 21
Figure 7. The role of SF in drug delivery.
Table 2. Application of SF in drug delivery.
Application Areas SF in Drug Delivery References
Oncology treatment To deliver drugs to tumor tissue, SFs are used as carriers to improve
the stability and bioavailability of drugs [166]
Wound healing
SFs promote cell migration, proliferation, and repair during wound
healing and can be used to prepare drug-delivery systems to promote
wound healing
[167]
Treatment of blood disorders
SFs provide reliable carriers for the transport and release of drugs for
the treatment of blood disorders, such as anticoagulants and
anti-platelet agents
[168]
Treatment of neurological disorders
SFs can be used to deliver drugs for the treatment of neurological
disorders, such as neuroprotective agents and anti-epileptic drugs, to
promote the protection and repair of nerve cells
[169]
Skin beauty and treatment
SFs are widely used in cosmetic skin products and delivery systems
for therapeutic drugs to improve skin texture, promote wound
healing, reduce scar formation, etc.
[170]
Infectious disease control
SFs are used as carriers for drug delivery systems to deliver antiviral,
antibacterial, and antifungal drugs, improving their efficacy
and bioaccessibility
[171]
Treatment of orthopedic diseases
SFs are used to deliver drugs for treating orthopedic diseases, such as
bone growth factors and anti-inflammatory drugs, and to promote
the growth and repair of bone cells
[106,107]
Cardiovascular disease treatment
SFs are used as carriers in drug delivery systems for the delivery of
drugs treating cardiovascular disease, such as anti-hypertensives and
anti-heart failure drugs, to alleviate the symptoms of
cardiovascular disease
[172]
Immune disease treatment
SFs can be used in drug delivery systems to deliver drugs for treating
immune diseases, such as anti-inflammatory drugs and
immunomodulators, to regulate the function of the immune system
and treat diseases
[102]
Dental treatment
SFs are widely used in dental therapeutic drug delivery systems for
the delivery of antibacterial drugs, natural anti-inflammatory agents,
and bone growth factors to promote dental restoration and healing
[173]
4.3. Prospects for SF in Clinical Applications
As a natural polymer with good biocompatibility, SF has been widely used in biomed-
ical applications, including ophthalmic surgical sutures, artificial corneas, artificial tendons,
Biomedicines 2023,11, 2244 12 of 21
orthopedic ligaments, cartilage engineering, artificial skin for wound surfaces in trauma-
tology, and anticoagulation stents in cardiology [
174
176
]. Although there is extensive
research into the application of SFs in different base materials, most of it is still at the
laboratory research stage, and few products have been successfully commercialized and are
actually used in clinical treatment (Table 3). SERI surgical scaffolds, Silk Voice injections,
and silk-substituted isoserine trauma dressings are the products in routine clinical use
today (Table 4).
Table 3. Selected SF material development companies.
Company Name Region SF Products
Sofregen Inc. US SERI surgical stents
Injectable fillers
Vaxess Technologies Inc. US Drug and vaccine delivery
Evolved by nature US
Skincare products, textile coatings, topical ophthalmic treatments, etc.
Cocoon Biotech Inc. US Drug delivery systems (hydrogels, osteoarticular microspheres, etc.)
Kraig Biocraft Laboratories Inc. US Special textiles
Oxford Biomaterials Ltd. UK Artificial blood vessels
Orthox Ltd. UK Meniscal repair stents, tissue stents
Suzhou Semtex Biotechnology Co. China Injectable gels, stents, and dressings
Suzhou Suhao Biomaterials Technology Co.
China Trauma dressings
Zhejiang Xingyue Biotechnology Co. China Raw materials such as SF gels, microspheres, solutions, and sponges
SF skin rejuvenation mask
Table 4. Some approved SF products.
Product Type Trademarks Uses Time to Market
SERI Surgical Stent Allergan
Full body contouring, brachioplasty,
abdominoplasty, breast fixation, breast
reconstruction, etc.
2013
Silk Voice Injection Sofregen Vocal cord dielectricity and vocal cord
insufficiency United States, 2018
Silk-substituted isoserine
wound dressing Soho Biotechnology Co. Wound healing China, 2012
With increasing in-depth research on SF, its application in the treatment and repair
of bone and cartilage tissues is becoming more promising. In regenerative medicine, the
binding of SF to cells or growth factors can promote cell attachment, proliferation, and differ-
entiation, while providing structural support to help rebuild tissues. The biocompatibility
and biodegradability of SF fibers make them ideal materials for manufacturing artificial
blood vessels, and SF can be used in the preparation of biomedical dressings to promote the
healing process of wounds. SF dressings provide a protective physical barrier to promote
cell regrowth and tissue repair, and SF can be used as a vehicle for drug delivery systems.
SF has good drug adsorption and sustained release performance, which can control the
release rate and dose of drugs and improve the efficacy and bioavailability of drugs; SF
can also be used to prepare artificial bone substitutes for bone-tissue engineering and bone
repair. Moreover, an SF scaffold can promote the attachment and growth of bone cells and
accelerate the process of bone regeneration and repair. SF has wide application prospects
in regenerative medicine, which can promote tissue regeneration and repair and provide
new treatments and means for disease treatment and rehabilitation [
155
,
177
,
178
]. In terms
of applications, most are concentrated in the fields of wound repair dressings [
179
,
180
]
and orthopedic repair materials [
31
]; in terms of morphology, SF membranes [
181
] and SF
scaffolds [
182
] are the materials with more applications. SF can be combined with other
biomaterials to form composite scaffolds, mimic the natural
in vivo
environment, increase
the
in vivo
fusion potential of scaffolds, implant destination cells to accelerate the healing
and regeneration of trauma sites, etc. It can be modified into different scaffolds such as
Biomedicines 2023,11, 2244 13 of 21
injectable and printable gels, porous sponges, and electrostatically spun two- and three-
dimensional structures [
130
,
134
]. The advantages of SF therapy are its obvious effects, low
side effects, and high safety, and more possibilities for SF-based scaffolds can be expected
through new modification techniques. In the future, the application of SF in the treatment
and repair of bone and cartilage tissues will be even more promising, while more research
is needed to explore its mechanism of action and dose determination.
5. Conclusions and Outlook
Repairing cartilage and osteochondral damage due to OA has always been a challenge
for clinicians. Currently, tissue engineering techniques have been the subject of much
research and have great potential in bone/cartilage repair. SF, as a natural material, is
widely used in tissue engineering due to its inexpensive availability, excellent biocom-
patibility, unique mechanical properties, and desirable processing properties. This paper
summarizes the progress of research on SF biomaterials in the field of bone/cartilage repair.
Compared with other types of biomaterials, SF has great research value and broad appli-
cation prospects. SF-based biomaterials have more robust mechanical properties through
loading peptides, gene-editing means, exosomes, nanoparticles, and growth factors, which
are more conducive to cell adhesion and growth and enhance bone/cartilage repair. For
example, Ding et al. significantly improved the cell recruitment ability of SF/HA scaffolds
by loading bone morphogenetic protein-2 (BMP-2) onto SF/HA scaffolds to accelerate
osteochondral repair [
183
], proliferation, and differentiation, promoting bone repair and
cartilage repair [184].
SF has good biocompatibility, biodegradability, and bio-absorbability, and has been
widely used in the medical field in recent years. Currently, clinical applications regarding
SF biomaterials are mainly focused on wound healing and wound dressings [
114
]. In
addition, there have also been clinical studies on SF scaffolds for meniscal cartilage repair,
with some success in recent years [
185
]. However, for OA and cartilage regeneration, SF
has some limitations. First of all, OA is a joint disease whose main features are cartilage
degeneration and joint inflammation. While SF can help maintain cartilage structure,
it does not prevent or reverse cartilage degeneration. This means that SF has a limited
therapeutic effect on OA. Secondly, SF also has certain limitations on cartilage regeneration.
Cartilage regeneration is a complex biological process that includes steps such as stem
cell differentiation, cell proliferation, and secretion of a matrix. Although SF has a certain
effect on cartilage structure, it does not directly promote cartilage regeneration. When
treating cartilage damage or joint degenerative diseases, stem cell therapy, biomaterial
implantation, or surgery are often required to promote cartilage regeneration and repair.
Overall, while SF plays an important role in maintaining cartilage structure and function, it
has certain limitations in OA and cartilage regeneration. The treatment of these conditions
often requires a combination of factors and treatments to achieve the best results [186].
In conclusion, SF-based biomaterials have broad application potential in the treatment
of OA, and future research can combine SF biomaterials with other bioactive molecules,
such as growth factors and gene therapy, to improve the effect of cartilage regeneration.
At the same time, according to the specific situation of patients, the personalized design
of SF biomaterials can help achieve better treatment results. More importantly, as the
preparation process of SF biomaterials is further improved, the mechanical properties and
stability of the materials are improved, and their degradation rate
in vivo
is prolonged.
Although SF biomaterials have achieved some success in the treatment of OA, they still face
some challenges and limitations. More clinical research and scientific exploration will help
further develop and refine this treatment, providing better treatment options for patients
with OA.
Biomedicines 2023,11, 2244 14 of 21
Author Contributions:
X.S., L.W. and L.Q.: data curation, formal analysis, writing—original draft,
software. J.Y.: resources, formal analysis. C.Z.: writing—review and editing. Z.X.: resources, formal
analysis. Y.Z.: resources, formal analysis. L.C.: conceptualization, formal analysis,
writing—review
and editing. N.H.: conceptualization, formal analysis, funding acquisition,
writing—review
and
editing, project administration. All authors have read and agreed to the published version of
the manuscript.
Funding:
General Project of National Natural Science Foundation of China (Project number: 82072443),
Excellent Project of Chongqing Overseas Returnee Entrepreneurship and Innovation Support Program
(Project number: CX2022032), General Project of Chongqing Technology Innovation and Applica-
tion Development Special Project (Project number: Cstc2020jscx-msxmx0094), the key project of
Chongqing Science and Health Joint Medical Research Project (Project number: 2019ZDXM014).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement:
The data presented in this study are available on request from the
corresponding author.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Qiao, K.; Xu, L.; Tang, J.; Wang, Q.; Lim, K.S.; Hooper, G.; Woodfield, T.B.F.; Liu, G.; Tian, K.; Zhang, W.; et al. The advances in
nanomedicine for bone and cartilage repair. J. Nanobiotechnol. 2022,20, 141. [CrossRef] [PubMed]
2.
Tuckermann, J.; Adams, R.H. The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat. Rev.
Rheumatol. 2021,17, 608–620. [CrossRef]
3.
Camacho, P.M.; Petak, S.M.; Binkley, N.; Diab, D.L.; Eldeiry, L.S.; Farooki, A.; Harris, S.T.; Hurley, D.L.; Kelly, J.; Lewiecki, E.M.;
et al. American association of clinical endocrinologists/american college of endocrinology clinical practice guidelines for the
diagnosis and treatment of postmenopausal osteoporosis—2020 update. Endocr. Pract. 2020,26, 564–570. [CrossRef]
4.
Chakhtoura, M.; Dagher, H.; Sharara, S.; Ajjour, S.; Chamoun, N.; Cauley, J.; Mahfoud, Z.; Boudreau, R.; El Hajj Fuleihan, G.
Systematic review of major osteoporotic fracture to hip fracture incidence rate ratios worldwide: Implications for Fracture Risk
Assessment Tool (FRAX)-derived estimates. J. Bone Miner. Res. 2021,36, 1942–1956. [CrossRef] [PubMed]
5.
Akizuki, S.; Yasukawa, Y.; Takizawa, T. Does arthroscopic abrasion arthroplasty promote cartilage regeneration in osteoarthritic
knees with eburnation? A prospective study of high tibial osteotomy with abrasion arthroplasty versus high tibial osteotomy
alone. Arthroscopy 1997,13, 9–17. [CrossRef]
6. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016,88, 2023–2038. [CrossRef]
7.
Vina, E.R.; Kwoh, C.K. Epidemiology of osteoarthritis: Literature update. Curr. Opin. Rheumatol.
2018
,30, 160–167. [CrossRef]
[PubMed]
8.
Litwic, A.; Edwards, M.H.; Dennison, E.M.; Cooper, C. Epidemiology and burden of osteoarthritis. Br. Med. Bull.
2013
,105,
185–199. [CrossRef] [PubMed]
9.
Li, G.; Yin, J.; Gao, J.; Cheng, T.S.; Pavlos, N.J.; Zhang, C.; Zheng, M.H. Subchondral bone in osteoarthritis: Insight into risk factors
and microstructural changes. Arthritis. Res. Ther. 2013,15, 223. [CrossRef]
10.
Medical Advisory Secretariat. Arthroscopic lavage and debridement for osteoarthritis of the knee: An evidence-based analysis.
Ont. Health Technol. Assess. Ser. 2005,5, 1–37.
11.
Xia, B.; Chen, D.; Zhang, J.; Hu, S.; Jin, H.; Tong, P. Osteoarthritis pathogenesis: A review of molecular mechanisms. Calcif. Tissue.
Int. 2014,95, 495–505. [CrossRef]
12.
Vonk, L.A.; Roël, G.; Hernigou, J.; Kaps, C.; Hernigou, P. Role of Matrix-Associated Autologous Chondrocyte Implantation with
Spheroids in the Treatment of Large Chondral Defects in the Knee: A Systematic Review. Int. J. Mol. Sci.
2021
,22, 7149. [CrossRef]
13.
Liu, Y.; Ma, Y.; Zhang, J.; Yuan, Y.; Wang, J. Exosomes: A Novel Therapeutic Agent for Cartilage and Bone Tissue Regeneration.
Dose Response 2019,17, 1559325819892702.
14.
Kolambkar, Y.M.; Boerckel, J.D.; Dupont, K.M.; Bajin, M.; Huebsch, N.; Mooney, D.J.; Hutmacher, D.W.; Guldberg, R.E. Spa-
tiotemporal delivery of bone morphogenetic protein enhances functional repair of segmental bone defects. Bone
2011
,49, 485–492.
[CrossRef]
15.
Medvedeva, E.V.; Grebenik, E.A.; Gornostaeva, S.N.; Telpuhov, V.I.; Lychagin, A.V.; Timashev, P.S.; Chagin, A.S. Repair of
Damaged Articular Cartilage: Current Approaches and Future Directions. Int. J. Mol. Sci. 2018,19, 2366. [CrossRef]
16.
Cheng, G.; Davoudi, Z.; Xing, X.; Yu, X.; Cheng, X.; Li, Z.; Deng, H.; Wang, Q. Advanced SF Biomaterials for Cartilage
Regeneration. ACS Biomater. Sci. Eng. 2018,4, 2704–2715.
17.
Clark, R.A.; Ghosh, K.; Tonnesen, M.G. Tissue engineering for cutaneous wounds. J. Investig. Dermatol.
2007
,127, 1018–1029.
[CrossRef] [PubMed]
Biomedicines 2023,11, 2244 15 of 21
18.
Snyder, Y.; Jana, S. Strategies for development of decellularized heart valve scaffolds for tissue engineering. Biomaterials
2022
,
288, 121675. [CrossRef] [PubMed]
19.
Tian, L.; Ma, J.; Li, W.; Zhang, X.; Gao, X. Microfiber Fabricated via Microfluidic Spinning toward Tissue Engineering Applications.
Macromol. Biosci. 2023,23, e2200429. [CrossRef]
20. Petre, D.G.; Leeuwenburgh, S.C.G. The Use of Fibers in Bone Tissue Engineering. Tissue Eng. Part B Rev. 2022,28, 141–159.
21.
Li, G.; Sun, S. Silk Fibroin-Based Biomaterials for Tissue Engineering Applications. Molecules
2022
,27, 2757. [CrossRef] [PubMed]
22.
Zhang, C.; Zhang, Y.; Shao, H.; Hu, X. Hybrid Silk Fibers Dry-Spun from Regenerated Silk Fibroin/Graphene Oxide Aqueous
Solutions. ACS Appl. Mater. Interfaces 2016,8, 3349–3358. [CrossRef] [PubMed]
23.
Song, W.; Muthana, M.; Mukherjee, J.; Falconer, R.J.; Biggs, C.A.; Zhao, X. Magnetic-Silk Core-Shell Nanoparticles as Potential
Carriers for Targeted Delivery of Curcumin into Human Breast Cancer Cells. ACS Biomater. Sci. Eng.
2017
,3, 1027–1038.
[CrossRef]
24.
Melke, J.; Midha, S.; Ghosh, S.; Ito, K.; Hofmann, S. Silk fibroin as biomaterial for bone tissue engineering. Acta. Biomater.
2016
,31,
1–16. [CrossRef]
25.
Huang, W.; Ling, S.; Li, C.; Omenetto, F.G.; Kaplan, D.L. Silkworm silk-based materials and devices generated using bio-
nanotechnology. Chem. Soc. Rev. 2018,47, 6486–6504. [CrossRef]
26.
Dong, Q.; Cai, J.; Wang, H.; Chen, S.; Liu, Y.; Yao, J.; Shao, Z.; Chen, X. Artificial ligament made from silk protein/Laponite hybrid
fibers. Acta Biomater. 2020,106, 102–113. [CrossRef]
27.
Ma, D.; Wang, Y.; Dai, W. Silk fibroin-based biomaterials for musculoskeletal tissue engineering. Mater. Sci. Eng. C Mater. Biol.
Appl. 2018,89, 456–469. [CrossRef]
28.
Wang, J.; Yang, Q.; Cheng, N.; Tao, X.; Zhang, Z.; Sun, X.; Zhang, Q. Collagen/silk fibroin composite scaffold incorporated with
PLGA microsphere for cartilage repair. Mater. Sci. Eng. C Mater. Biol. Appl. 2016,61, 705–711. [CrossRef]
29.
Sheng, R.; Chen, J.; Wang, H.; Luo, Y.; Liu, J.; Chen, Z.; Mo, Q.; Chi, J.; Ling, C.; Tan, X.; et al. Nanosilicate-Reinforced Silk
Fibroin Hydrogel for Endogenous Regeneration of Both Cartilage and Subchondral Bone. Adv. Healthc. Mater.
2022
,11, e2200602.
[CrossRef]
30.
Jeyakumar, V.; Amraish, N.; Niculescu-Morsza, E.; Bauer, C.; Pahr, D.; Nehrer, S. Decellularized Cartilage Extracellular Matrix
Incorporated Silk Fibroin Hybrid Scaffolds for Endochondral Ossification Mediated Bone Regeneration. Int. J. Mol. Sci.
2021
,
22, 4055. [CrossRef]
31.
Zhou, Z.; Cui, J.; Wu, S.; Geng, Z.; Su, J. Silk fibroin-based biomaterials for cartilage/osteochondral repair. Theranostics
2022
,12,
5103–5124. [CrossRef] [PubMed]
32.
Zhang, W.; Ling, C.; Zhang, A.; Liu, H.; Jiang, Y.; Li, X.; Sheng, R.; Yao, Q.; Chen, J. An all-silk-derived functional nanosphere
matrix for sequential biomolecule delivery and in situ osteochondral regeneration. Bioact. Mater.
2020
,5, 832–843. [CrossRef]
[PubMed]
33.
Maleki, H.; Shahbazi, M.A.; Montes, S.; Hosseini, S.H.; Eskandari, M.R.; Zaunschirm, S.; Verwanger, T.; Mathur, S.; Milow,
B.; Krammer, B.; et al. Mechanically Strong Silica-Silk Fibroin Bioaerogel: A Hybrid Scaffold with Ordered Honeycomb
Micromorphology and Multiscale Porosity for Bone Regeneration. ACS Appl. Mater. Interfaces 2019,11, 17256–17269. [PubMed]
34.
Zheng, A.; Cao, L.; Liu, Y.; Wu, J.; Zeng, D.; Hu, L.; Zhang, X.; Jiang, X. Biocompatible silk/calcium silicate/sodium alginate
composite scaffolds for bone tissue engineering. Carbohydr. Polym. 2018,199, 244–255. [CrossRef]
35.
Chen, F.; Porter, D.; Vollrath, F. Structure and physical properties of silkworm cocoons. J. R. Soc. Interface
2012
,9, 2299–2308.
[CrossRef]
36.
Zhang, Y.; Sheng, R.; Chen, J.; Wang, H.; Zhu, Y.; Cao, Z.; Zhao, X.; Wang, Z.; Liu, C.; Chen, Z.; et al. Silk Fibroin and Sericin
Differentially Potentiate the Paracrine and Regenerative Functions of Stem Cells through Multiomics Analysis. Adv. Mater.
2023
,
13, e2210517.
37.
Wang, C.; Xia, K.; Zhang, Y.; Kaplan, D.L. Silk-Based Advanced Materials for Soft Electronics. Acc. Chem. Res.
2019
,52, 2916–2927.
[CrossRef]
38.
Zhang, Y.Q.; Ma, Y.; Xia, Y.Y.; Shen, W.D.; Mao, J.P.; Zha, X.M.; Shirai, K.; Kiguchi, K. Synthesis of silk fibroin-insulin bioconjugates
and their characterization and activities in vivo. J. Biomed. Mater. Res. B Appl. Biomater. 2006,79, 275–283.
39.
Carrascoza Mayen, J.F.; Lupan, A.; Cosar, C.; Kun, A.Z.; Silaghi-Dumitrescu, R. On the roles of the alanine and serine in the
β-sheet structure of fibroin. Biophys. Chem. 2015,197, 10–17.
40.
Zhao, Y.; Zhu, Z.S.; Guan, J.; Wu, S.J. Processing, mechanical properties and bio-applications of silk fibroin-based high-strength
hydrogels. Acta Biomater. 2021,125, 57–71. [CrossRef]
41.
Takei, F.; Kikuchi, Y.; Kikuchi, A.; Mizuno, S.; Shimura, K. Further evidence for importance of the subunit combination of silk
fibroin in its efficient secretion from the posterior silk gland cells. J. Cell Biol. 1987,105, 175–180.
42.
Tanaka, K.; Mori, K.; Mizuno, S. Immunological identification of the major disulfide-linked light component of silk fibroin.
J. Biochem. 1993,114, 1–4.
43.
Inoue, S.; Tanaka, K.; Arisaka, F.; Kimura, S.; Ohtomo, K.; Mizuno, S. Silk fibroin of Bombyx mori is secreted, assembling a
high molecular mass elementary unit consisting of H-chain, L-chain, and P25, with a 6:6:1 molar ratio. J. Biol. Chem.
2000
,275,
40517–40528. [CrossRef] [PubMed]
44.
Zhou, C.Z.; Confalonieri, F.; Medina, N.; Zivanovic, Y.; Esnault, C.; Yang, T.; Jacquet, M.; Janin, J.; Duguet, M.; Perasso, R.; et al.
Fine organization of Bombyx mori fibroin heavy chain gene. Nucleic Acids Res. 2000,28, 2413–2419. [CrossRef]
Biomedicines 2023,11, 2244 16 of 21
45.
Tanaka, K.; Kajiyama, N.; Ishikura, K.; Waga, S.; Kikuchi, A.; Ohtomo, K.; Takagi, T.; Mizuno, S. Determination of the site of
disulfide linkage between heavy and light chains of silk fibroin produced by Bombyx mori. Biochim. Biophys. Acta
1999
,1432,
92–103. [PubMed]
46.
Hui, C.C.; Matsuno, K.; Suzuki, Y. Fibroin gene promoter contains a cluster of homeodomain binding sites that interact with three
silk gland factors. J. Mol. Biol. 1990,213, 651–670. [CrossRef] [PubMed]
47.
Kikuchi, Y.; Mori, K.; Suzuki, S.; Yamaguchi, K.; Mizuno, S. Structure of the Bombyx mori fibroin light-chain-encoding gene:
Upstream sequence elements common to the light and heavy chain. Gene 1992,110, 151–158. [PubMed]
48.
Tomeh, M.A.; Hadianamrei, R.; Zhao, X. Silk Fibroin as a Functional Biomaterial for Drug and Gene Delivery. Pharmaceutics
2019
,
11, 494. [CrossRef]
49.
Qi, Y.; Wang, H.; Wei, K.; Yang, Y.; Zheng, R.Y.; Kim, I.S.; Zhang, K.Q. A Review of Structure Construction of Silk Fibroin
Biomaterials from Single Structures to Multi-Level Structures. Int. J. Mol. Sci. 2017,18, 237. [CrossRef]
50.
Zhou, C.Z.; Confalonieri, F.; Jacquet, M.; Perasso, R.; Li, Z.G.; Janin, J. Silk fibroin: Structural implications of a remarkable amino
acid sequence. Proteins 2001,44, 119–122. [CrossRef]
51.
Boi, M.; Marchiori, G.; Berni, M.; Gambardella, A.; Salamanna, F.; Visani, A.; Bianchi, M.; Fini, M.; Filardo, G. Nanoindentation:
An advanced procedure to investigate osteochondral engineered tissues. J. Mech. Behav. Biomed. Mater.
2019
,96, 79–87. [CrossRef]
52.
Lu, Q.; Zhu, H.; Zhang, C.; Zhang, F.; Zhang, B.; Kaplan, D.L. Silk self-assembly mechanisms and control from thermodynamics
to kinetics. Biomacromolecules 2012,13, 826–832.
53.
Rockwood, D.N.; Preda, R.C.; Yücel, T.; Wang, X.; Lovett, M.L.; Kaplan, D.L. Materials fabrication from Bombyx mori silk fibroin.
Nat. Protoc. 2011,6, 1612–1631. [CrossRef]
54. Kratky, O.; Schauenstein, E.; Sekora, A. An Unstable Lattice in Silk Fibroin. Nature 1950,165, 319–320.
55.
Cebe, P.; Partlow, B.P.; Kaplan, D.L.; Wurm, A.; Zhuravlev, E.; Schick, C. Silk I and Silk II studied by fast scanning calorimetry.
Acta Biomater. 2017,55, 323–332. [CrossRef] [PubMed]
56.
Xiao, Y.; Liu, Y.; Zhang, W.; Qi, P.; Ren, J.; Pei, Y.; Ling, S. Formation, Structure, and Mechanical Performance of Silk Nanofibrils
Produced by Heat-Induced Self-Assembly. Macromol. Rapid Commun. 2021,42, e2000435. [PubMed]
57.
Chen, W.; Xu, Y.; Li, H.; Dai, Y.; Zhou, G.; Zhou, Z.; Xia, H.; Liu, H. Tanshinone IIA Delivery Silk Fibroin Scaffolds Significantly
Enhance Articular Cartilage Defect Repairing via Promoting Cartilage Regeneration. ACS Appl. Mater. Interfaces
2020
,12,
21470–21480. [CrossRef]
58.
Hashimoto, T.; Kojima, K.; Tamada, Y. Higher Gene Expression Related to Wound Healing by Fibroblasts on Silk Fibroin
Biomaterial than on Collagen. Molecules 2020,25, 1939. [CrossRef]
59.
Xiao, H.; Huang, W.; Xiong, K.; Ruan, S.; Yuan, C.; Mo, G.; Tian, R.; Zhou, S.; She, R.; Ye, P.; et al. Osteochondral repair using
scaffolds with gradient pore sizes constructed with silk fibroin, chitosan, and nano-hydroxyapatite. Int. J. Nanomed.
2019
,14,
2011–2027.
60.
Koullali, B.; Zhang, Y.; Peterson, A.; Raia, N.; Kaplan, D.L.; House, M.D. Cervical Augmentation with an Injectable Silk-Based Gel:
Biocompatibility in a Rat Model of Pregnancy. Reprod. Sci. 2020,27, 1215–1221.
61.
Fountain, J.N.; Hawker, M.J.; Hartle, L.; Wu, J.; Montanari, V.; Sahoo, J.K.; Davis, L.M.; Kaplan, D.L.; Kumar, K. Towards Non-stick
Silk: Tuning the Hydrophobicity of Silk Fibroin Protein. Chembiochem 2022,23, e202200429.
62.
Sun, S.; Ding, C.; Liu, X.; Zhao, Y.; Zhang, J.; Ding, Q.; Zhang, Y.; Zhang, Y.; Hao, M.; Zheng, Y.; et al. Silk pro-
tein/polyvinylpyrrolidone nanofiber membranes loaded with puerarin accelerate wound healing in mice by reducing
the inflammatory response. Biomater. Adv. 2022,135, 212734.
63.
Liang, Y.; Tang, B.; Sharma, A.; Perera, D.; Allardyce, B.J.; Ghosh, S.; Schniepp, H.C.; Rajkhowa, R. Silk Protein Paper with In Situ
Synthesized Silver Nanoparticles. Macromol. Biosci. 2021,21, e2000357. [CrossRef] [PubMed]
64.
Griswold, E.; Cappello, J.; Ghandehari, H. Silk-elastinlike protein-based hydrogels for drug delivery and embolization. Adv. Drug
Deliv. Rev. 2022,191, 114579. [CrossRef]
65.
Kim, S.H.; Hong, H.; Ajiteru, O.; Sultan, M.T.; Lee, Y.J.; Lee, J.S.; Lee, O.J.; Lee, H.; Park, H.S.; Choi, K.Y.; et al. 3D bioprinted silk
fibroin hydrogels for tissue engineering. Nat. Protoc. 2021,16, 5484–5532. [CrossRef] [PubMed]
66. Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014,32, 773–785. [CrossRef]
67.
Trucco, D.; Sharma, A.; Manferdini, C.; Gabusi, E.; Petretta, M.; Desando, G.; Ricotti, L.; Chakraborty, J.; Ghosh, S.; Lisignoli, G.
Modeling and Fabrication of Silk Fibroin-Gelatin-Based Constructs Using Extrusion-Based Three-Dimensional Bioprinting. ACS
Biomater. Sci. Eng. 2021,7, 3306–3320. [CrossRef] [PubMed]
68.
Jia, L.; Hua, Y.; Zeng, J.; Liu, W.; Wang, D.; Zhou, G.; Liu, X.; Jiang, H. Bioprinting and regeneration of auricular cartilage using a
bioactive bioink based on microporous photocrosslinkable acellular cartilage matrix. Bioact. Mater. 2022,16, 66–81.
69.
Li, Z.; Zhang, X.; Yuan, T.; Zhang, Y.; Luo, C.; Zhang, J.; Liu, Y.; Fan, W. Addition of Platelet-Rich Plasma to Silk Fibroin Hydrogel
Bioprinting for Cartilage Regeneration. Tissue Eng. Part A 2020,26, 886–895. [CrossRef]
70.
Deng, C.; Yang, J.; He, H.; Ma, Z.; Wang, W.; Zhang, Y.; Li, T.; He, C.; Wang, J. 3D bio-printed biphasic scaffolds with dual
modification of silk fibroin for the integrated repair of osteochondral defects. Biomater. Sci. 2021,9, 4891–4903. [CrossRef]
71.
Liu, J.; Fang, Q.; Yu, X.; Wan, Y.; Xiao, B. Chitosan-Based Nanofibrous Membrane Unit with Gradient Compositional and
Structural Features for Mimicking Calcified Layer in Osteochondral Matrix. Int. J. Mol. Sci.
2018
,19, 2330. [CrossRef] [PubMed]
72.
Zhang, F.; Zuo, B.; Fan, Z.; Xie, Z.; Lu, Q.; Zhang, X.; Kaplan, D.L. Mechanisms and control of silk-based electrospinning.
Biomacromolecules 2012,13, 798–804. [CrossRef]
Biomedicines 2023,11, 2244 17 of 21
73.
Kankala, R.K.; Zhu, K.; Li, J.; Wang, C.S.; Wang, S.B.; Chen, A.Z. Fabrication of arbitrary 3D components in cardiac surgery: From
macro-, micro- to nanoscale. Biofabrication 2017,9, 032002. [CrossRef] [PubMed]
74.
Guvendiren, M.; Burdick, J.A. Engineering synthetic hydrogel microenvironments to instruct stem cells. Curr. Opin. Biotechnol.
2013,24, 841–846. [CrossRef]
75. Li, J.; Mooney, D.J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 2016,1, 16071. [CrossRef]
76.
Thorrez, L.; Shansky, J.; Wang, L.; Fast, L.; VandenDriessche, T.; Chuah, M.; Mooney, D.; Vandenburgh, H. Growth, differentiation,
transplantation and survival of human skeletal myofibers on biodegradable scaffolds. Biomaterials 2008,29, 75–84.
77.
Ran, J.; Fei, Y.; Wang, C.; Ruan, D.; Hu, Y.; Zheng, Z.; Chen, X.; Yin, Z.; Tang, C.; Chen, Y.; et al. An Off-the-Shelf Tissue Engineered
Cartilage Composed of Optimally Sized Pellets of Cartilage Progenitor/Stem Cells. ACS Biomater. Sci. Eng.
2021
,7, 881–892.
[CrossRef]
78.
Kankala, R.K.; Zhao, J.; Liu, C.G.; Song, X.J.; Yang, D.Y.; Zhu, K.; Wang, S.B.; Zhang, Y.S.; Chen, A.Z. Highly Porous Microcarriers
for Minimally Invasive In Situ Skeletal Muscle Cell Delivery. Small 2019,15, e1901397. [CrossRef]
79.
Wang, X.; Yucel, T.; Lu, Q.; Hu, X.; Kaplan, D.L. Silk nanospheres and microspheres from silk/pva blend films for drug delivery.
Biomaterials 2010,31, 1025–1035. [CrossRef]
80.
Hua, X.; Shu, L.; Li, J. Multiscale modelling for investigating the long-term time-dependent biphasic behaviour of the articular
cartilage in the natural hip joint. Biomech. Model. Mechanobiol. 2022,21, 1145–1155. [CrossRef]
81. Boyde, A. The Bone Cartilage Interface and Osteoarthritis. Calcif. Tissue Int. 2021,109, 303–328.
82.
Karns, M.R.; Epperson, R.T.; Baran, S.; Nielsen, M.B.; Taylor, N.B.; Burks, R.T. Revisiting the Anterior Glenoid: An Analysis of the
Calcified Cartilage Layer, Capsulolabral Complex, and Glenoid Bone Density. Arthroscopy 2018,34, 2309–2318. [CrossRef]
83.
Schreiner, A.J.; Stoker, A.M.; Bozynski, C.C.; Kuroki, K.; Stannard, J.P.; Cook, J.L. Clinical Application of the Basic Science of
Articular Cartilage Pathology and Treatment. J. Knee Surg. 2020,33, 1056–1068. [CrossRef] [PubMed]
84.
Suri, S.; Gill, S.E.; Massena de Camin, S.; Wilson, D.; McWilliams, D.F.; Walsh, D.A. Neurovascular invasion at the osteochondral
junction and in osteophytes in osteoarthritis. Ann. Rheum. Dis. 2007,66, 1423–1428. [CrossRef] [PubMed]
85.
Mostakhdemin, M.; Nand, A.; Ramezani, M. Articular and Artificial Cartilage, Characteristics, Properties and Testing
Approaches—A Review. Polymers 2021,13, 2000. [CrossRef]
86.
Deng, B.; Wang, F.; Yin, L.; Chen, C.; Guo, L.; Chen, H.; Gong, X.; Li, Y.; Yang, L. Quantitative study on morphology of calcified
cartilage zone in OARSI 04 cartilage from osteoarthritic knees. Curr. Res. Transl. Med. 2016,64, 149–154. [CrossRef] [PubMed]
87.
Ealla, K.K.R.; Veeraraghavan, V.P.; Ravula, N.R.; Durga, C.S.; Ramani, P.; Sahu, V.; Poola, P.K.; Patil, S.; Panta, P. Silk Hydrogel for
Tissue Engineering: A Review. J. Contemp. Dent. Pract. 2022,23, 467–477. [PubMed]
88.
Hoemann, C.D.; Lafantaisie-Favreau, C.H.; Lascau-Coman, V.; Chen, G.; Guzmán-Morales, J. The cartilage-bone interface. J. Knee
Surg. 2012,25, 85–97. [CrossRef] [PubMed]
89.
Abramoff, B.; Caldera, F.E. Osteoarthritis: Pathology, Diagnosis, and Treatment Options. Med. Clin. N. Am.
2020
,104, 293–311.
[CrossRef]
90.
Geyer, M.; Schönfeld, C. Novel Insights into the Pathogenesis of Osteoarthritis. Curr. Rheumatol. Rev.
2018
,14, 98–107. [CrossRef]
91. Moo, E.K.; Tanska, P.; Federico, S.; Al-Saffar, Y.; Herzog, W.; Korhonen, R.K. Collagen fibres determine the crack morphology in
articular cartilage. Acta Biomater. 2021,126, 301–314. [PubMed]
92.
Michalek, A.J.; Kuxhaus, L.; Jaremczuk, D.; Zaino, N.L. Proteoglycans contribute locally to swelling, but globally to compressive
mechanics, in intact cervine medial meniscus. J. Biomech. 2018,74, 86–91. [CrossRef] [PubMed]
93.
Wolschrijn, C.F.; Weijs, W.A. Development of the subchondral bone layer of the medial coronoid process of the canine ulna. Anat.
Rec. Part A Discov. Mol. Cell. Evol. Biol. 2005,284, 439–445. [CrossRef] [PubMed]
94.
Liang, G.; Vanhouten, J.; Macica, C.M. An atypical degenerative osteoarthropathy in Hyp mice is characterized by a loss in the
mineralized zone of articular cartilage. Calcif. Tissue Int. 2011,89, 151–162.
95.
Qian, J.J.; Xu, Q.; Xu, W.M.; Cai, R.; Huang, G.C. Expression of VEGF-A Signaling Pathway in Cartilage of ACLT-induced
Osteoarthritis Mouse Model. J. Orthop. Surg. Res. 2021,16, 379. [CrossRef]
96.
Matsuzaki, T.; Alvarez-Garcia, O.; Mokuda, S.; Nagira, K.; Olmer, M.; Gamini, R.; Miyata, K.; Akasaki, Y.; Su, A.I.; Asahara, H.;
et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci. Transl.
Med. 2018,10, eaan0746. [CrossRef]
97.
Thonar, E.J.; Manicourt, D.M.; Williams, J.; Lenz, M.E.; Sweet, M.B.; Schnitzer, T.J.; Otten, L.; Glant, T.; Kuettner, K.E. Circulating
keratan sulfate: A marker of cartilage proteoglycan catabolism in osteoarthritis. J. Rheumatol. Suppl. 1991,27, 24–26.
98.
Slovacek, H.; Khanna, R.; Poredos, P.; Poredos, P.; Jezovnik, M.; Hoppensteadt, D.; Fareed, J.; Hopkinson, W. Interrelationship of
MMP-9, Proteoglycan-4, and Inflammation in Osteoarthritis Patients Undergoing Total Hip Arthroplasty. Clin. Appl. Thromb.
Hemost. 2021,27, 1076029621995569. [CrossRef]
99.
Wu, T.; Chen, Y.; Liu, W.; Tong, K.L.; Suen, C.W.; Huang, S.; Hou, H.; She, G.; Zhang, H.; Zheng, X.; et al. Ginsenoside Rb1/TGF-
β
1
loaded biodegradable silk fibroin-gelatin porous scaffolds for inflammation inhibition and cartilage regeneration. Mater. Sci. Eng.
C Mater. Biol. Appl. 2020,111, 110757.
100.
Zhang, W.; Zhang, Y.; Zhang, A.; Ling, C.; Sheng, R.; Li, X.; Yao, Q.; Chen, J. Enzymatically crosslinked silk-nanosilicate reinforced
hydrogel with dual-lineage bioactivity for osteochondral tissue engineering. Mater. Sci. Eng. C Mater. Biol. Appl.
2021
,127, 112215.
[CrossRef]
Biomedicines 2023,11, 2244 18 of 21
101.
Wu, Y.; Zhou, L.; Li, Y.; Lou, X. Osteoblast-derived extracellular matrix coated PLLA/silk fibroin composite nanofibers promote
osteogenic differentiation of bone mesenchymal stem cells. J. Biomed. Mater. Res. A 2022,110, 525–534. [CrossRef]
102.
Geão, C.; Costa-Pinto, A.R.; Cunha-Reis, C.; Ribeiro, V.P.; Vieira, S.; Oliveira, J.M.; Reis, R.L.; Oliveira, A.L. Thermal annealed silk
fibroin membranes for periodontal guided tissue regeneration. Journal of materials science. J. Mater. Sci. Mater. Med.
2019
,30, 27.
[PubMed]
103.
Pignolo, R.J.; Wang, H.; Kaplan, F.S. Fibrodysplasia Ossificans Progressiva (FOP): A Segmental Progeroid Syndrome. Front.
Endocrinol. 2020,10, 908. [CrossRef] [PubMed]
104.
Ribeiro, V.P.; Pina, S.; Costa, J.B.; Cengiz, I.F.; García-Fernández, L.; Fernández-Gutiérrez, M.D.M.; Paiva, O.C.; Oliveira, A.L.;
San-Román, J.; Oliveira, J.M.; et al. Enzymatically Cross-Linked Silk Fibroin-Based Hierarchical Scaffolds for Osteochondral
Regeneration. ACS Appl. Mater. Interfaces 2019,11, 3781–3799. [CrossRef] [PubMed]
105.
Bhattacharjee, P.; Kundu, B.; Naskar, D.; Kim, H.W.; Maiti, T.K.; Bhattacharya, D.; Kundu, S.C. Silk scaffolds in bone tissue
engineering: An overview. Acta Biomater. 2017,63, 1–17. [CrossRef] [PubMed]
106.
Li, M.; You, J.; Qin, Q.; Liu, M.; Yang, Y.; Jia, K.; Zhang, Y.; Zhou, Y. A Comprehensive Review on Silk Fibroin as a Persuasive
Biomaterial for Bone Tissue Engineering. Int. J. Mol. Sci. 2023,24, 2660. [CrossRef]
107.
Choi, J.H.; Kim, D.K.; Song, J.E.; Oliveira, J.M.; Reis, R.L.; Khang, G. Silk Fibroin-Based Scaffold for Bone Tissue Engineering. Adv.
Exp. Med. Biol. 2018,1077, 371–387. [PubMed]
108. Kasoju, N.; Bora, U. Silk fibroin in tissue engineering. Adv. Healthc. Mater. 2012,1, 393–412. [CrossRef]
109.
Cai, Y.; Guo, J.; Chen, C.; Yao, C.; Chung, S.M.; Yao, J.; Lee, I.S.; Kong, X. Silk fibroin membrane used for guided bone tissue
regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2017,70, 148–154. [CrossRef]
110.
Luetchford, K.A.; Chaudhuri, J.B.; De Bank, P.A. Silk fibroin/gelatin microcarriers as scaffolds for bone tissue engineering. Mater.
Sci. Eng. C Mater. Biol. Appl. 2020,106, 110116. [CrossRef]
111.
Fitzpatrick, V.; Martín-Moldes, Z.; Deck, A.; Torres-Sanchez, R.; Valat, A.; Cairns, D.; Li, C.; Kaplan, D.L. Functionalized 3D-
printed silk-hydroxyapatite scaffolds for enhanced bone regeneration with innervation and vascularization. Biomaterials
2021
,
276, 120995. [CrossRef] [PubMed]
112.
Jouan, Y.; Bouchemla, Z.; Bardèche-Trystram, B.; Sana, J.; Andrique, C.; Ea, H.K.; Richette, P.; Latourte, A.; Cohen-Solal, M.; Hay, E.
Lin28a induces SOX9 and chondrocyte reprogramming via HMGA2 and blunts cartilage loss in mice. Sci. Adv.
2022
,8, eabn3106.
[PubMed]
113.
Meinel, L.; Hofmann, S.; Betz, O.; Fajardo, R.; Merkle, H.P.; Langer, R.; Evans, C.H.; Vunjak-Novakovic, G.; Kaplan, D.L.
Osteogenesis by human mesenchymal stem cells cultured on silk biomaterials: Comparison of adenovirus mediated gene transfer
and protein delivery of BMP-2. Biomaterials 2006,27, 4993–5002. [PubMed]
114.
Meinel, L.; Fajardo, R.; Hofmann, S.; Langer, R.; Chen, J.; Snyder, B.; Vunjak-Novakovic, G.; Kaplan, D. Silk implants for the
healing of critical size bone defects. Bone 2005,37, 688–698. [CrossRef]
115.
He, P.; Sahoo, S.; Ng, K.S.; Chen, K.; Toh, S.L.; Goh, J.C. Enhanced osteoinductivity and osteoconductivity through hydroxyapatite
coating of silk-based tissue-engineered ligament scaffold. J. Biomed. Mater. Res. A 2013,101, 555–566. [CrossRef]
116.
Ghanaati, S.; Unger, R.E.; Webber, M.J.; Barbeck, M.; Orth, C.; Kirkpatrick, J.A.; Booms, P.; Motta, A.; Migliaresi, C.; Sader, R.A.;
et al. Scaffold vascularization
in vivo
driven by primary human osteoblasts in concert with host inflammatory cells. Biomaterials
2011,32, 8150–8160. [CrossRef]
117.
Farokhi, M.; Mottaghitalab, F.; Shokrgozar, M.A.; Ai, J.; Hadjati, J.; Azami, M. Bio-hybrid silk fibroin/calcium phosphate/PLGA
nanocomposite scaffold to control the delivery of vascular endothelial growth factor. Mater. Sci. Eng. C Mater. Biol. Appl.
2014
,35,
401–410. [CrossRef]
118.
Ho-Shui-Ling, A.; Bolander, J.; Rustom, L.E.; Johnson, A.W.; Luyten, F.P.; Picart, C. Bone regeneration strategies: Engineered
scaffolds, bioactive molecules and stem cells current stage and future perspectives. Biomaterials 2018,180, 143–162. [CrossRef]
119.
Zhang, W.; Zhu, C.; Ye, D.; Xu, L.; Zhang, X.; Wu, Q.; Zhang, X.; Kaplan, D.L.; Jiang, X. Porous silk scaffolds for delivery of
growth factors and stem cells to enhance bone regeneration. PLoS ONE 2014,9, e102371.
120.
Xu, C.; Sun, Y.; Jansen, J.A.; Li, M.; Wei, L.; Wu, Y.; Liu, Y. Calcium phosphate ceramics and synergistic bioactive agents for
osteogenesis in implant dentistry. Tissue Eng. Part C Methods 2023,29, 197–215. [CrossRef]
121.
Lv, L.; Cheng, W.; Wang, S.; Lin, S.; Dang, J.; Ran, Z.; Zhu, H.; Xu, W.; Huang, Z.; Xu, P.; et al. Poly(
β
-amino ester) Dual-Drug-
Loaded Hydrogels with Antibacterial and Osteogenic Properties for Bone Repair. ACS Biomater. Sci. Eng.
2023
,9, 1976–1990.
[CrossRef] [PubMed]
122.
Liu, K.; Li, L.; Chen, J.; Li, Y.; Wen, W.; Lu, L.; Li, L.; Li, H.; Liu, M.; Zhou, C.; et al. Bone ECM-like 3D Printing Scaffold with
Liquid Crystalline and Viscoelastic Microenvironment for Bone Regeneration. ACS Nano
2022
,16, 21020–21035. [CrossRef]
[PubMed]
123.
Shen, X.; Zhang, Y.; Gu, Y.; Xu, Y.; Liu, Y.; Li, B.; Chen, L. Sequential and sustained release of SDF-1 and BMP-2 from silk
fibroin-nanohydroxyapatite scaffold for the enhancement of bone regeneration. Biomaterials 2016,106, 205–216. [CrossRef]
124.
Trubiani, O.; Marconi, G.D.; Pierdomenico, S.D.; Piattelli, A.; Diomede, F.; Pizzicannella, J. Human Oral Stem Cells, Biomaterials
and Extracellular Vesicles: A Promising Tool in Bone Tissue Repair. Int. J. Mol. Sci. 2019,20, 4987. [CrossRef]
125.
Jeong, J.H.; Hur, S.S.; Lobionda, S.; Chaycham, S.; Oh, J.S.; Lee, Y.K.; Hwang, Y. Heparin-mimicking polymer-based hydrogel
matrix regulates macrophage polarization by controlling cell adhesion. Biochem. Biophys. Res. Commun.
2023
,642, 154–161.
[CrossRef]
Biomedicines 2023,11, 2244 19 of 21
126.
Kojima, N.; Matsuo, T.; Sakai, Y. Rapid hepatic cell attachment onto biodegradable polymer surfaces without toxicity using an
avidin-biotin binding system. Biomaterials 2006,27, 4904–4910. [CrossRef]
127.
Kaur, M.; Sharma, S.; Sinha, V.R. Polymer based microspheres of aceclofenac as sustained release parenterals for prolonged
anti-inflammatory effect. Mater. Sci. Eng. C Mater. Biol. Appl. 2017,72, 492–500. [CrossRef]
128.
Tanetsugu, Y.; Tagami, T.; Terukina, T.; Ogawa, T.; Ohta, M.; Ozeki, T. Development of a Sustainable Release System for a
Ranibizumab Biosimilar Using Poly(lactic-co-glycolic acid) Biodegradable Polymer-Based Microparticles as a Platform. Biol.
Pharm. Bull. 2017,40, 145–150. [CrossRef]
129.
Arif, Z.U.; Khalid, M.Y.; Noroozi, R.; Sadeghianmaryan, A.; Jalalvand, M.; Hossain, M. Recent advances in 3D-printed polylactide
and polycaprolactone-based biomaterials for tissue engineering applications. Int. J. Biol. Macromol.
2022
,218, 930–968. [CrossRef]
[PubMed]
130.
Diaz-Gomez, L.; García-González, C.A.; Wang, J.; Yang, F.; Aznar-Cervantes, S.; Cenis, J.L.; Reyes, R.; Delgado, A.; Évora, C.;
Concheiro, A.; et al. Biodegradable PCL/fibroin/hydroxyapatite porous scaffolds prepared by supercritical foaming for bone
regeneration. Int. J. Pharm. 2017,527, 115–125. [CrossRef] [PubMed]
131.
Li, D.W.; He, J.; He, F.L.; Liu, Y.L.; Liu, Y.Y.; Ye, Y.J.; Deng, X.; Yin, D.C. Silk fibroin/chitosan thin film promotes osteogenic and
adipogenic differentiation of rat bone marrow-derived mesenchymal stem cells. J. Biomater. Appl.
2018
,32, 1164–1173. [CrossRef]
[PubMed]
132.
Liu, C.S.; Feng, B.W.; He, S.R.; Liu, Y.M.; Chen, L.; Chen, Y.L.; Yao, Z.Y.; Jian, M.Q. Preparation and evaluation of a silk fibroin-
polycaprolactone biodegradable biomimetic tracheal scaffold. J. Biomed. Mater. Res. B Appl. Biomater.
2022
,110, 1292–1305.
[CrossRef] [PubMed]
133.
Mazurek, Ł.; Szudzik, M.; Rybka, M.; Konop, M. Silk Fibroin Biomaterials and Their Beneficial Role in Skin Wound Healing.
Biomolecules 2022,12, 1852. [CrossRef] [PubMed]
134.
Pudkon, W.; Laomeephol, C.; Damrongsakkul, S.; Kanokpanont, S.; Ratanavaraporn, J. Comparative Study of Silk Fibroin-Based
Hydrogels and Their Potential as Material for 3-Dimensional (3D) Printing. Molecules 2021,26, 3887. [CrossRef]
135.
Hong, M.H.; Lee, J.H.; Jung, H.S.; Shin, H.; Shin, H. Biomineralization of bone tissue: Calcium phosphate-based inorganics in
collagen fibrillar organic matrices. Biomater. Res. 2022,26, 42. [CrossRef]
136.
Khurshid, Z.; Alfarhan, M.F.; Mazher, J.; Bayan, Y.; Cooper, P.R.; Dias, G.J.; Adanir, N.; Ratnayake, J. Extraction of Hydroxyapatite
from Camel Bone for Bone Tissue Engineering Application. Molecules 2022,27, 7946. [CrossRef]
137.
Pazarçeviren, A.E.; Tezcaner, A.; Keskin, D.; Kolukısa, S.T.; Sürdem, S.; Evis, Z. Boron-doped Biphasic
Hydroxyapatite/β-Tricalcium
Phosphate for Bone Tissue Engineering. Biol. Trace Elem. Res. 2021,199, 968–980. [CrossRef]
138.
Jing, T.; Yi, L.; Xu, L.; Chen, C.; Liu, F. The incorporation of
β
-tricalcium phosphate nanoparticles within silk fibroin composite
scaffolds for enhanced bone regeneration: An in vitro and in vivo study. J. Biomater. Appl. 2022,36, 1567–1578. [CrossRef]
139.
Jia, X.; Zhou, J.; Ning, J.; Li, M.; Yao, Y.; Wang, X.; Jian, Y.; Zhao, K. The polycaprolactone/silk fibroin/carbonate hydroxyapatite
electrospun scaffold promotes bone reconstruction by regulating the polarization of macrophages. Regen. Biomater.
2022
,
9, rbac035. [CrossRef]
140.
Hong, H.; Seo, Y.B.; Kim, D.Y.; Lee, J.S.; Lee, Y.J.; Lee, H.; Ajiteru, O.; Sultan, M.T.; Lee, O.J.; Kim, S.H.; et al. Digital light
processing 3D printed silk fibroin hydrogel for cartilage tissue engineering. Biomaterials 2020,232, 119679. [CrossRef]
141.
Bai, B.; Hou, M.; Hao, J.; Liu, Y.; Ji, G.; Zhou, G. Research progress in seed cells for cartilage tissue engineering. Regen. Med.
2022
,
17, 659–675. [CrossRef] [PubMed]
142.
Stefanik, J.J.; Guermazi, A.; Roemer, F.W.; Peat, G.; Niu, J.; Segal, N.A.; Lewis, C.E.; Nevitt, M.; Felson, D.T. Changes in
patellofemoral and tibiofemoral joint cartilage damage and bone marrow lesions over 7 years: The Multicenter Osteoarthritis
Study. Osteoarthr. Cartil. 2016,24, 1160–1166. [CrossRef]
143.
Kulchar, R.J.; Denzer, B.R.; Chavre, B.M.; Takegami, M.; Patterson, J. A Review of the Use of Microparticles for Cartilage Tissue
Engineering. Int. J. Mol. Sci. 2021,22, 10292. [CrossRef]
144.
Huang, K.; Li, Q.; Li, Y.; Yao, Z.; Luo, D.; Rao, P.; Xiao, J. Cartilage Tissue Regeneration: The Roles of Cells, Stimulating Factors
and Scaffolds. Curr. Stem. Cell Res. Ther. 2018,13, 547–567. [CrossRef] [PubMed]
145.
Rainbow, R.S.; Kwon, H.; Foote, A.T.; Preda, R.C.; Kaplan, D.L.; Zeng, L. Muscle cell-derived factors inhibit inflammatory
stimuli-induced damage in hMSC-derived chondrocytes. Osteoarthr. Cartil. 2013,21, 990–998. [CrossRef]
146.
Armiento, A.R.; Stoddart, M.J.; Alini, M.; Eglin, D. Biomaterials for articular cartilage tissue engineering: Learning from biology.
Acta Biomater. 2018,65, 1–20. [CrossRef] [PubMed]
147.
Liu, Z.; Zheng, Z.; Chen, K.; Li, Y.; Wang, X.; Li, G. A heparin-functionalized woven stent graft for endovascular exclusion.
Colloids Surf. B Biointerfaces 2019,180, 118–126. [CrossRef]
148.
Wang, T.; Li, Y.; Liu, J.; Fang, Y.; Guo, W.; Liu, Y.; Li, X.; Li, G.; Wang, X.; Zheng, Z.; et al. Intraarticularly injectable silk hydrogel
microspheres with enhanced mechanical and structural stability to attenuate osteoarthritis. Biomaterials
2022
,286, 121611.
[CrossRef]
149.
Wang, W.; Liu, Y.; Wang, S.; Fu, X.; Zhao, T.; Chen, X.; Shao, Z. Physically Cross-Linked Silk Fibroin-Based Tough Hydrogel
Electrolyte with Exceptional Water Retention and Freezing Tolerance. ACS Appl. Mater. Interfaces
2020
,12, 25353–25362. [CrossRef]
150.
Saha, S.; Kundu, B.; Kirkham, J.; Wood, D.; Kundu, S.C.; Yang, X.B. Osteochondral tissue engineering
in vivo
: A comparative
study using layered silk fibroin scaffolds from mulberry and nonmulberry silkworms. PLoS ONE 2013,8, e80004. [CrossRef]
Biomedicines 2023,11, 2244 20 of 21
151.
Shi, W.; Sun, M.; Hu, X.; Ren, B.; Cheng, J.; Li, C.; Duan, X.; Fu, X.; Zhang, J.; Chen, H.; et al. Structurally and Functionally
Optimized Silk-Fibroin-Gelatin Scaffold Using 3D Printing to Repair Cartilage Injury In Vitro and In Vivo. Adv. Mater.
2017
,
29, 1701089. [CrossRef]
152.
Wang, X.; Wenk, E.; Hu, X.; Castro, G.R.; Meinel, L.; Wang, X.; Li, C.; Merkle, H.; Kaplan, D.L. Silk coatings on PLGA and alginate
microspheres for protein delivery. Biomaterials 2007,28, 4161–4169. [CrossRef]
153.
Li, T.; Song, X.; Weng, C.; Wang, X.; Gu, L.; Gong, X.; Wei, Q.; Duan, X.; Yang, L.; Chen, C. Silk fibroin/carboxymethyl chitosan
hydrogel with tunable biomechanical properties has application potential as cartilage scaffold. Int. J. Biol. Macromol.
2019
,137,
382–391. [CrossRef] [PubMed]
154.
Liu, W.; Li, Z.; Zheng, L.; Zhang, X.; Liu, P.; Yang, T.; Han, B. Electrospun fibrous silk fibroin/poly(L-lactic acid) scaffold for
cartilage tissue engineering. Tissue Eng. Regen. Med. 2016,13, 516–526. [CrossRef] [PubMed]
155.
Naomi, R.; Ratanavaraporn, J.; Fauzi, M.B. Comprehensive Review of Hybrid Collagen and Silk Fibroin for Cutaneous Wound
Healing. Materials 2020,13, 3097. [CrossRef] [PubMed]
156.
Sharafat-Vaziri, A.; Khorasani, S.; Darzi, M.; Saffarian, Z.; Alizadeh, Z.; Tahmasebi, M.N.; Kazemnejad, S. Safety and efficacy of
engineered tissue composed of silk fibroin/collagen and autologous chondrocytes in two patients with cartilage defects: A pilot
clinical trial study. Knee 2020,27, 1300–1309. [CrossRef]
157.
Jaipaew, J.; Wangkulangkul, P.; Meesane, J.; Raungrut, P.; Puttawibul, P. Mimicked cartilage scaffolds of silk fibroin/hyaluronic
acid with stem cells for osteoarthritis surgery: Morphological, mechanical, and physical clues. Mater. Sci. Eng. C Mater. Biol. Appl.
2016,64, 173–182. [CrossRef]
158.
Wani, S.U.D.; Veerabhadrappa, G.H. Silk Fibroin Based Drug Delivery Applications: Promises and Challenges. Curr. Drug Targets
2018,19, 1177–1190. [CrossRef]
159.
Xu, J.; Wang, Y.; Ding, M.; Song, G.; Wu, M.; Kang, Z.; Wang, J. Sequence-structure characterization of recombinant polypeptides
derived from silk fibroin heavy chain. Mater. Sci. Eng. C Mater. Biol. Appl. 2020,111, 110831. [CrossRef]
160.
Bargel, H.; Trossmann, V.T.; Sommer, C.; Scheibel, T. Bioselectivity of silk protein-based materials and their bio-inspired
applications. Beilstein J. Nanotechnol. 2022,13, 902–921. [CrossRef]
161.
Ratanavaraporn, J.; Soontornvipart, K.; Shuangshoti, S.; Shuangshoti, S.; Damrongsakkul, S. Localized delivery of curcumin from
injectable gelatin/Thai silk fibroin microspheres for anti-inflammatory treatment of osteoarthritis in a rat model. Inflammopharma-
cology 2017,25, 211–221. [CrossRef]
162.
Seib, F.P.; Pritchard, E.M.; Kaplan, D.L. Self-assembling doxorubicin silk hydrogels for the focal treatment of primary breast
cancer. Adv. Funct. Mater. 2013,23, 58–65. [CrossRef]
163.
Qu, J.; Liu, Y.; Yu, Y.; Li, J.; Luo, J.; Li, M. Silk fibroin nanoparticles prepared by electrospray as controlled release carriers of
cisplatin. Mater. Sci. Eng. C Mater. Biol. Appl. 2014,44, 166–174. [CrossRef] [PubMed]
164.
Sharma, S.; Bano, S.; Ghosh, A.S.; Mandal, M.; Kim, H.W.; Dey, T.; Kundu, S.C. Silk fibroin nanoparticles support
in vitro
sustained
antibiotic release and osteogenesis on titanium surface. Nanomedicine 2016,12, 1193–1204. [CrossRef]
165.
Hassani Besheli, N.; Mottaghitalab, F.; Eslami, M.; Gholami, M.; Kundu, S.C.; Kaplan, D.L.; Farokhi, M. Sustainable Release of
Vancomycin from Silk Fibroin Nanoparticles for Treating Severe Bone Infection in Rat Tibia Osteomyelitis Model. ACS Appl.
Mater. Interfaces 2017,9, 5128–5138. [CrossRef]
166.
Wang, Z.; Yang, Z.; Jiang, J.; Shi, Z.; Mao, Y.; Qin, N.; Tao, T.H. Silk Microneedle Patch Capable of On-Demand Multidrug Delivery
to the Brain for Glioblastoma Treatment. Adv. Mater. 2022,34, e2106606. [CrossRef]
167.
Lehmann, T.; Vaughn, A.E.; Seal, S.; Liechty, K.W.; Zgheib, C. Silk Fibroin-Based Therapeutics for Impaired Wound Healing.
Pharmaceutics 2022,14, 651. [CrossRef] [PubMed]
168.
Di Buduo, C.A.; Laurent, P.A.; Zaninetti, C.; Lordier, L.; Soprano, P.M.; Ntai, A.; Barozzi, S.; La Spada, A.; Biunno, I.; Raslova, H.;
et al. Miniaturized 3D bone marrow tissue model to assess response to Thrombopoietin-receptor agonists in patients. Elife
2021
,
10, e58775. [CrossRef] [PubMed]
169.
Yang, C.; Li, S.; Huang, X.; Chen, X.; Shan, H.; Chen, X.; Tao, L.; Zhang, M. Silk Fibroin Hydrogels Could Be Therapeutic
Biomaterials for Neurological Diseases. Oxidative Med. Cell. Longev. 2022,2022, 2076680. [CrossRef]
170.
Gholipourmalekabadi, M.; Sapru, S.; Samadikuchaksaraei, A.; Reis, R.L.; Kaplan, D.L.; Kundu, S.C. Silk fibroin for skin injury
repair: Where do things stand? Adv. Drug Deliv. Rev. 2020,153, 28–53. [CrossRef]
171.
Crakes, K.R.; Herrera, C.; Morgan, J.L.; Olstad, K.; Hessell, A.J.; Ziprin, P.; LiWang, P.J.; Dandekar, S. Efficacy of silk fibroin
biomaterial vehicle for
in vivo
mucosal delivery of Griffithsin and protection against HIV and SHIV infection ex vivo. J. Int. AIDS
Soc. 2020,23, e25628. [CrossRef]
172.
Lam, Y.T.; Tan, R.P.; Michael, P.L.; Lau, K.; Yang, N.; Rnjak-Kovacina, J.; Wise, S.G. Bioengineering silk into blood vessels. Biochem.
Soc. Trans. 2021,49, 2271–2286. [CrossRef] [PubMed]
173.
He, S.; Fu, X.; Wang, L.; Xue, Y.; Zhou, L.; Qiao, S.; An, J.; Xia, T. Self-Assemble Silk Fibroin Microcapsules for Cartilage
Regeneration through Gene Delivery and Immune Regulation. Small 2023, e2302799. [CrossRef] [PubMed]
174.
Liu, J.; Lawrence, B.D.; Liu, A.; Schwab, I.R.; Oliveira, L.A.; Rosenblatt, M.I. Silk fibroin as a biomaterial substrate for corneal
epithelial cell sheet generation. Investig. Ophthalmol. Vis. Sci. 2012,53, 4130–4138. [CrossRef]
175.
Jeong, L.; Kim, M.H.; Jung, J.Y.; Min, B.M.; Park, W.H. Effect of silk fibroin nanofibers containing silver sulfadiazine on wound
healing. Int. J. Nanomed. 2014,9, 5277–5287.
Biomedicines 2023,11, 2244 21 of 21
176.
Nakayama, K.; Chinen, S.; Teshima, J.; Tamada, Y.; Hirabayashi, M.; Hochi, S. Silk fibroin sheet multilayer suitable for vitrification
of in vitro-matured bovine oocytes. Theriogenology 2020,145, 109–114. [CrossRef]
177.
Perrone, G.S.; Leisk, G.G.; Lo, T.J.; Moreau, J.E.; Haas, D.S.; Papenburg, B.J.; Golden, E.B.; Partlow, B.P.; Fox, S.E.; Ibrahim, A.M.;
et al. The use of silk-based devices for fracture fixation. Nat. Commun. 2014,5, 3385. [CrossRef]
178.
Yuan, T.; Li, Z.; Zhang, Y.; Shen, K.; Zhang, X.; Xie, R.; Liu, F.; Fan, W. Injectable Ultrasonication-Induced Silk Fibroin Hydrogel
for Cartilage Repair and Regeneration. Tissue Eng. Part A 2021,27, 1213–1224. [CrossRef]
179.
Chouhan, D.; Mandal, B.B. Silk biomaterials in wound healing and skin regeneration therapeutics: From bench to bedside. Acta
Biomater. 2020,103, 24–51. [CrossRef]
180.
Sultan, M.T.; Hong, H.; Lee, O.J.; Ajiteru, O.; Lee, Y.J.; Lee, J.S.; Lee, H.; Kim, S.H.; Park, C.H. Silk Fibroin-Based Biomaterials for
Hemostatic Applications. Biomolecules 2022,12, 660. [CrossRef]
181.
Huang, J.; Qin, J.; Zhang, P.; Chen, X.; You, X.; Zhang, F.; Zuo, B.; Yao, M. Facile preparation of a strong chitosan-silk biocomposite
film. Carbohydr. Polym. 2020,229, 115515. [CrossRef]
182.
Kundu, B.; Rajkhowa, R.; Kundu, S.C.; Wang, X. Silk fibroin biomaterials for tissue regenerations. Adv. Drug Deliv. Rev.
2013
,65,
457–470. [CrossRef] [PubMed]
183.
Ding, Z.; Fan, Z.; Huang, X.; Lu, Q.; Xu, W.; Kaplan, D.L. Silk-Hydroxyapatite Nanoscale Scaffolds with Programmable Growth
Factor Delivery for Bone Repair. ACS Appl. Mater. Interfaces 2016,8, 24463–24470. [CrossRef] [PubMed]
184.
Chen, S.; Chen, X.; Geng, Z.; Su, J. The horizon of bone organoid: A perspective on construction and application. Bioact. Mater.
2022,18, 15–25. [CrossRef]
185.
Warnecke, D.; Stein, S.; Haffner-Luntzer, M.; de Roy, L.; Skaer, N.; Walker, R.; Kessler, O.; Ignatius, A.; Dürselen, L. Biomechanical,
structural and biological characterisation of a new silk fibroin scaffold for meniscal repair. J. Mech. Behav. Biomed. Mater.
2018
,86,
314–324. [CrossRef] [PubMed]
186.
Rajput, M.; Mondal, P.; Yadav, P.; Chatterjee, K. Light-based 3D bioprinting of bone tissue scaffolds with tunable mechanical
properties and architecture from photocurable silk fibroin. Int. J. Biol. Macromol. 2022,202, 644–656. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Therefore, future work could find another cross-linking protocol is then required to generate high-strength hydrogels. It has been reported that SF and Gel can promote the secretion of GAG and collagen by chondrocytes (Chen et al., 2019;Wu et al., 2020;Lee et al., 2021;Yuan et al., 2021;Wu et al., 2022;Su et al., 2023;Zheng et al., 2023). Our study is consistent with previous findings that SF material and Gel material promote chondrocytes to produce more ECM deposition relative to the control group; the deposition of GAG and HYP in the SF group was higher than the control group. ...
Article
Full-text available
Suitable hydrogel materials for cartilage tissue repair should exhibit high strength and toughness, and excellent biocompatibility. However, the mechanical properties of most hydrogels cannot meet the complex mechanical requirements of articular cartilage tissues. Given this situation, we have adopted a chemical cross-linking method using hexafluoro isopropanol to mediate the cross-linking of Silk Fibroin (SF) and deionized water (DI), which promoted the formation of β-sheets, generating “high-toughness” Silk Fibroin hydrogels. The introduction of Gelatin (Gel) served to increase the content of β-sheets and increase the tensile modulus from 24.51 ± 2.07 MPa to 39.75 ± 6.54 MPa, which significantly enhanced the flexibility of the hydrogel and meets the mechanical requirements of cartilage tissue. In addition, in vitro biological experiments have shown that the introduction of Gel promotes cell proliferation and enhances the production of cartilage extracellular matrix by chondrocytes. In vivo experiments have demonstrated that SF/Gel hydrogel promotes articular cartilage regeneration more effectively than SF hydrogel, as evidenced by improvements in gross appearance, imaging, and histology. This study has established that high-strength SF/Gel hydrogel prepared by applying the binary-solvent-induced conformation transition strategy has potential applications in cartilage tissue repair and regeneration and is a feasible biomaterial for osteochondral regeneration.
... Fibroin has attracted intense research in recent decades, particularly in wound healing, due to the promising results obtained in vitro and in vivo by employing fibroin in different forms [28][29][30][31][32][33]. In addition to skin regeneration, until now, a variety of fibroin-based hydrogels, 3D porous scaffolds and mats have been fabricated for regenerating different tissues, including nerves, tendons, bone and cartilage, ligaments and blood vessels [34][35][36][37][38][39][40]. ...
Article
Full-text available
In order to reduce the toxicological impact on healthy cells and to improve the therapeutic response, many drug delivery systems have been fabricated and analysed, involving the use of different natural and synthetic materials at macro-, micro- and nanoscales. Among the natural materials which have demonstrated a huge potential for the development of effective drug delivery systems, silk fibroin has emerged for its excellent biological properties and for the possibility to be processed in a wide range of forms, which can be compliant with multiple active molecules and pharmaceutical ingredients for the treatment of various diseases. This review aims at presenting silk fibroin as an interesting biopolymer for applications in drug delivery systems, exploring the results obtained in recent works in terms of technological progress and effectiveness in vitro and in vivo.
Article
Full-text available
This review will present the latest research related to the production and application of spider silk and silk-based materials in reconstructive and regenerative medicine and tissue engineering, with a focus on musculoskeletal tissues, and including skin regeneration and tissue repair of bone and cartilage, ligaments, muscle tissue, peripheral nerves, and artificial blood vessels. Natural spider silk synthesis is reviewed, and the further recombinant production of spider silk proteins. Research insights into possible spider silk structures, like fibers (1D), coatings (2D), and 3D constructs, including porous structures, hydrogels, and organ-on-chip designs, have been reviewed considering a design of bioactive materials for smart medical implants and drug delivery systems. Silk is one of the toughest natural materials, with high strain at failure and mechanical strength. Novel biomaterials with silk fibroin can mimic the tissue structure and promote regeneration and new tissue growth. Silk proteins are important in designing tissue-on-chip or organ-on-chip technologies and micro devices for the precise engineering of artificial tissues and organs, disease modeling, and the further selection of adequate medical treatments. Recent research indicates that silk (films, hydrogels, capsules, or liposomes coated with silk proteins) has the potential to provide controlled drug release at the target destination. However, even with clear advantages, there are still challenges that need further research, including clinical trials.
Article
Full-text available
Effective treatments for cartilage defects are currently lacking. Gene delivery using proper delivery systems has shown great potential in cartilage regeneration. However, the inflammatory microenvironment generated by the defected cartilage severely affects the system's delivery efficiency. Therefore, this study reports a silk fibroin microcapsule (SFM) structure based on layer‐by‐layer self‐assembly, in which interleukin‐4 (IL‐4) is modified on silk by click chemistry and loaded with lysyl oxidase plasmid DNA (LOX pDNA). The silk microcapsules display good biocompatibility and the release rate of genes can be adjusted by controlling the number of self‐assembled layers. Moreover, the functionalized SFMs mixed with methacrylated gelatin (GelMA) exhibit good injectability. The IL‐4 on the outer layer of the SFM can regulate macrophages to polarize toward the M2 type, thereby promoting cartilage matrix repair and inhibiting inflammation. The LOX pDNA loaded inside can be effectively delivered into cells to promote extracellular matrix generation, significantly promoting cartilage regeneration. The results of this study provide a promising biomaterial for cartilage repair, and this novel silk‐based microcapsule delivery system can also provide strategies for the treatment of other diseases.
Article
Full-text available
Silk fibroin (SF) and sericin (SS), the two major proteins of silk, are attractive biomaterials with great potential in tissue engineering and regenerative medicine. However, their biochemical interactions with stem cells remain unclear. In this study, we employed multiomics to obtain a global view of the cellular processes and pathways of mesenchymal stem cells (MSCs) triggered by SF and SS to discern cell-biomaterial interactions at an in-depth, high-throughput molecular level. Integrated RNA sequencing and proteomic analysis confirmed that SF and SS initiated widespread but distinct cellular responses and potentiated the paracrine functions of MSCs that regulate extracellular matrix deposition, angiogenesis, and immunomodulation through differentially activating the integrin/PI3K/Akt and glycolysis signaling pathways. These paracrine signals of MSCs stimulated by SF and SS effectively improved skin regeneration by regulating the behavior of multiple resident cells (fibroblasts, endothelial cells, and macrophages) in the skin wound microenvironment. Compared to SS, SF exhibited better immunomodulatory effects in vitro and in vivo, indicating its greater potential as a carrier material of MSCs for skin regeneration. This study provides comprehensive and reliable insights into the cellular interactions with SF and SS, enabling the future development of silk-based therapeutics for tissue engineering and stem cell therapy. This article is protected by copyright. All rights reserved.
Article
Full-text available
Bone tissue engineering (BTE) utilizes a special mix of scaffolds, cells, and bioactive factors to regulate the microenvironment of bone regeneration and form a three-dimensional bone simulation structure to regenerate bone tissue. Silk fibroin (SF) is perhaps the most encouraging material for BTE given its tunable mechanical properties, controllable biodegradability, and excellent biocompatibility. Numerous studies have confirmed the significance of SF for stimulating bone formation. In this review, we start by introducing the structure and characteristics of SF. After that, the immunological mechanism of SF for osteogenesis is summarized, and various forms of SF biomaterials and the latest development prospects of SF in BTE are emphatically introduced. Biomaterials based on SF have great potential in bone tissue engineering, and this review will serve as a resource for future design and research.
Article
Full-text available
The skin, acting as the outer protection of the human body, is most vulnerable to injury. Wound healing can often be impaired, leading to chronic, hard-to-heal wounds. For this reason, searching for the most effective dressings that can significantly enhance the wound healing process is necessary. In this regard, silk fibroin, a protein derived from silk fibres that has excellent properties, is noteworthy. Silk fibroin is highly biocompatible and biodegradable. It can easily make various dressings, which can be loaded with additional substances to improve healing. Dressings based on silk fibroin have anti-inflammatory, pro-angiogenic properties and significantly accelerate skin wound healing, even compared to commercially available wound dressings. Animal studies confirm the beneficial influence of silk fibroin in wound healing. Clinical research focusing on fibroin dressings is also promising. These properties make silk fibroin a remarkable natural material for creating innovative, simple, and effective dressings for skin wound healing. In this review, we summarise the application of silk fibroin biomaterials as wound dressings in full-thickness, burn, and diabetic wounds in preclinical and clinical settings.
Article
Full-text available
Waste tissues such as mammalian bone are a valuable source from which to extract hydroxyapatite. Camel bone-based hydroxyapatite (CBHA) was extracted from the femur of camel bones using a defatting and deproteinization procedure. The extracted CBHA was mechanically, chemically, physically, morphologically and structurally characterized. Fourier-Transform InfraRed (FTIR) spectra, Micro-Raman, and X-ray diffraction analysis confirmed successful extraction of hydroxyapatite. The mechanical properties of the CBHA scaffold were measured using a Universal Instron compression tester. Scanning electron microscopy showed the presence of a characteristic interconnected porous architecture with pore diameter ranging from 50–600 µm and micro-computer tomography (Micro-CT) analysis identified a mean porosity of 73.93. Thermogravimetric analysis showed that the CBHA was stable up to 1000 °C and lost only 1.435% of its weight. Inductively coupled plasma–mass spectrometry (ICP-MS) and Energy-dispersive-X-ray (EDX) analysis demonstrated the presence of significant amounts of calcium and phosphorus and trace ions of sodium, magnesium, zinc, lead and strontium. Following 21 days of incubation in simulated body fluid (SBF), the pH fluctuated between 10–10.45 and a gradual increase in weight loss was observed. In conclusion, the extracted CBHA is a promising material for future use in bone tissue regeneration applications.
Article
Implant-supported dental prosthetics are widely used in dental practice. Sufficient peri-implant bone tissue is a crucial prerequisite for the long-term success of this treatment, as insufficient peri-implant bone volume hampers dental implant installation and negatively influences dental implant stability. However, due to tooth extraction, bone metabolism diseases, and trauma, bone defects in the jaw are common in patients, particularly in the elderly and those suffering from underlying conditions. If this is the case, alveolar ridge has to be augmented for reliable implant placement. Various biomaterials, growth factors or growth factor-based products, and trace elements have been tested and used for alveolar ridge augmentation. Among those biomaterials, calcium phosphates (CaPs) are the most popular due to their promising biocompatibility, great osteoconductivity, and distinguishing osteogenesis. Combining CaPs with growth factors or trace elements can further favor bone defect repair. This review mainly focuses on applying artificial CaP biomaterials and their combination with bioactive agents to repair bone defects in implant dentistry.
Article
In this study, we developed a poly(β-amino ester) (PBAE) hydrogel for the double release of vancomycin (VAN) and total flavonoids of Rhizoma Drynariae (TFRD). VAN was covalently bonded to PBAE polymer chains and was released to enhance the antimicrobial effect first. TFRD chitosan (CS) microspheres were physically dispersed in the scaffold, TFRD was released from the microspheres, and osteogenesis was induced subsequently. The scaffold had good porosity (90.12 ± 3.27%), and the cumulative release rate of the two drugs in PBS (pH 7.4) solution exceeded 80%. In vitro antimicrobial assays demonstrated the antibacterial properties of the scaffold against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). Besides these, cell viability assays indicated that the scaffold had good biocompatibility. Moreover, alkaline phosphatase and matrix mineralization were expressed more than in the control group. Overall, cell experiments confirmed that the scaffolds have enhanced osteogenic differentiation capabilities. In conclusion, the dual-drug-loaded scaffold with antibacterial and bone regeneration effects is promising in the field of bone repair.
Article
Microfibers, a type of long, thin, and flexible material, can be assembled into functional three-dimensional (3D) structures by folding, binding and weaving. As a novel spinning method, combining microfluidic technology and wet spinning, microfluidic spinning technology can precisely control the size, morphology, structure, and composition of the microfibers. Particularly, the process is mild and rapid, which is suitable for preparing microfibers using biocompatible materials and, without affecting the viability of cells encapsulated. Furthermore, owing to the controllability of microfluidic spinning, microfibers with well-defined structures (such as hollow structures) will contribute to the exchange of nutrients or guide cell orientation. Thus, this method is often used to fabricate microfibers as cell scaffolds for cell encapsulation or adhesion, and can be further applied to biomimetic fibrous tissues. In this review, we focus on different fiber structures prepared by microfluidic spinning technology, including solid, hollow, and heterogeneous structures, generated from three essential elements: spinning platform, fiber composition, and solidification methods. Furthermore, we describe the application of microfibers with different structures in tissue engineering, such as blood vessels, skeletal muscle, bone, nerves, and lung bronchi. Finally, the challenges and future development prospects of microfluidic spinning technology in tissue engineering applications are discussed. This article is protected by copyright. All rights reserved
Article
The physicochemical properties of biomaterials influence cell adhesion, shape, and polarization of macrophages. In this study, we aimed to evaluate the polarization of macrophages in terms of the regulation of cell adhesion and how synthetic mimics for heparin and poly(sodium-4-styrenesulfonate) can regulate macrophage polarization by modulating cell shape, focal adhesion, cell traction force, and intracellular tension. Our initial findings showed that macrophages cultured with heparin-mimicking polymer-based hydrogel matrix showed reduced expression of cell adhesion markers such as integrins, vinculin, RhoA, and ROCK1/2 and reduced cell shape, elongation, cell-matrix traction force, and intracellular tension. Furthermore, we observed a significant decrease in cell adhesion in cells cultured on the hydrogel, resulting in the promotion of M1 polarization. These findings offer insights into the important roles of cell-matrix interactions in macrophage polarization and offer a platform for heparin-mimicking polymer-based hydrogel matrices to induce M1 polarization by inducing cell adhesion without classical activators.
Article
Implanting a 3D printing scaffold is an effective therapeutic strategy for personalized bone repair. As the key factor for the success of bone tissue engineering, the scaffold should provide an appropriate bone regeneration microenvironment and excellent mechanical properties. In fact, the most ideal osteogenic microenvironment is undoubtedly provided by natural bone extracellular matrix (ECM), which exhibits liquid crystalline and viscoelastic characteristics. However, mimicking a bone ECM-like microenvironment in a 3D structure with outstanding mechanical properties is a huge challenge. Herein, we develop a facile approach to fabricate a bionic scaffold perfectly combining bone ECM-like microenvironment and robust mechanical properties. Creatively, 3D printing a poly(l-lactide) (PLLA) scaffold was effectively strengthened via layer-by-layer electrostatic self-assembly of chitin whiskers. More importantly, a kind of chitin whisker/chitosan composite hydrogel with bone ECM-like liquid crystalline state and viscoelasticity was infused into the robust PLLA scaffold to build the bone ECM-like microenvironment in 3D structure, thus highly promoting bone regeneration. Moreover, deferoxamine, an angiogenic factor, was encapsulated in the composite hydrogel and sustainably released, playing a long-term role in angiogenesis and thereby further promoting osteogenesis. This scaffold with bone ECM-like microenvironment and excellent mechanical properties can be considered as an effective implantation for bone repair.