You are on page 1of 19

Biomaterials 287 (2022) 121638

Contents lists available at ScienceDirect

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

Silk sericin-based materials for biomedical applications


Jia Liu a, 1, Lin Shi a, 1, Yan Deng a, b, 1, Meizhen Zou a, b, Bo Cai a, Yu Song a, Zheng Wang a, c, **,
Lin Wang a, b, *
a
Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
430022, China
b
Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
c
Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China

A R T I C L E I N F O A B S T R A C T

Keywords: Silk sericin, a natural protein extracted from silkworm cocoons, has been extensively studied and utilized in the
Silk sericin biomedical field because of its superior biological activities and controllable chemical-physical properties.
Biomaterials Sericin is biocompatible and naturally cell adhesive, enabling cell attachment, proliferation, and differentiation
Tissue engineering
in sericin-based materials. Moreover, its abundant functional groups from variable amino acids composition
Drug delivery
allow sericin to be chemically modified and cross-linked to form versatile constructs serving as alternative
matrixes for biomedical applications. Recently, sericin has been constructed into various types of biomaterials for
tissue engineering and regenerative medicine, including various bulk constructions (films, hydrogels, scaffolds,
conduits, and devices) and micro-nano formulations. In this review, we systemically summarize the properties of
silk sericin, introduce its different forms, and demonstrate their newly-developed as well as potential biomedical
applications.

1. Introduction silk sericin has been ignored and discarded as a waste from the textile
industry for thousands of years, causing environmental pollution and a
Silk is a type of ancient material that has emerging values in waste of natural resources [9]. However, with the advances in sericin
biomedical applications. As a natural protein-polymer secreted in the investigation, the discarded sericin is revealed to possess excellent bio­
glands of lepidopteran insects, the resources of silk are abundant with a logical activities as well as fibroin. Other than good-biocompatibility
short production cycle [1]. In countries like China and India, the history and low-immunogenicity that are very essential for biomaterials,
of utilizing silk can be dated back thousands of years ago, mostly in recently, it is demonstrated that sericin can support cell attachment and
textile industries. Since the initial applications such as silk-based sutures proliferation [10], and stimulate cell differentiation [11–13]. Further­
in medical practice, and excellent biological properties of silk are more, sericin has excellent humidity preservation and antioxidant ac­
gradually revealed, silk has been considered a promising natural mate­ tivities [14,15]. Through modification, precipitation, or cross-linking
rial for biomedical applications, especially in tissue engineering, with other polymers, sericin could be prepared as stable and controllable
drug/gene delivery, and the emerging smart wearable electronics [1–3]. biomaterials, whose morphology, size, and properties (elasticity,
Silk mainly contains two components: fibroin and sericin. Silk fibroin strength, and biodegradability) are able to be tuned precisely. Therefore,
has not only been developed as biomedical devices (surgical sutures) it has attracted attention in the fields of cosmeceuticals, cell culture, and
[4], but also becomes an attractive candidate for tissue engineering and tissue engineering [16,17]. The recovery and utilization of sericin would
drug delivery system due to its suitable elasticity, mechanical strength, not only minimize environmental problems but also achieve great po­
controllable degradation, and biocompatibility [5–8]. On the contrary, tential value in biomedical research and medical practice.

* Corresponding author. Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of
Science and Technology, Wuhan, 430022, China.
** Corresponding author. Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of
Science and Technology, Wuhan, 430022, China.
E-mail addresses: zhengwang@hust.edu.cn (Z. Wang), lin_wang@hust.edu.cn (L. Wang).
1
, these authors contributed equally to this work.

https://doi.org/10.1016/j.biomaterials.2022.121638
Received 21 January 2022; Received in revised form 4 June 2022; Accepted 14 June 2022
Available online 17 June 2022
0142-9612/© 2022 Published by Elsevier Ltd.
J. Liu et al. Biomaterials 287 (2022) 121638

(hydrogels [18,19], films [20,21], conduits [22–25], scaffolds [26–28]),


and micro-nano structures [29] (Fig. 1). It is worth mentioning that the
unique amino acid composition of sericin contributes to its amphipathic
and acid-base responsiveness, making it a good candidate for drug de­
livery [30]. This review outlines the structure and properties of silk
sericin, summarizes and discusses different types of sericin-based plat­
forms and their applications in tissue engineering and drug delivery, and
offers prospects for future applications.

2. Characteristics and processing methods of sericin

2.1. Sources and types of silkworm cocoons

Silks are generally spun into fibers in the glands of lepidopteran in­
sects, including silkworms, spiders, flies, mites, and scorpions [31]. The
most commonly-used silks originate from silkworms of Bombycidae and
Saturniidae families. According to whether they eat mulberry leaves,
silkworms are divided into mulberry and non-mulberry categories [32].
Silk from Bombycidae family belongs to mulberry silks, such as Bombyx
mori (B. mori), while temperate and tropical tasar silks from Saturniidae
family belong to the non-mulberry silk categories, such as Antheraea
assamensis (A. assamensis), Antheraea pernyi (A. pernyi), and Antheraea
mylitta (A. mylitta) [33,34]. The cocoon layer is mainly composed of silk
fibroin (60%–80%) in the center and sericin (15%–35%) in the periph­
ery, whose proportions are dependent on the species of silkworms [35].
Fig. 1. The biological activities of sericin, different formats of sericin-based With the extensive exploration of sericin and the help of bioengineering,
biomaterials, and their biological applications. new types of silkworm cocoons containing more than 40% or even up to
90% of sericin (from silk fibroin-deficient cocoons) have been developed
At present, sericin-based biomaterials have been integrated into [35].
tissue engineering constructs to meet various repair and regeneration
demands for different types of tissues, including bulk materials

Fig. 2. Schematic diagram of sericin extraction methods. For wild-type cocoons, there are many different extraction methods for sericin isolation, including physical
extraction methods, biological extraction methods, and chemical extraction methods. The sericin is purified by dialysis and obtained by freeze-drying. For silk
fibroin-deficient cocoons, the intact sericin could be isolated through dissolution in LiBr solution (6 M), dialysis in water, and concentration in PEG solution,
successively.

2
J. Liu et al. Biomaterials 287 (2022) 121638

2.2. Extraction and properties of sericin Table 1


Different sericin extraction methods.
2.2.1. Structure and composition Methods Advantages Limitations Reference
Approximately 90% of the components in sericin are proteins (20 (s)
kDa–400 kDa, 18 amino acids). The remaining components include Thermal-dependent methods
pigments, waxes, sugars, mineral salts, and other impurities [36]. Unlike HTHP (autoclave) No purification Low yield; Heat- [10,49,52,
fibroin, sericin is water-soluble due to its high proportion of polar amino process required; caused degradation; 54,56,57]
acids (>50%), including serine, aspartic acid, and lysine [35,37], which Low cost; Low Destruction of β-sheet
toxicity; Suitable for structures;
allow physical/chemical modifications and crosslinking. The sericin safety evaluation; Abrogating BMP-2/4
protein wrapped around fibroin has a stratified structure, which is High collagen induction
divided into inner, middle, and outer layers according to the water production; High
solubility. From the inner to outer layers (far away from fibroin), the antioxidant activity
and tyrosinase
amount of hydrophobic amino acids in sericin gradually decreases,
inhibition activity
while the content of hydrophilic amino acids gradually increases. Infrared heating High yield and Extra equipment [44]
Therefore, the outer layer has the best water solubility and can be dis­ quality of silk sericin
solved in boiling water within 2 h [37]. In addition, there are two aro­ (SS)
matic amino acids in sericin (tyrosine and phenylalanine) [35], which Chemical reagent-dependent method
Alkaline solution: Application on a Mechanical damage [10,48,
may endow sericin with intrinsic fluorescence [38]. This photo­ Na2CO3, NaOH large scale; Low cost; of silk fibroin (SF); 52]
luminescence property inherits after chemical modification or physical High yield; High Complex purification
crosslinking [39], making sericin a promising biomaterial for biological antioxidant activity process; Large
detection and tracing in vivo. amounts of
wastewater;
The secondary structure of sericin mainly contains random coils and
Extensive
β-sheets, corresponding to the amorphous and crystalline regions, degradation;
respectively [16]. Due to its high content of random coils, sericin ex­ Unbeneficial in
hibits the characteristics of amorphous materials in the dry state, such as fabricating self-
fragility and low mechanical strength. The strength and crystallinity of standing matrices and
self-assembly
sericin-based materials can be controlled by temperature regulation and
particles due to high
chemical modification. Generally, the high temperature tends to destroy water solubility; loss
the β-sheets in sericin [40]. On the contrary, chemical crosslinking (e.g., antityrosinase
glutaraldehyde) leads to stable β-sheet structure formation, thus activity; Massive
reduction of β-sheet
improving the crystallinity and strength of sericin-based materials [41].
Neutral soaps (NS) High quality of High cost; Time- [43,58,
In addition, ethanol-induced dehydration is conducive to the formation fibroin fiber consuming; 59]
of β-sheets from random coils in sericin [42]. Environmental
pollution; Difficulty
2.2.2. Extraction methods in sericin recycling
and reuse
In the textile industry, silkworm cocoons are usually boiled in a so­
Acidic solution: Lower degree of Mechanical damage [10,45,
lution containing sodium carbonate and Marseille soap to remove citric acid, degradation than of SF; Complex 46]
sericin for isolating fibroin. This industrial degumming process is a tartaric acid alkali degumming purification process
traditional and effective approach to separate sericin and fibroin, which Urea buffer Low degree of Inefficient; Extremely [10,52,
degradation; High high cytotoxicity; 60]
however suffers from the high cost of Marseille soap and difficulties in
anti-tyrosinase Massive reduction of
sericin recycling [43]. To cope with these issues, the high-temperature activity for inhibition β-sheet; Abrogating
high-pressure method (HTHP) is employed to dissolve sericin of co­ of melanogenesis BMP-2/4 induction
coons in boiled water under ambient or high pressure, avoiding complex LiBr, LiSCN, NaCl Low degree of Inefficient; Complex [38,61,
refining and purification processes [10]. However, this approach usually degradation; Suitable purification process; 62]
for structural studies Short storage period
results in a low yield, largely because only the outer layer of sericin is
and fabricating pure
dissolved. To enhance sericin dissolution, an infrared (IR) dyeing ma­ sericin based bulk
chine is utilized to heat silkworm cocoons [44]. In this process, water materials
molecules act as abrasives during energy transfer by electromagnetic Surfactant: silk Similar degumming Extra synthesis [58,59]
protein rate to the NS; process of surfactant;
waves, thereby enhancing the sericin shedding. In combination with
surfactant (SPS), Environment Potential biosecurity
heating, some chemical reagents are employed to improve the degum­ alkyl friendly concerns
ming efficiency, including alkaline reagents (sodium carbonate and so­ polyglycoside
dium hydroxide) [34], acid reagents (citric acid and tartaric acid) [45, (APG)
46], and proteases (papain, bromelain, pancreatin, and fungal protease) Biological enzyme-dependent method
Papain, bromelain, Suitable for Low and narrow MW [47,
[47] (Fig. 2). Among them, boiling silkworm cocoons in sodium car­
pancreatin, industrial range of SS peptides; 63–65]
bonate solution (0.02 M) is the most widely used laboratory degumming fungal protease production;
method with high extraction efficiency for both mulberry silkworm completely remove
cocoons (B. mori) and non-mulberry silkworm cocoons (A. mylitta) [48]. SS;
It is important to note that extraction conditions (temperature, time, Homogeneous low Tedious degumming
molecular weight procedure
ratio of cocoons to water, and chemical reagents) not only determine the peptides for cell
yield but also affect the molecular weights and characteristics of isolated culture supplements
sericin. Higher temperature, longer duration, and alkaline reagents may
increase the yield, but they lead to denaturation and degradation of
sericin [49]. 140 Nd-s) allows facile isolation of intact sericin under mild conditions.
To obtain intact protein, sericin is directly isolated from the middle Our group extracted sericin protein from the fibroin-deficient silkworm
silk gland of fifth instar larvae [50,51]. In addition, the emergence of cocoons using lithium bromide (LiBr) solution (6 M) at 35 ◦ C, which
natural fibroin-deficient mutant silkworms (e.g., B. mori, 185 Nd-s, and exhibited a well-preserved protein profile at 60 kDa, 100 kDa, 130 kDa,

3
J. Liu et al. Biomaterials 287 (2022) 121638

and >250 kDa. This gentle LiBr extraction method not only preserves the biomedical applications.
protein structures but also provides a high yield of sericin (>90%) [38].
The sericin proteins obtained by different extraction methods have 2.2.3.2. Ability to support cell growth. Sericin is demonstrated to pro­
different amino acid profiles, conformation, and molecular weights, as mote the proliferation and maintain the functions of various types of
well as different physical-chemical properties and biological activities cells, including normal mammalian, tumor, and insect cells [78,79].
[52]. As a foreign protein, sericin would be recognized by the pattern This characteristic may be attributed to the following aspects: (1) Sericin
recognition receptors of immune cells, which are affected by the protein can substitute bovine serum albumin (BSA) or fetal bovine serum (FBS)
conformation of sericin, especially β-sheet structures [53]. Since the to support cell culture as a nutrient. For example, serum-free DMEM
conformation of sericin is highly dependent on the extraction methods, medium supplemented with sericin (0.05%) can improve the anti-aging
the receptors activated by sericin and its bioactivities would be varied by ability of skin fibroblasts [80], and RPMI1640 medium containing
the degumming conditions. For instance, in the sericin obtained by sericin (0.01%) could maintain the secretory function of pancreatic islet
sonication at 37 ◦ C, the high molecular weight (HMW) fraction of pro­ cells to exert hypoglycemic ability [81]. (2) Sericin is capable of sup­
tein (MW > 30 kDa) possesses more abundance of β-sheet structures porting cell adhesion. The sericin-based film was used to culture mouse
compared with the low molecular weight fraction [54]. Of note, the fibroblasts (L929), which supported cell attachment and proliferation as
sericin protein with the high amount of β-sheet could significantly in­ well as styrene culture plate [10]. (3) Sericin can resist cell apoptosis
crease the bone morphogenic protein-2/4 (BMP-2/4) expression in induced by ultraviolet irradiation via inhibiting caspase-3 activation
macrophages and monocytes via activation of Toll-like receptors [33]. (4) Sericin could also promote cell differentiation. Recently,
(TLR-2/4), which recognize the foreign materials in the body. Thus, such sericin was demonstrated to induce rapid differentiation and maturation
HMW sericin protein has great osteogenic activity. However, the sericin of primary human retinal pigment epithelial cells (hRPEs) through
isolated by harsh extraction methods (e.g., heat or urea) mainly contains NF-κB pathway. The visual cycle-related genes (RPE65, RDH10, and
random coils, which hardly enhance the BMP-2/4 expression. Of note, CRALBP) were significantly upregulated after sericin treatment in hRPEs
the heating treatment could even denature the HMW sericin and [12,13]. Moreover, sericin modification on the surface of commercially
diminish its BMP-2/4 induction ability, likely due to the destruction of titanium alloy promoted the adhesion, proliferation, and differentiation
β-sheet structures. Moreover, the degradation of peptide sequences of osteoblasts, and improved the activity of alkaline phosphatase,
during the degumming process would also weaken the bioactivities of thereby promoting bone tissue repair [11].
sericin. For example, the sericin extracted by heat method exhibited These forementioned benefits of sericin on cell culture make it
weaker activities on lactate suppression and interferon-β induction in suitable for tissue engineering, and sericin-based biomaterials have been
vesicular stomatitis virus (VSV) infected RD cells compared with the widely investigated and utilized in bone tissue engineering [82], adipose
intact sericin isolated by LiBr method, likely due to partial degradation tissue engineering [83,84], nerve injury repair [22,25], skin injury
of F5-SP peptide sequence (SEDSSEVDIDLGNLG, an α-glucosidase in­ repair [85], as well as corneal epithelial repair [86].
hibitor) in obtained sericin [55]. Therefore, it is necessary to choose the
appropriate methods for sericin isolation according to the actual appli­ 2.2.3.3. Antioxidant activity. The antioxidant properties of sericin are
cation requirements and silkworm cocoons (Table 1). closely related to its reactive oxygen species (ROS) scavenging activity,
inhibition of lipid peroxidation, anti-tyrosinase, and anti-elastase ac­
2.2.3. Biological activities tivities [14,15]. Moreover, sericin may enhance the activity of antioxi­
dant enzymes such as superoxide dismutase (SOD), catalase (CAT), and
2.2.3.1. Biocompatibility and immunogenicity. Biocompatibility is one of glutathione peroxidase (GPx) [87,88]. The antioxidant activities are also
the most important considerations for materials utilized for biomedical related to its high contents of serine and threonine, which provide hy­
applications [66,67]. Due to limited understanding and studies, sericin droxyl groups to chelate ions (e.g., copper and iron) [15]. In addition,
was once considered the major cause of adverse effects in silk applica­ the pigment molecules (e.g., flavonoids and carotenoids) accumulated in
tions, such as inflammation, allergenicity, and immunogenicity [68]. In sericin layers may be one of the causes that endow sericin with antiox­
a clinical study, exposure to silk was recognized as an independent idant properties and anti-tyrosinase activity [52,89]. Of note, the anti­
predictor of asthma [69]. The biosafety concerns of sericin have slowed oxidant properties of sericin are partly determined by the cocoon strains
down and impeded its biomedical applications for decades. and affected by the extraction methods [90–92].
Recently, extensive evidence has demonstrated that sericin itself Oxidative stress damage occurs widely in various polar physical
does not cause a significant immune response, while the mixture of environments and pathological conditions [93]. Due to its antioxidation,
sericin and fibroin proteins may induce chronic immune responses with sericin has been used in a variety of fields related to oxidative stress. It is
unclear mechanisms [70–73]. The studies validated that sericin and reported that sericin could effectively absorb ultraviolet (UV), prevent
sericin-based membranes mildly increased the levels of tumor necrosis oxidative damage, and reduce apoptosis of human keratinocytes
factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) of murine macro­ (HaCaT) [94,95]. In addition, the sericin (B. mori) enhanced the resis­
phages (RAW264.7) in vitro, which were comparable to the polystyrene tance of human epidermal cells (A431) against chlorpromazine-induced
plates [51,74,75]. Moreover, in both rabbits and mice, sericin exhibited phototoxicity via increasing intracellular glutathione [95]. These
immunological inertia [76,77]. To clarify the biosafety of sericin, we studies indicate the antioxidant and photoprotective effects of sericin.
have extracted the degraded and intact sericin from wild-type and Besides UV irradiation, sericin has a protective effect against
fibroin-deficient mutant silkworm cocoons (B. mori), respectively, and oxidative stress damage caused by ultra-low temperatures [96,97].
evaluated their biocompatibility in BALB/c mice [77]. To investigate the Cryopreservation is a key technology for breeding and passage, how­
inflammatory responses, the intact sericin-based hydrogel was subcu­ ever, ultra-low temperature freezing induces strong oxidative stress for
taneously inoculated in BALB/c mice, which did not increase the infil­ cells, tissues, and organs, which reduces their survival rate and biolog­
tration of macrophages or neutrophils in the tissue of hydrogel ical function. Given the benefits of sericin in cell culture and its anti­
implantation site compared with alginate hydrogel and fibroin hydrogel oxidant capacity, sericin-based cryoprotectants have been developed
at Day 3, 10 and 17. Moreover, these sericin proteins did not signifi­ and applied to protect mammalian cells, buffalo semen, and even human
cantly elevate the levels of IgG (total IgG and sericin-specific IgG) and sperm in cryopreservation [98–100]. The sericin addition (0.25%–0.5%)
IgE compared with fibrinogen (an FDA-approved biomaterial), indi­ not only reduced the malondialdehyde in seminal plasma but also
cating the low immunogenicity and allergenicity of sericin. These find­ increased the activities of GPx and SOD, therefore improving the
ings indicate that sericin is a biocompatible natural protein suitable for integrity of sperm plasma membrane and survival rate of sperm in

4
J. Liu et al. Biomaterials 287 (2022) 121638

Table 2
Biomedical applications of sericin-based bulk materials.
Materials Applications Research stage Types of silk Reference
(s)

Films
Sericin Skin tissue engineering In vitro A. mylitta [42]
Sericin Bone tissue engineering In vitro A. pernyi [103]
Sericin/Collagen Wound dressing in vivo, Wistar rats B. mori [104]
Sericin/Bacterial Cellulose Wound healing In vitro B. mori [105]
Sericin/Hydroxyapatite Bone tissue engineering In vitro B. mori [106]
Sericin/Ti-RGD Osseointegration In vitro A. mylitta [11]
Sericin/Agarose-Glycerol Chronic wound treatment In vitro Bivoltine sericin [20]
AgNPs-Sericin/PVA Antibacterial dressing In vitro Sericin powder [107]
AgNPs-Sericin/Agarose Antibacterial dressing In vitro 872. Silkworms [108]
AgNPs-PDA-Sericin/PVA Antibacterial dressing In vitro Unclear [109]
AgNPs-PDA-Sericin/Agarose Wound dressing In vitro 872. Silkworms [110]
ZnONPs-Sericin/PVA Antibacterial dressing In vitro Sericin powder [21]
Ag@ZnONPs-Sericin/Agarose Antibacterial dressing In vitro Unclear [111]
Silver Zinc Sulfadiazine/Sericin Cream Second-degree burn wounds Clinical Trial B. mori [112]
Sericin Cream Pruritus in hemodialysis patients Clinical Trial B. mori [113]
Sericin/Chitosan Cream Pressure Sore Clinical Trial, Recruiting B. mori [133]
Silk Mat Guided bone regeneration (GBR) Clinical Trial B. mori [114]
Hydrogels
Sericin Ischemic Stroke In vitro B. mori, 140Nd-s [18]
Sericin Cartilage regeneration In vitro B. mori, 140Nd-s [19]
Sericin Cell culture and drug delivery In vitro 140 Nd-s [38]
Sericin Ischemic myocardial infarction; In vitro B. mori, 140 Nd- [115]
s
Sericin/Agarose Wound dressing In vitro B. mori [116]
Sericin/Chitosan Skin tissue regeneration in vivo, C57BL/6 mice A. mylitta [117]
Sericin/Alginate Cell and drug delivery In vitro B. mori, 185 Nd- [118]
s
Sericin/Dextran Drug delivery system in vivo, C57BL/6 mice B. mori, Baiyu [39]
Sericin/Polyacrylamide Dermal reconstruction In vitro A. mylitta [119]
Sericin/Poly (γ-glutamic acid) Wound dressing in vivo, Sprague-Dawley Sericin powder [120]
rats
Sericin/Poly (NIPAm/MBA) Wound dressing in vivo, Sprague-Dawley Sericin powder [121]
rats
Sericin/Poly (NIPAm/LMSH) Antibacterial wound dressing in vivo, Sprague-Dawley Sericin powder [122]
rats
Sericin/Graphene oxide Calvarial bone regeneration in vivo, Sprague-Dawley 140 Nd-s [123]
rats
EBN/Sericin hypertrophic scars development at split-thickness skin graft Clinical Trial, Not yet B. mori [133]
donor site recruiting
Scaffolds
Sericin/PVA Full-thickness wound healing in vivo, Sprague-Dawley B. mori [124]
rats
Sericin/Chitosan Wound dressing In vitro B. mori [26]
Sericin/Chitosan Chronic nerve compression treatment in vivo, Sprague-Dawley B. mori, 140 Nd- [27]
rats s
Sericin/Collagen Cartilage regeneration In vitro B. mori [125]
Sericin/Carboxymethyl Cellulose Wound dressing In vitro A. mylitta [126]
Sericin/HAp Osteogenic differentiation In vitro A. pernyi [127]
Sericin/HAp Bone biomineralization In vitro A. pernyi [128]
Sericin/PVA/Glycerin Wound dressing in vivo, Wistar rats B. mori [129]
Sericin/microstructured Bacterial Cellulose Prospective gut repair In vitro B. mori [28]
(mBC)
Sericin/Carbon-Nanotubes (CNTs) Ischemic stroke damage treatment in vivo, C57BL/6 J mice B. mori, 140 Nd- [130]
s
Sericin/PVA Wound dressing for split-thickness skin graft donor site Clinical Trial, Phase 2 B. mori [131]
Sericin/PHMB/Bacterial Cellulose Wound dressing for split-thickness skin graft donor site Clinical Trial, Phase 2 B. mori [132,133]
Sericin/Collagen Wound dressing for split-thickness skin graft donor site Clinical Trial, Recruiting B. mori [133]
Sericin/Chitosan pad Wound dressing for split-thickness skin graft donor site Clinical Trial B. mori [133]
Conduits
Sericin Peripheral nerve regeneration in vivo, Sprague-Dawley B. mori, 140 Nd- [134]
rats s
Sericin/Silicone Peripheral nerve regeneration in vivo, Sprague-Dawley B. mori, 140 Nd- [22]
rats s
Sericin/Fibroin-PLGA Peripheral nerve regeneration in vivo, Sprague-Dawley B. mori [23]
rats
Sericin/Carbon Nanotube Peripheral nerve injury in vivo, Sprague-Dawley B. mori, 140 Nd- [24]
rats s
Devices
XSBR/SSCNT Sensors Human motion and skin temperature detection – Sericin powder [135]
SSCNT ink Human health and activity monitoring – B. mori [136]
Carbon black/Sericin@Fabrics Sweat loss monitoring – Unclear [137]
Flexible Li–S full battery – Unclear [138]
(continued on next page)

5
J. Liu et al. Biomaterials 287 (2022) 121638

Table 2 (continued )
Materials Applications Research stage Types of silk Reference
(s)

Sericin/S/NOCF and Li/NOCF


NOCF: (N/O-codoped carbonized silk
fabric)
SS@CNTs/Acetate Fabrics Textile strain sensor with electromagnetic shielding – Unclear [139]
Ag/Sericin/Au Resistance switching device – Unclear [140]

Table 3 now been widely used for tissue engineering and drug delivery. A variety
Biomedical applications of sericin-based nanoparticles. of sericin-based materials have been fabricated, including bulk materials
Uses of sericin Type of NPs Type of silk Reference (e.g. films, hydrogels, scaffolds, and conduits) and nano-/microparticles.
(s) In this section, we summarized different types of sericin-based materials
Drug delivery and their applications, and reviewed the clinical studies of sericin-based
Coating and AuNP@sericin (in vitro) unclear [177] products (Tables 2 and 3).
stabilizing AgNP@sericin (in vitro) SS powder [173]
agents MSN@sericin (in vivo) B. mori [165]
FGO@sericin (in vitro) B. mori [162] 3.1. Bulk formats
Sericin-drug FA-SND (in vivo) B. mori [172]
coupling FA-Ser-Chol (in vitro) SS power [178]
Pure sericin RSV@Ser (in vitro) unclear [167] 3.1.1. Films
nanocarrier ATR@Ser (in vivo) B. mori [166] Films (or membranes) are highly flexible and elastic formations that
Sericin-polymer Sericin/PBLG (in vivo) B. mori [169,170] allow gas exchange, water vapor transmission, and bacteria segregation
nanocarrier Sericin/poly- B. mori [168]
[141]. Skin, an important organ consisting of epidermis and dermis
ethylcyanoacrylate (in
vivo) layers, is easily damaged by acute trauma or chronic ulcers [142]. Thus,
Sericin/PLA (in vitro) B. mori [171] a variety of films have emerged to promote wound healing as dressing
SC-GG-RBA (in vitro) B. mori [179] materials [143].
MA-GG–SS–CS (in vitro) B. mori [180] As sericin possesses great biocompatibility and inherent bioactivities
Reducing agent
Nano Ag (in vitro) SS powder [181]
(supporting cell adhesion and proliferation, antioxidant property), it is
Nano Ag (in vitro) B. mori/ [175] suitable to be generated as films for skin regeneration [71]. For instance,
Samiacynthiaricini the glutaraldehyde-crosslinked sericin films and sericin/collagen com­
Nano manganese dioxide SS powder [176] posite films have been prepared for wound healing, which could support
(in vivo)
the attachment and proliferation of various types of cells (e.g., fibro­
Others
Anti-bacterial Sericin/TiO2 (in vitro) Raw silk filaments [182] blasts and keratinocytes) [42,104]. Sericin was also loaded in bacterial
anti- Sericin/alginate (in vivo) B. mori [183] cellulose (BC) films to fabricate sericin-functionalized BC composites
inflammatory (Fig. 3A) [105]. The sericin released from composites accelerated the
Would healing Sericin/magnesium Unclear [184] healing process, improved extracellular matrix production, and reduced
hydroxide (in vitro)
scar tissue formation. Furthermore, to endow the dressing materials
with antibacterial functions, the nanoparticles with antibacterial activ­
cryopreservation [98,100]. Thus, sericin would be an alternative with ity (silver or zinc oxide nanoparticles) were embedded in sericin-based
low cost to serum in cryopreservation for protecting cells and embryos films [108,111,144–146].
from oxidative stress damage. Sericin films can also promote bone regeneration. As shown in
Fig. 3B, sericin was wrapped on the metallic implant to enhance the
2.2.3.4. Metabolic regulation activity. As a dietary additive, sericin is adhesion, proliferation, and differentiation of osteoblast-like cells,
demonstrated to regulate glycolipid metabolism. In a type 2 diabetes rat which could benefit bone regeneration [11]. In another work, the
model induced by high-glucose/fat diet combined with streptozotocin, calcium-deficient carbonate apatite was in situ mineralized on sericin
oral administration of sericin significantly increased the levels of liver film to generate a hydroxyapatite/sericin composite film, which was
glycogen and insulin signaling pathway-related factors (IR, IRS-1, PI3K, capable of promoting the adhesion and viability of human osteosarcoma
and AKT) and reduced the blood glucose, suggesting that sericin may MG-63 cells [106].
regulate glucose metabolism through insulin-PI3K/AKT signaling
pathway [88,101]. Moreover, sericin could also regulate the lipid 3.1.2. Hydrogels
metabolism of obese animals [102]. Interestingly, sericin could facilitate Hydrogel is a three-dimensional (3D) network containing physically/
antiviral response by regulating glucose metabolism [55]. The sericin chemically crosslinked materials and a large amount of aqueous solu­
(B. Mori) and its derived F5-SP peptide (SEDSSEVDIDLGNLG) were tion, which is widely used in drug delivery and tissue engineering [147].
identified to be α-glucosidase inhibitors, which could suppress cellular Sericin protein contains a lot of polar amino acids that offer plentiful
glycolysis. By reducing lactate production, sericin significantly side groups for chemical crosslinking [148]. However, the sericin ob­
enhanced the innate immune response by promoting interferon tained by harsh degumming methods is inevitably degraded, which is
expression through RIG-I-MAVS signaling pathway, thus resulting in difficult to form stable hydrogels itself. To generate stable sericin net­
excellent anti-RNA virus activity in vitro and in vivo. These studies works, the degraded sericin was usually combined with other polymers
indicate the potential value of sericin for the treatment of metabolic (e.g., collagen, gelatin, chitosan, and poly (vinyl alcohol)) to form
diseases and viral infections. composite hydrogels [16,29,119,120,149]. For instance, a sericin/poly
(NIPAm/LMSH) nanocomposite hydrogel with antibacterial properties
3. Sericin-based biomaterials with different formats and was developed as a wound dressing [122]. The embedding of sericin
corresponding applications increased the internal pore sizes of interpenetrating network (IPN)
hydrogels, and interestingly shifted the structure of networks from
Due to its good biocompatibility and bioactivities, silk sericin has honeycomb to layered. Importantly, the sericin composite hydrogels
could adsorb bacteria, and interrupt the physiological activities of the

6
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 3. Preparation and applications of sericin-based films. (A) Sericin-Bacterial cellulose (BC) composite films were fabricated by a solution impregnation method,
which could accelerate the wound healing process, improve extracellular matrix production, and reduce scar formation [105]. (B) Sericin film was wrapped on the
metallic implant to enhance the adhesion, proliferation, and differentiation of osteoblast-like cells, thus promoting bone regeneration [11].

attached bacteria by released micromolecular sericin, thereby achieving this hydrogel could reduce the infarct size, inhibit the inflammatory
an antibacterial effect. Similarly, a lysozyme-loaded sericin/agarose response and apoptosis, and promote the formation of new blood vessels
hydrogel showed excellent antimicrobial activities and water absorption by injection into the acute myocardial infarction area in mice, thus
properties (Fig. 4A), possessing great potential in skin regeneration leading to the recovery of cardiac function [115].
applications [150]. In addition, some other composite hydrogels (e.g., Notably, using the degraded sericin, we have also generated an
sericin/poly (γ-glutamic acid), sericin/polyacrylamide, and ser­ injectable, photo-crosslinked pure sericin hydrogel [19]. The meth­
icin/chitosan hydrogels) were prepared as wound dressings, since acryloyl groups were introduced to sericin to synthesize sericin meth­
sericin could encourage fibroblasts proliferation and migration [85,117, acryloyl (SerMA), which could be crosslinked in situ under UV
119,120]. Aside from preparing simple hybrid hydrogels, our group irradiation through photo-polymerization. This SerMA hydrogel could
introduced hydrazide groups to sericin molecules. The sericin-derivative support cell attachment and proliferation as well as the intact sericin
was subsequently crosslinked with oxidized dextran to fabricate an hydrogels. Subsequently, graphene oxide was mingled into the
injectable sericin/dextran composite hydrogel for sustained release of photo-crosslinked SerMA hydrogels (SMH/GO) to enhance the me­
doxorubicin (DOX) in tumor local [39]. This hydrogel induced little chanical strength, whose compressive stress was increased by ~2 folds
immune response and inherited the photoluminescent property of (Fig. 4C) [123]. Notably, the sericin promoted bone marrow stromal
sericin for in vivo tracing. In addition to polymers, inorganic nano­ cells (BMSCs) migration, and the embedded graphene oxide specifically
particles (AgNPs or Ag@MOF-GO) also have been incorporated into encouraged osteogenesis differentiation of BMSCs. Thus, as an artificial
sericin hydrogels to endow them with antibacterial properties or he­ bone substitute, the hydrogel significantly accelerated calvarial bone
mostatic activity [151,152]. regeneration in mice compared with the commercial artificial bone
Since the harsh extraction procedures induce severe degradation and (Osteobone). Moreover, the degraded sericin-derivative (SerMA) was
denaturation of sericin, stable pure sericin networks were rarely pre­ used to generate AgNPs embedded cryogels (SMC@Ag), which exhibited
pared and investigated. To overcome the limitation, our group isolated improved hemostatic and antibacterial activities compared with com­
the intact sericin using a mild extraction method (LiBr method) from mercial gelatin sponge [154].
fibroin-deficient mutant silkworm strains (185 Nd-s and 140 Nd-s) [38].
Such sericin could be readily crosslinked with chemical agents (e.g., 3.1.3. Scaffolds
glutaraldehyde, genipin) or self-assemble under physical sonication to During tissue regeneration, biomaterial scaffolds are desired to
form pure sericin hydrogels [38,153]. As shown in Fig. 4B, the provide an instructive environment to recruit cells and promote cell
glutaraldehyde-crosslinked sericin hydrogel with photoluminescence proliferation, therefore accelerating healing cascades [155]. Sericin has
was able to support cell adhesion, proliferation, and long-term survival been prepared as scaffolds through foam processing, solvent casting, or
[38]. Moreover, the sericin hydrogel could sustained-release bioactive freeze-drying methods for tissue engineering.
reagents, making it a promising candidate for tissue engineering and For promoting skin tissue regeneration, a sericin/polyvinyl alcohol
regenerative medicine. Besides, the intact sericin was crosslinked with (PVA) scaffold was generated by freeze-drying and subsequently
genipin to generate a pure sericin hydrogel for repairing ischemic stroke precipitated in ethanol [124]. The scaffolds could promote L929 fibro­
and ischemic myocardial infarction [18,115]. This hydrogel could sup­ blasts proliferation in vitro, and significantly accelerate the full-thickness
port the attachment and growth of neurons in vitro and exhibited wound healing in rats. Moreover, a sericin/PVA scaffold containing
intrinsically neurotrophic and neuroprotective effects [18]. In addition, glycerin was fabricated [129]. The addition of glycerin effectively

7
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 4. Preparation and applications of sericin-based hydrogels. (A) A lysozyme-loaded sericin/agarose hydrogel was generated by the solution impregnation
method, which showed excellent antimicrobial activities and great water absorption properties, making it a potential alternative for wound dressing [150]. (B) Intact
sericin isolation from fibroin-deficient mutant silkworm strains, which could be readily crosslinked to fabricate sericin hydrogel [38]. (C) A sericin/graphene oxide
composite hydrogel was generated for calvarial bone regeneration, which could promote BMSCs migration and osteogenesis differentiation [123].

improved the flexibility properties and reduced the wound adhesion differentiation of BMSCs.
properties due to its uniform pore structure, which might be contributed For nerve regeneration, our group fabricated a nerve growth factor
to the effect of glycerin in reducing the phase separation between sericin (NGF)-loaded chitosan/sericin composite scaffold for the treatment of
and PVA. Importantly, the sustained release of sericin made the scaffolds chronic nerve compression (Fig. 5A) [27]. This scaffold could promote
appropriate for long-term healing of the wound. Schwann cells adhesion and proliferation. In a preclinical chronic nerve
For bone and cartilage regeneration, the sericin scaffolds mineralized compression rat model, the NGF-loaded scaffold promoted nerve func­
with hydroxyapatite (HAps) were prepared by a wet precipitation tional recovery via alleviating neuralgia, enhancing nerve conduction
method [127,128]. These scaffolds could not only support BMSCs velocity, accelerating microstructure recuperation, and improving
adhesion and proliferation but also promote the osteogenic gastrocnemius muscles dystrophy. Moreover, an injectable

8
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 5. Preparation and applications of sericin-based scaffolds and conduits. (A) A chitosan/sericin composite scaffold loaded with nerve growth factor (NGF) was
fabricated and applied for treating chronic nerve compression [27]. It can promote nerve functional recovery in a preclinical chronic nerve compression rat model via
alleviating neuralgia, enhancing nerve conduction velocity, accelerating microstructure recuperation, and improving gastrocnemius muscles. (B) A sericin nerve
conduit loaded with clobetasol was designed and fabricated for repairing a 10 mm gap in peripheral nerve in the rat injury model [24]. (C) A carbon nanotube/sericin
nerve conduit was employed to recover the peripheral nerve injury, showing great regenerative performance combined with electrical stimulation [25].

carbon-nanotubes-doped sericin scaffold (CNTs-SS) with programmable functions in a nerve injury rat model (10-mm gap defect) (Fig. 5C) [25].
shape-memory property was generated for treating ischemic stroke
[130]. The shape of CNTs-SS could be pre-designed according to the 3.1.5. Devices
personalized stroke damage, thus allowing injection into the cavity of As a degradable and biocompatible protein, sericin recently has
mice to cover the damage. It was worth noting that CNTs-SS scaffold attracted attention in the fields of electronics and sensing apart from
could deliver bone marrow mesenchymal stem cells (BMSCs) to the tissue engineering and drug delivery. The hydrophilic sericin usually
brain tissue and then functionally promote BMSCs’ neuronal differen­ serves as a binder and stabilizer to improve the electrical properties of
tiation, thus promoting stroke repair. flexible wearable sensors. Flexible devices based on nanocarbon mate­
rials are one of the important methods to produce wearable electronics.
3.1.4. Conduits The sericin-modified carbon nanotubes (CNTs) were combined with
Nerve guidance conduit (NGC) is generally formed with a hollow carboxylic styrene-butadiene rubber (XSBR) to generate a flexible
tube shape composed of natural or synthetic materials, which serves as network, which could serve as multi-functional sensors for detecting
an alternative to grafted nerve tissue for repairing peripheral nerve weak deformation and temperature variation (Fig. 6) [135]. Such
injury [134]. Due to the good biocompatibility and biodegradability, wearable devices provide a convenient strategy for monitoring human
sericin has been employed to generate conduits for nerve regeneration. healthcare and motion. Similarly, in combination with sericin and CNTs,
In our previous work, a nerve guidance conduit was generated by sericin a bio-ink (SSCNT) was generated to draw multifunctional circuits on
and silicone for peripheral nerve regeneration [22]. This sericin/silicone various substrate surfaces, and subsequently fabricate electrocardio­
conduit effectively promoted recovery of nerve morphology and func­ gram electrodes, respiration sensors, or electrochemical sensors for
tions during repairing a 5-mm gap defect in a sciatic nerve transection human healthcare monitoring [136].
model. To improve the repair performance, clobetasol (a glucocorticoid Lithium-sulfur batteries are promising high-energy power sources for
receptor agonist) was loaded in a sericin nerve conduit for treating a flexible wearable electronics, but are limited by the instability and low
longer gap defect (10 mm) [24] (Fig. 5B). The clobetasol-loaded sericin utilization of lithium-sulfur as well as poor electrode flexibility. The
conduit successfully repaired the nerve defect in rats, during which the sericin was utilized as adhesive binders to stabilize sulfur cathode, and
degradation products of conduit promoted neurotrophic factors secre­ then combined with the carbonized fibroin fabric to fabricate lith­
tion and clobetasol upregulated the myelin-related genes in Schwann iophilic and thiophilic stable electrodes with high areal capacity and
cells. Moreover, carbon nanotube (CNT) was introduced into the sericin great bending capability [138]. Moreover, sericin was selected to
conduit to endow it with electrical conductivity, which improved the generate a resistance-switching device with multilevel nonvolatile
performance of CNT/sericin conduit for restoring nerve structure and memory, as it possessed large resistance OFF/ON ratio and remained

9
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 6. Fabrication process and applications of the XSBR/SSCNT sensors [135]. Schematic illustration of the preparation (A) of flexible XSBR/SSCNT sensors and the
applications for human motion (B) and temperature detection (C).

insulation in native state [140]. The Ag/sericin/Au sandwich structure wound pain. In addition, no adverse events such as infection occurred
was performed as a simple metal-insulator-metal (MIM) cell structure [133]. The team also intends to conduct some other clinical trials,
and displayed significantly multilevel resistance-switching memory including the clinical efficacy of sericin/collagen hybrid scaffold on
performance. STSG donor sites, the clinical efficacy of sericin hydrogel sheet
impregnated with bird’s nest extract for the prevention of scar formation
3.1.6. Clinical studies in patients with STSG donor sites, and the clinical efficacy of ser­
Currently, most studies of sericin-based materials remain at the icin/chitosan pad on STSG donor sites. However, the results of these
laboratory stage, and only several clinical studies have been performed. clinical trials are currently unclear. Seong-Gon Kim et al. conducted a
In a randomized double-blind clinical trial, silver zinc sulfadiazine clinical trial of silk mats for guided bone regeneration (GBR), which
cream supplemented with sericin was used for treating second-degree contains both fibroin and sericin. Compared with the commercial dPTFE
burn wounds. The sericin-loaded cream (22.42 ± 6.33 days) signifi­ membrane, the effect of the silk mat on GBR after third molar extraction
cantly shortened wound healing time compared with the cream without surgery was comparable, and no obvious complications occurred [114].
sericin (29.28 ± 9.27 days), and no infection or severe adverse reactions Currently, clinical trials of sericin-based materials are still sparse, the
occurred in patients [112]. This is the first case of sericin safely and types of products are relatively simple, and the application scenarios are
effectively used in clinical research. In another clinical trial, topical circumscribed. This may be one of the reasons that hinder the
administration of sericin cream significantly reduced pruritus in he­ commercialization of sericin products. Therefore, more efforts and at­
modialysis patients and improved quality of life in uremia patients tempts are needed in this regard.
compared to placebo [113].
To demonstrate the potential of sericin in other applications, Por­ 3.2. Particles
nanong Aramwit et al. developed a sericin/PVA hybrid scaffold, which
promoted wound healing more efficiently than the PVA scaffold without 3.2.1. Nanoparticles
sericin in rats. This result was further confirmed in a clinical trial of With the advancement of nanotechnology, nanomaterials have been
treating split-thickness skin graft (STSG) donor site. The sericin/PVA widely used for controlled drug delivery and diagnosis. Generally,
scaffold markedly shortened the time for complete healing of the pa­ nanostructure endows materials with unique physical and chemical
tient’s wound skin compared to the commercially available Bactigras® properties, such as high surface area, dispersion, and enhanced perme­
dressing [131]. ability and retention (EPR) effects, which make nanomaterials exten­
In another study by this team, a bacterial cellulose wound dressing sively applied in drug delivery [156,157]. Due to its modifiable structure
(BCSP) containing sericin and polyhexamethylene biguanide was and excellent biological characteristics (e.g., amphiphilicity, pH
developed. When implanted subcutaneously in rats, the BCSP elicited a responsiveness, biodegradability, and good biocompatibility), sericin
lower inflammatory response compared to Bactigras®. Clinical patch possesses unique advantages in fabricating nano- and micro-scale ma­
experiments were performed on the normal skin of healthy volunteers, terials [16]. In Table 3, we have summarized the recent studies of
and no skin irritation symptoms appeared, such as erythema, melanin sericin-based micro/nano-structures.
accumulation, edema, and papules [132]. Further clinical trials were The stability and dispersibility of nanoparticles (NPs) in the physi­
conducted for the treatment of STSG donor sites with this dressing ological environment are important factors, because aggregation of
(BCSP). Although BCSP did not significantly accelerate the wound either the NPs themselves or the blood cells caused by NPs may cause
healing process compared with Bactigras®, the BCSP treatment inflammation and acute toxicity [158]. Thus, the NPs should be modi­
encouragingly improved the wound quality and reduced the patient’s fied or coated with dispersion stabilizers to enhance stability and reduce

10
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 7. Preparation and applications of sericin-based nanostructures. (A) Sericin was wrapped on mesoporous silica nanoparticles (MSNs) to improve their dispersion
and stability in aqueous solutions. At the same time, serving as a gatekeeper, sericin responded to protease and acidic condition to control the release of loaded drugs
[165]. (B) Sericin-PBLG micelles enhanced the delivery of DOX for the treatment of drug-resistant tumors [169]. (C) Sericin-based nanoparticles underwent a charge
reversal process due to the change of amino/carboxyl ratio under different pH conditions, thereby achieving pH-dependent cellular internalization and drug release
in the tumor [30]. (D) Sericin was used as a reducing agent to assist the synthesis of silver nanoparticles [173].

serum protein adsorption in the circulatory system [159–161]. Sericin degraded sericin peptides, thus leading to DOX release (Fig. 7A).
contains abundant polar amino acids, which can interact with water Except for coating nanoparticles, sericin itself can be prepared as
molecules to form a hydration layer. In this respect, sericin is possible to nanocarriers for insoluble molecules (e.g., resveratrol and resveratrol) to
be used as a dispersion stabilizer to enhance the stability of enhance their therapeutic effects and reduce systemic toxicity [166,
nanoparticles. 167]. Moreover, nanocomposites formed by sericin and other polymers
Fluorinated graphene (FGO) is a high-efficiency drug carrier and (e.g., synthetic polymers, polysaccharides, and proteins) have been
photoacoustic contrast agent. Its highly inert and repellent surface generated as therapeutic drug carriers [168–171].
makes it extremely unstable in aqueous solutions. To solve this problem, Sericin/poly-ethylcyanoacrylate nanospheres were prepared through
sericin was developed as a stabilizer to fabricate water-steady FGO interfacial polymerization to improve the bioavailability of fenofibrate
nanocarriers, which efficiently loaded curcumin and achieved a pro­ (a lipid-lowering drug) [168]. In another study, an amphiphilic polymer
longed circulation in blood [162]. Similarly, by coating with sericin, the micelle (sericin backbone and poly (γ-benzyl-L-glutamate) side chains)
nanomaterials (e.g., carbon nanotubes, zeolitic imidazolate was fabricated through ring-opening polymerization [169]. Because of
framework-8) can obtain enhanced stability and biocompatibility [139, the high cell membrane penetration, such micelles effectively delivered
163,164]. In our previous study, sericin was wrapped on DOX-loaded DOX into tumor cells (Fig. 7B). On this basis, vitamin B12 was further
mesoporous silica nanoparticles (MSNs) as a gatekeeper through conjugated on sericin molecules to enhance the targeting capability of
pH-sensitive hydrazone linkages, forming a pH/protease dual respon­ micelle for CD320-overexpressed gastric cancer cells [170].
sive drug delivery system [165]. The sericin coating endowed silica Given the abundant acidic and basic amino acids, sericin qualifies as
nanoparticles with good dispersibility and prevented drug premature a unique pH-sensitive material to construct “smart” nanocarriers. For
leakage. When the nanoparticles were internalized into tumor cells instance, a pH-responsive charge reversal sericin/chitosan nanoparticle
through endocytosis, the acidity microenvironment of lysosomes (SSC@NP) was prepared by crosslinking negatively-charged sericin with
cleaved the pH-sensitive hydrazone bonds and the protease in lysosomes positively-charged chitosan [30]. This nanoparticle exhibited a negative

11
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 8. Preparation and applications of sericin-based microstructures. (A) The sericin microspheres were synthesized by an emulsification method for sustained
release of DOX, which were used for the treatment of metastatic lung cancer by pulmonary administration [189]. (B) Preparation process and morphology control of
sericin-based microparticles [186]. (C) Fabrication of microstructures using sericin lithography, and fluorescence images of osteoblasts proliferated on sericin
scaffolds with selective patterning [190].

charge in the neutral condition, while the surface charge converted to be friendly method.
positive under acidic condition (pH 6.0) due to the increased amino/­
carboxyl ratio. Owing to this charge reversal property, SSC@NP signif­ 3.2.2. Microparticles
icantly increased cellular internalization under tumor acidic condition To enable topical drug delivery in certain tissues or organs with
by 6.0-fold compared with the neutral environment, and sufficiently special structures, and deliver larger particles (or cells), sericin micro­
delivered DOX into cancer cells (Fig. 7C) [30]. Similarly, a pH-sensitive particles are developed as an effective supplement to the drug delivery
charge-reversal fluorinated graphene oxide was prepared by modifying systems. Various methods are utilized for fabricating sericin-based mi­
cationic polymer (polyethyleneimine) and sericin, which could shift the croparticles [185–189].
surface charge in response to environmental pH values for promoting For treating metastatic lung cancer, our group prepared sericin mi­
circulation stability and enhancing intracellular drug delivery [162]. In croparticles (SMPs) using a water-in-oil (w/o) emulsification method,
addition to adjusting the ratio of acidic and basic amino acids to achieve followed by DOX loading and coating with tannic acid (TA)/ferric irons
pH responsiveness, abundant modifiable functional groups of sericin can (Fe3+) based metal-organic networks (Fig. 8A). These reasonably
also be employed to introduce pH-sensitive properties [165,172]. For designed microparticles could be effectively deposited in lungs and
instance, we grafted DOX to sericin molecules through the hydrazone remain for more than 5 days through pulmonary administration in vivo,
bonds to generate a pH-sensitive sericin-drug conjugate, which could and continuously release anticancer drugs in a pH-dependent manner.
release DOX in acidic lysosomes [172]. The DOX-loaded microparticles (DOX@SMP-MON) significantly
In addition to serving as drug carriers, sericin is also used as a bio­ reduced the number of metastatic nodules in lungs in the metastatic
mineralized matrix or reducing agent for generating sericin/inorganic breast cancer (4T1) model [189].
nanocomposites [173–176]. For instance, sericin was used as a template A one-step green route to fabricate sericin-based microcapsules
to mediate the nucleation of hydroxyapatite crystals [127]. Similarly, without the aid of organic solvents was reported as well (Fig. 8B) [186].
the self-assembled sericin nanofibers were used as templates to regulate The sericin aqueous solution (1% wt) was mixed with certain volumes of
silica nucleation and assembly for preparing nano-silica drug carriers CaCl2 solution (0.2 M) and stirred at 37 ◦ C for 24 h at pH 7.4. The sericin
[174]. As a reducing agent, sericin was used to synthesize silver nano­ conformation was changed through chelation of Ca ions, resulting in
particles (AgNPs), which simultaneously served as a stabilizer [173] microcapsules formation by self-assembly. The morphology of micro­
(Fig. 7D). Interestingly, the reducing activity of sericin varies with capsules could be easily regulated to form bionic structures (discoidal,
different origins of cocoons. Sericin derived from non-mulberry silk­ biconcave, cocoon-like, and tubular structures) by adjusting the con­
worms exhibits higher reducing activity than that from mulberry silk­ centration of calcium ions and stirring speed [186]. In addition, the pH
worms, which is beneficial to AgNPs formation [175]. This simple values of buffers and the types of ions could also affect the assembly of
synthesis strategy requires neither additional reducing agents nor sericin microspheres [188]. In another work, the 2-isocyanatoethyl
excessive heat treatments, that is an economical and environmentally methacrylate (IEM) modified sericin has been synthesized for

12
J. Liu et al. Biomaterials 287 (2022) 121638

Fig. 9. Prospects of exploration and applications of silk sericin.

preparing a photo-crosslinkable sericin photoresist (Fig. 8C). Through of sericin-alginate beads (inner core) and chitosan (outer shell) via
photolithography, it was easy to precisely control the sericin cross­ ionotropic gelation [191]. The microcapsules prevented graft rejection
linking to customize the specific spatial morphology and complex by isolating the immune environment of the host. Meanwhile, the
patterning of microstructures. These high-resolution microstructures porous structures of microcapsules allowed nutrient exchange, ensuring
could be used as a suitable substrate for cell culture and guide cells to the survival and biological metabolism of liver cells in the capsules. This
grow in a particular direction without additional adhesion ligands microcapsule-based cell therapy strategy is expected to generate an
[190]. enriched population of metabolically and functionally active cells for
Transplanted cells for tissue engineering are usually vulnerable to hepatocytes transplantation in acute liver failure without the addition of
the host immune system and exhibit a very low proliferation rate. To immunosuppressants.
address this problem, Nayak et al. prepared the microcapsules composed

13
J. Liu et al. Biomaterials 287 (2022) 121638

4. Perspectives (5) The sericin contains a large number of random coils in the
structure that would increase the brittleness of sericin-based
With recent advances and the development of sericin-based bio­ materials [16]. Besides, the harsh physical/chemical extraction
materials for tissue engineering and regeneration medicine, sericin, the methods further decrease the molecular weights of isolated
by-product of silk industry, begins to show great potential in biomedical peptides, which further results in defective properties [16,34,48].
research and applications. The relevant developments may change the Thus, the sericin biomaterials usually suffer from poor mechan­
current status of silk industry and promote the transformation and ical properties.
modernization of traditional industries as well as tissue engineering and
regeneration medicine. According to FDA’s classification management rules for medical
Although plenty of silk sericin-derived biomaterials (from nano-size devices, it is essential to demonstrate the substantial equivalence of new
to bulk) have been developed and investigated, no sericin-derived medical devices to a predicate device (a legally marketed one), including
medical products were available on the market until now. On the con­ the safety and effectiveness. But as aforementioned, there are various
trary, the silk fibroin, which also originates from silk cocoons, has been challenges in the development of sericin-derived products, such as the
manufactured to be commercialized medical products for different biosafety, efficacy, mechanical property, and batch difference. To
application scenarios (e.g., SERI® surgical scaffold, Sofsilk™ Silk Su­ address these issues, great research efforts on sericin need to be directed
tures, Silk Voice®, Sidaiyi® wound dressing). After comparing the two towards (1) the development of novel sericin extraction techniques that
proteins from silkworm cocoons, we speculate the following issues that could efficiently isolate sericin from silkworm cocoons (both wild-type
may hinder the development and clinical translation of sericin and mutant cocoons) with minimized degradation and denaturation;
biomaterials: (2) identification of peptide sequences and conformation of sericin; (3)
active preclinical and clinical studies on the biosafety and effectiveness
(1) Silk sericin has been regarded as the waste of textile industry for of sericin and sericin-based formulations; (4) in-depth mechanism
thousands of years, which leads to significantly less attention to exploration on the bioactivities of sericin and its derived peptides; and
sericin compared with fibroin. Moreover, the long-term misun­ (5) enhancement of the mechanical properties of sericin-based
derstanding of sericin’s immunogenicity had hindered the materials.
development and application of sericin-based biomaterials. As a Of note, among these sericin-based materials summarized in this
result, the studies on sericin are far less than those on fibroin, review, the wearable electronic devices generated by sericin could
including the structure characterization, exploration of biological conventionally monitor personal healthcare, such as human movement,
activities, and product development. skin temperature, and sweat loss. These devices could be directly
(2) It is a challenge to obtain sericin protein with high quality attached to human skin or clothes avoiding the safety concerns of
(without degradation and denaturation) on a large or industrial sericin, which may be easier for translation or commercialization.
scale. Although we have dissolved and isolated the intact sericin Overall, despite some “uncultivated lands” on sericin investigation,
protein from the fibroin-deficient silkworm cocoons using a mild such natural protein exhibits great potential for the generation of novel
extraction method, the resources of mutant silkworm cocoons are materials for tissue engineering and regeneration medicine. In the
quite limited. For the wild-type silkworm cocoons, including both future, it is encouraged to develop sericin-based materials with intelli­
mulberry and non-mulberry categories, all the extraction pro­ gent responsiveness and on-demand maneuverability, which may be one
cesses would induce unfavorable degradation and denaturation of the promising research directions for sericin materials. In addition,
of sericin protein. This issue dramatically limits the in-depth different forms of materials based on sericin, such as the “inks” for 3D
studies on sericin properties and fabrication of sericin-based printing [192], microneedles [193], wearable electronic devices, and
biomaterials. even smart micro/nano robots can be developed for various biomedical
(3) The immunogenicity of sericin has long been controversial, applications (Fig. 9). The applications of sericin in these interdisci­
although many studies indicated it to be a safe and suitable plinary fields are also worth exploring.
biomaterial. Recently, we have investigated the biocompatibility
of sericin derived from wild-type and fibroin-deficient mutant Declaration of competing interest
silkworm cocoons (B. mori) in BALB/c mice, and demonstrated
the low immunogenicity and allergenicity. However, this study The authors declare that they have no known competing financial
only focused on the mulberry silkworm cocoons and tested the interests or personal relationships that could have appeared to influence
immune responses in mice. Thus, the biocompatibility of the work reported in this paper.
different types of silkworm cocoons should be further studied,
and the immune responses of sericin in other animals or even Acknowledgments
humans need to be examined.
(4) The underlying mechanisms of sericin promoting tissue repair are This work was supported by the National Natural Science Foundation
not clear. In our previous work, we explored the mechanism of of China [82072167, 81873931, 82072068, and 81974382]; the Joint
sericin in stimulating angiogenesis and found that sericin Fund of Ministry of Education for Equipment Pre-research
enhanced vascular endothelial growth factor-a (VEGFa) expres­ [6141A02022626], the Major Scientific and Technological Innovation
sion via activating extracellular regulated protein kinases (ERK) Projects in Hubei Province [2018ACA136], the Integrated Innovative
phosphorylation [115]. According to the RNA sequencing, Team for Major Human Diseases Program of Tongji Medical College of
sericin-based scaffold has been demonstrated to induce BMSCs HUST, the Academic Doctor Supporting Program of Tongji Medical
migration and osteogenesis differentiation via activating College of HUST, and the Union Hospital Foundation for Young Scientist
mitogen-activated protein kinases (MAPK), tumor necrosis factor (2021xhqh01).
(TNF), and chemokine signaling pathways, thereby improving
bone regeneration [123]. However, we did not identify the spe­ References
cific peptide sequence on sericin that activates these signaling
and pathways. Since sericin is a mixture of multiple proteins (or [1] S. Blake, N.Y. Kim, N. Kong, J. Ouyang, W. Tao, Silk’s cancer applications as a
biodegradable material, Mater. Today Sustain. 13 (2021), 100069, https://doi.
peptides) and the extraction procedures cleave these proteins, it org/10.1016/j.mtsust.2021.100069.
is difficult to identify the specific receptors and related signal
pathways until every single fragment could be isolated.

14
J. Liu et al. Biomaterials 287 (2022) 121638

[2] W. Huang, S. Ling, C. Li, F.G. Omenetto, D.L. Kaplan, Silkworm silk-based [27] L. Zhang, W. Yang, K. Tao, Y. Song, H. Xie, J. Wang, X. Li, X. Shuai, J. Gao,
materials and devices generated using bio-nanotechnology, Chem. Soc. Rev. 47 P. Chang, G. Wang, Z. Wang, L. Wang, Sustained local release of NGF from a
(17) (2018) 6486–6504, https://doi.org/10.1039/C8CS00187A. chitosan-sericin composite scaffold for treating chronic nerve compression, ACS
[3] C. Holland, K. Numata, J. Rnjak-Kovacina, F.P. Seib, The biomedical use of silk: Appl. Mater. Interfaces 9 (4) (2017) 3432–3444, https://doi.org/10.1021/
past, present, future, Adv. Healthc. Mater. 8 (1) (2019), e1800465, https://doi. acsami.6b14691.
org/10.1002/adhm.201800465. [28] L. Lamboni, C. Xu, J. Clasohm, J. Yang, M. Saumer, K.H. Schafer, G. Yang, Silk
[4] F.G. Omenetto, D.L. Kaplan, New opportunities for an ancient material, Science sericin-enhanced microstructured bacterial cellulose as tissue engineering
329 (5991) (2010) 528–531, https://doi.org/10.1126/science.1188936. scaffold towards prospective gut repair, Mater. Sci. Eng., C 102 (2019) 502–510,
[5] Y. Wang, H.J. Kim, G. Vunjak-Novakovic, D.L. Kaplan, Stem cell-based tissue https://doi.org/10.1016/j.msec.2019.04.043.
engineering with silk biomaterials, Biomaterials 27 (36) (2006) 6064–6082, [29] G. Das, H.S. Shin, E.V.R. Campos, L.F. Fraceto, M. Del Pilar Rodriguez-Torres, K.
https://doi.org/10.1016/j.biomaterials.2006.07.008. C.F. Mariano, D.R. de Araujo, F. Fernandez-Luqueno, R. Grillo, J.K. Patra, Sericin
[6] B. Kundu, R. Rajkhowa, S.C. Kundu, X. Wang, Silk fibroin biomaterials for tissue based nanoformulations: a comprehensive review on molecular mechanisms of
regenerations, Adv. Drug Deliv. Rev. 65 (4) (2013) 457–470, https://doi.org/ interaction with organisms to biological applications, J. Nanobiotechnol. 19 (1)
10.1016/j.addr.2012.09.043. (2021) 30, https://doi.org/10.1186/s12951-021-00774-y.
[7] M. Gholipourmalekabadi, S. Sapru, A. Samadikuchaksaraei, R.L. Reis, D. [30] D. Hu, Z. Xu, Z. Hu, B. Hu, M. Yang, L. Zhu, pH-Triggered charge-reversal silk
L. Kaplan, S.C. Kundu, Silk fibroin for skin injury repair: where do things stand? sericin-based nanoparticles for enhanced cellular uptake and doxorubicin
Adv. Drug Deliv. Rev. 153 (2020) 28–53, https://doi.org/10.1016/j. delivery, ACS Sustainable Chem. Eng. 5 (2) (2017) 1638–1647, https://doi.org/
addr.2019.09.003. 10.1021/acssuschemeng.6b02392.
[8] P.W. Madden, I. Klyubin, M.J. Ahearne, Silk fibroin safety in the eye: a review [31] G.H. Altman, F. Diaz, C. Jakuba, T. Calabro, R.L. Horan, J. Chen, H. Lu,
that highlights a concern, BMJ Open Ophthalmol. 5 (1) (2020), e000510, https:// J. Richmond, D.L. Kaplan, Silk-based biomaterials, Biomaterials 24 (3) (2003)
doi.org/10.1136/bmjophth-2020-000510. 401–416, https://doi.org/10.1016/s0142-9612(02)00353-8.
[9] P. Aramwit, T. Siritientong, T. Srichana, Potential applications of silk sericin, a [32] R. Konwarh, B.K. Bhunia, B.B. Mandal, Opportunities and challenges in exploring
natural protein from textile industry by-products, Waste Manag. Res. 30 (3) Indian non-mulberry silk for biomedical applications, Proc. Indian Natl. Sci.
(2012) 217–224, https://doi.org/10.1177/0734242X11404733. Acad. 83 (1) (2017) 85–101, https://doi.org/10.16943/ptinsa/2017/41288.
[10] P. Aramwit, S. Kanokpanont, T. Nakpheng, T. Srichana, The effect of sericin from [33] R. Dash, M. Mandal, S.K. Ghosh, S.C. Kundu, Silk sericin protein of tropical tasar
various extraction methods on cell viability and collagen production, Int. J. Mol. silkworm inhibits UVB-induced apoptosis in human skin keratinocytes, Mol. Cell.
Sci. 11 (5) (2010) 2200–2211, https://doi.org/10.3390/ijms11052200. Biochem. 311 (1–2) (2008) 111–119, https://doi.org/10.1007/s11010-008-
[11] S. Nayak, T. Dey, D. Naskar, S.C. Kundu, The promotion of osseointegration of 9702-z.
titanium surfaces by coating with silk protein sericin, Biomaterials 34 (12) (2013) [34] B. Kundu, N.E. Kurland, V.K. Yadavalli, S.C. Kundu, Isolation and processing of
2855–2864, https://doi.org/10.1016/j.biomaterials.2013.01.019. silk proteins for biomedical applications, Int. J. Biol. Macromol. 70 (2014) 70–77,
[12] J.R. Eidet, S. Reppe, L. Pasovic, O.K. Olstad, T. Lyberg, A.Z. Khan, I.G. Fostad, D. https://doi.org/10.1016/j.ijbiomac.2014.06.022.
F. Chen, T.P. Utheim, The silk-protein sericin induces rapid melanization of [35] T.T. Cao, Y.Q. Zhang, Processing and characterization of silk sericin from Bombyx
cultured primary human retinal pigment epithelial cells by activating the NF-κB mori and its application in biomaterials and biomedicines, Mater. Sci. Eng., C 61
pathway, Sci. Rep. 6 (2016), 22671, https://doi.org/10.1038/srep22671. (2016) 940–952, https://doi.org/10.1016/j.msec.2015.12.082.
[13] L. Pasovic, T.P. Utheim, S. Reppe, A.Z. Khan, C.J. Jackson, B. Thiede, J.P. Berg, E. [36] E. Bari, S. Perteghella, S. Farago, M.L. Torre, Association of silk sericin and
B. Messelt, J.R. Eidet, Improvement of storage medium for cultured human retinal platelet lysate: premises for the formulation of wound healing active medications,
pigment epithelial cells using factorial design, Sci. Rep. 8 (1) (2018) 5688, Int. J. Biol. Macromol. 119 (2018) 37–47, https://doi.org/10.1016/j.
https://doi.org/10.1038/s41598-018-24121-8. ijbiomac.2018.07.142.
[14] N. Kato, S. Sato, A. Yamanaka, H. Yamada, N. Fuwa, M. Nomura, Silk protein, [37] Y.J. Wang, Y.Q. Zhag, Three-layered sericins around the silk fibroin fiber from
sericin, inhibits lipid peroxidation and tyrosinase activity, Biosci., Biotechnol., Bombyx mori cocoon and their amino acid composition, Adv. Mater. Res.
Biochem. 62 (1) (1998) 145–147, https://doi.org/10.1271/bbb.62.145. 175–176 (2011) 158–163. https://dx.doi.org/10.4028/www.scientific.net/AMR.
[15] K. Jena, J.P. Pandey, R. Kumari, A.K. Sinha, V.P. Gupta, G.P. Singh, Free radical 175-176.158.
scavenging potential of sericin obtained from various ecoraces of tasar cocoons [38] Z. Wang, Y. Zhang, J. Zhang, L. Huang, J. Liu, Y. Li, G. Zhang, S.C. Kundu,
and its cosmeceuticals implication, Int. J. Biol. Macromol. 120 (Pt A) (2018) L. Wang, Exploring natural silk protein sericin for regenerative medicine: an
255–262, https://doi.org/10.1016/j.ijbiomac.2018.08.090. injectable, photoluminescent, cell-adhesive 3D hydrogel, Sci. Rep. 4 (2014) 7064,
[16] L. Lamboni, M. Gauthier, G. Yang, Q. Wang, Silk sericin: a versatile material for https://doi.org/10.1038/srep07064.
tissue engineering and drug delivery, Biotechnol. Adv. 33 (8) (2015) 1855–1867, [39] J. Liu, C. Qi, K. Tao, J. Zhang, J. Zhang, L. Xu, X. Jiang, Y. Zhang, L. Huang, Q. Li,
https://doi.org/10.1016/j.biotechadv.2015.10.014. H. Xie, J. Gao, X. Shuai, G. Wang, Z. Wang, L. Wang, Sericin/dextran injectable
[17] R.I. Kunz, R.M. Brancalhao, L.F. Ribeiro, M.R. Natali, Silkworm sericin: properties hydrogel as an optically trackable drug delivery system for malignant melanoma
and biomedical applications, BioMed Res. Int. 2016 (2016), 8175701, https:// treatment, ACS Appl. Mater. Interfaces 8 (10) (2016) 6411–6422, https://doi.
doi.org/10.1155/2016/8175701. org/10.1021/acsami.6b00959.
[18] Z. Wang, J. Wang, Y. Jin, Z. Luo, W. Yang, H. Xie, K. Huang, L. Wang, [40] Y.N. Jo, B.D. Park, I.C. Um, Effect of storage and drying temperature on the
A neuroprotective sericin hydrogel as an effective neuronal cell carrier for the gelation behavior and structural characteristics of sericin, Int. J. Biol. Macromol.
repair of ischemic stroke, ACS Appl. Mater. Interfaces 7 (44) (2015) 81 (2015) 936–941, https://doi.org/10.1016/j.ijbiomac.2015.09.016.
24629–24640, https://doi.org/10.1021/acsami.5b06804. [41] H. Teramoto, M. Miyazawa, Analysis of structural properties and formation of
[19] C. Qi, J. Liu, Y. Jin, L. Xu, G. Wang, Z. Wang, L. Wang, Photo-crosslinkable, Sericin fiber by infrared spectroscopy, J. Insect Biotechnol. Sericol. 72 (3) (2003)
injectable sericin hydrogel as 3D biomimetic extracellular matrix for minimally 157–162, https://doi.org/10.11416/jibs.72.157.
invasive repairing cartilage, Biomaterials 163 (2018) 89–104, https://doi.org/ [42] S. Nayak, S. Talukdar, S.C. Kundu, Potential of 2D crosslinked sericin membranes
10.1016/j.biomaterials.2018.02.016. with improved biostability for skin tissue engineering, Cell Tissue Res. 347 (3)
[20] S. Tyeb, N. Kumar, A. Kumar, V. Verma, Flexible agar-sericin hydrogel film (2012) 783–794, https://doi.org/10.1007/s00441-011-1269-4.
dressing for chronic wounds, Carbohydr. Polym. 200 (2018) 572–582, https:// [43] M. Yuksek, D. Kocak, A. Beyit, N.J.A.i.E.B. Merdan, Effect of degumming
doi.org/10.1016/j.carbpol.2018.08.030. performed with different type natural soaps and through ultrasonic method on
[21] L. Ai, Y. Wang, G. Tao, P. Zhao, A. Umar, P. Wang, H. He, Polydopamine-based the properties of silk fiber, Adv. Environ. Biol. 6 (2) (2012) 801–808.
surface modification of ZnO nanoparticles on sericin/polyvinyl alcohol composite [44] D. Gupta, A. Agrawal, H. Chaudhary, M. Gulrajani, C. Gupta, Cleaner process for
film for antibacterial application, Molecules 24 (3) (2019) 503, https://doi.org/ extraction of sericin using infrared, J. Clean. Prod. 52 (2013) 488–494, https://
10.3390/molecules24030503. doi.org/10.1016/j.jclepro.2013.03.016.
[22] H. Xie, W. Yang, J. Chen, J. Zhang, X. Lu, X. Zhao, K. Huang, H. Li, P. Chang, [45] A. Kurioka, F. Kurioka, M. Yamazaki, Characterization of sericin powder prepared
Z. Wang, L. Wang, A silk sericin/silicone nerve guidance conduit promotes from citric acid-degraded sericin polypeptides of the silkworm, Bombyx Mori,
regeneration of a transected sciatic nerve, Adv. Healthc. Mater. 4 (15) (2015) Biosci. Biotechnol. Biochem. 68 (4) (2004) 774–780, https://doi.org/10.1271/
2195–2205, https://doi.org/10.1002/adhm.201500355. bbb.68.774.
[23] J. Rao, Y. Cheng, Y. Liu, Z. Ye, B. Zhan, D. Quan, Y. Xu, A multi-walled silk [46] G. Freddi, G. Allara, G.J.C.T. Candiani, Degumming of silk fabrics with tartaric
fibroin/silk sericin nerve conduit coated with poly(lactic-co-glycolic acid) sheath acid, J. Soc. Dye. Colour. 112 (7–8) (2010) 191–195, https://doi.org/10.1111/
for peripheral nerve regeneration, Mater. Sci. Eng., C 73 (2017) 319–332, https:// j.1478-4408.1996.tb01817.x.
doi.org/10.1016/j.msec.2016.12.085. [47] N.M. Mahmoodi, M. Arami, F. Mazaheri, S. Rahimi, Degradation of sericin
[24] L. Zhang, W. Yang, H. Xie, H. Wang, J. Wang, Q. Su, X. Li, Y. Song, G. Wang, (degumming) of Persian silk by ultrasound and enzymes as a cleaner and
L. Wang, Z. Wang, Sericin nerve guidance conduit delivering therapeutically environmentally friendly process, J. Clean. Prod. 18 (2) (2010) 146–151, https://
repurposed clobetasol for functional and structural regeneration of transected doi.org/10.1016/j.jclepro.2009.10.003.
peripheral nerves, ACS Biomater. Sci. Eng. 5 (3) (2019) 1426–1439, https://doi. [48] H. Yun, H. Oh, M.K. Kim, H.W. Kwak, J.Y. Lee, I.C. Um, S.K. Vootla, K.H. Lee,
org/10.1021/acsbiomaterials.8b01297. Extraction conditions of Antheraea mylitta sericin with high yields and minimum
[25] X. Li, W. Yang, H. Xie, J. Wang, L. Zhang, Z. Wang, L. Wang, CNT/Sericin molecular weight degradation, Int. J. Biol. Macromol. 52 (2013) 59–65, https://
conductive nerve guidance conduit promotes functional recovery of transected doi.org/10.1016/j.ijbiomac.2012.09.017.
peripheral nerve injury in a rat model, ACS Appl. Mater. Interfaces 12 (33) (2020) [49] W. Lamoolphak, W. De-Eknamkul, A. Shotipruk, Hydrothermal production and
36860–36872, https://doi.org/10.1021/acsami.0c08457. characterization of protein and amino acids from silk waste, Bioresour. Technol.
[26] Z. Karahaliloglu, E. Kilicay, E.B. Denkbas, Antibacterial chitosan/silk sericin 3D 99 (16) (2008) 7678–7685, https://doi.org/10.1016/j.biortech.2008.01.072.
porous scaffolds as a wound dressing material, Artif. Cell Nanomed. Biotechnol.
45 (6) (2017) 1–14, https://doi.org/10.1080/21691401.2016.1203796.

15
J. Liu et al. Biomaterials 287 (2022) 121638

[50] K.U. Sprague, The Bombyx mori silk proteins: characterization of large applications, Acta Biomater. 5 (8) (2009) 3007–3020, https://doi.org/10.1016/j.
polypeptides, Biochemistry 14 (5) (1975) 925–931, https://doi.org/10.1021/ actbio.2009.03.026.
bi00676a008. [75] B. Panilaitis, G.H. Altman, J. Chen, H.J. Jin, V. Karageorgiou, D.L. Kaplan,
[51] B.C. Dash, B.B. Mandal, S.C. Kundu, Silk gland sericin protein membranes: Macrophage responses to silk, Biomaterials 24 (18) (2003) 3079–3085, https://
fabrication and characterization for potential biotechnological applications, doi.org/10.1016/s0142-9612(03)00158-3.
J. Biotechnol. 144 (4) (2009) 321–329, https://doi.org/10.1016/j. [76] Y.Q. Zhang, Y. Ma, Y.Y. Xia, W.D. Shen, J.P. Mao, R.Y. Xue, Silk sericin-insulin
jbiotec.2009.09.019. bioconjugates: synthesis, characterization and biological activity, J. Contr.
[52] P. Aramwit, S. Damrongsakkul, S. Kanokpanont, T. Srichana, Properties and Release 115 (3) (2006) 307–315, https://doi.org/10.1016/j.jconrel.2006.08.019.
antityrosinase activity of sericin from various extraction methods, Biotechnol. [77] Z. Jiao, Y. Song, Y. Jin, C. Zhang, D. Peng, Z. Chen, P. Chang, S.C. Kundu,
Appl. Biochem. 55 (2) (2010) 91–98, https://doi.org/10.1042/BA20090186. G. Wang, Z. Wang, L. Wang, In vivo characterizations of the immune properties of
[53] S.G. Kim, H. Kweon, Y.Y. Jo, Toll-like receptor and silk sericin for tissue sericin: an ancient material with emerging value in biomedical applications,
engineering, Int. J. Ind. Entomol. 42 (1) (2021) 1–6, https://doi.org/10.7852/ Macromol. Biosci. 17 (12) (2017), https://doi.org/10.1002/mabi.201700229.
ijie.2021.42.1.1. [78] S. Terada, T. Nishimura, M. Sasaki, H. Yamada, M. Miki, Sericin, a protein derived
[54] Y.Y. Jo, H. Kweon, D.W. Kim, K. Baek, W.S. Chae, Y.J. Kang, J.H. Oh, S.G. Kim, from silkworms, accelerates the proliferation of several mammalian cell lines
U. Garagiola, Silk sericin application increases bone morphogenic protein-2/4 including a hybridoma, Cytotechnology 40 (1–3) (2002) 3–12, https://doi.org/
expression via a toll-like receptor-mediated pathway, Int. J. Biol. Macromol. 190 10.1023/A:1023993400608.
(2021) 607–617, https://doi.org/10.1016/j.ijbiomac.2021.09.021. [79] S. Terada, M. Sasaki, K. Yanagihara, H. Yamada, Preparation of silk protein
[55] Y. Li, Y. Huang, P. Pan, X. Che, Y. Zhang, Y. Zhang, A. Amal, X. Li, W. Niu, N. Luo, sericin as mitogenic factor for better mammalian cell culture, J. Biosci. Bioeng.
W. Zhang, D. Gao, Q. Tan, Q. Zhang, X. Xing, Z. Luo, J. Wu, Sericin and sericin- 100 (6) (2005) 667–671, https://doi.org/10.1263/jbb.100.667.
derived peptide alleviate viral pathogenesis in mice though inhibiting lactate [80] K. Thitinan, M. Pannamas, L.J.J.o.A.S. Natthanej, In-vitro characterization of silk
production and facilitating antiviral response, Appl. Mater. Today 25 (2021), sericin as an anti-aging agent, J. Agric. Sci. 5 (3) (2013) 13–28, https://doi.org/
https://doi.org/10.1016/j.apmt.2021.101256. 10.5539/jas.v5n3p54.
[56] W.H. Wang, W.S. Lin, C.H. Shih, C.Y. Chen, S.H. Kuo, W.L. Li, Y.S. Lin, [81] A. Ogawa, S. Terada, M. Miki, T. Kimura, A. Yamaguchi, M. Sasaki, H. Yamada,
Functionality of silk cocoon (Bombyx mori L.) sericin extracts obtained through Supplementation of Sericin Is Effective in Maintenance of Islet Survival and
high-temperature hydrothermal method, Materials 14 (18) (2021), https://doi. Function under Serum-free Culture, Springer Netherlands, Dordrecht, 2006,
org/10.3390/ma14185314. pp. 107–112, https://doi.org/10.1007/1-4020-4457-7_15.
[57] C. Bungthong, S. Siriamornpun, Changes in amino acid profiles and bioactive [82] A. Veiga, F. Castro, C.C. Reis, A. Sousa, A.L. Oliveira, F. Rocha, Hydroxyapatite/
compounds of Thai silk cocoons as affected by water extraction, Molecules 26 (7) sericin composites: a simple synthesis route under near-physiological conditions
(2021), https://doi.org/10.3390/molecules26072033. of temperature and pH and preliminary study of the effect of sericin on the
[58] F. Wang, T.T. Cao, Y.Q. Zhang, Effect of silk protein surfactant on silk degumming biomineralization process, Mater. Sci. Eng., C 108 (2020), 110400, https://doi.
and its properties, Mater. Sci. Eng., C 55 (2015) 131–136, https://doi.org/ org/10.1016/j.msec.2019.110400.
10.1016/j.msec.2015.05.041. [83] S. Dinescu, B. Galateanu, M. Albu, A. Cimpean, A. Dinischiotu, M. Costache,
[59] F. Wang, Y.Q. Zhang, Effects of alkyl polyglycoside (APG) on Bombyx mori silk Sericin enhances the bioperformance of collagen-based matrices preseeded with
degumming and the mechanical properties of silk fibroin fibre, Mater. Sci. Eng., C human-adipose derived stem cells (hADSCs), Int. J. Mol. Sci. 14 (1) (2013)
74 (2017) 152–158, https://doi.org/10.1016/j.msec.2017.02.015. 1870–1889, https://doi.org/10.3390/ijms14011870.
[60] S. Cherdchom, A. Sereemaspun, P. Aramwit, Urea-extracted sericin is potentially [84] S. Tyeb, P.A. Shiekh, V. Verma, A. Kumar, Adipose-derived stem cells (ADSCs)
better than kojic acid in the inhibition of melanogenesis through increased loaded gelatin-sericin-laminin cryogels for tissue regeneration in diabetic
reactive oxygen species generation, J. Tradit. Complement. Med. 11 (6) (2021) wounds, Biomacromolecules 21 (2) (2020) 294–304, https://doi.org/10.1021/
570–580, https://doi.org/10.1016/j.jtcme.2021.06.005. acs.biomac.9b01355.
[61] Y. Takasu, H. Yamada, K. Tsubouchi, Isolation of three main sericin components [85] S. Sapru, S. Das, M. Mandal, A.K. Ghosh, S.C. Kundu, Nonmulberry silk protein
from the cocoon of the silkworm, Bombyx mori, Biosci. Biotechnol. Biochem. 66 sericin blend hydrogels for skin tissue regeneration - in vitro and in vivo, Int. J.
(12) (2002) 2715–2718, https://doi.org/10.1271/bbb.66.2715. Biol. Macromol. 137 (2019) 545–553, https://doi.org/10.1016/j.
[62] M.M. Ali, S.B. Arumugam, Effect of crude extract of Bombyx mori coccoons in ijbiomac.2019.06.121.
hyperlipidemia and atherosclerosis, J. Ayurveda Integr. Med. 2 (2) (2011) 72–78, [86] N. Nagai, Y. Fukuoka, M. Ishii, H. Otake, T. Yamamoto, A. Taga, N. Okamoto,
https://doi.org/10.4103/0975-9476.82527. Y. Shimomura, Instillation of sericin enhances corneal wound healing through the
[63] G. Freddi, R. Mossotti, R. Innocenti, Degumming of silk fabric with several ERK pathway in rat debrided corneal epithelium, Int. J. Mol. Sci. 19 (4) (2018)
proteases, J. Biotechnol. 106 (1) (2003) 101–112, https://doi.org/10.1016/j. 1123, https://doi.org/10.3390/ijms19041123.
jbiotec.2003.09.006. [87] M. Ersel, Y. Uyanikgil, F. Karbek Akarca, E. Ozcete, Y.A. Altunci, F. Karabey,
[64] S.J. Eom, N.H. Lee, M.C. Kang, Y.H. Kim, T.G. Lim, K.M. Song, Silk peptide T. Cavusoglu, A. Meral, G. Yigitturk, E. Oyku Cetin, Effects of silk sericin on
production from whole silkworm cocoon using ultrasound and enzymatic incision wound healing in a dorsal skin flap wound healing rat model, Med. Sci.
treatment and its suppression of solar ultraviolet-induced skin inflammation, Mon. Int. Med. J. Exp. Clin. Res. 22 (2016) 1064–1078, https://doi.org/
Ultrason. Sonochem. 61 (2020), 104803, https://doi.org/10.1016/j. 10.12659/msm.897981.
ultsonch.2019.104803. [88] X. Dong, S.X. Zhao, X.L. Yin, H.Y. Wang, Z.G. Wei, Y.Q. Zhang, Silk sericin has
[65] L. Zhu, J. Lin, L. Pei, Y. Luo, D. Li, Z. Huang, Recent advances in environmentally significantly hypoglycaemic effect in type 2 diabetic mice via anti-oxidation and
friendly and green degumming processes of silk for textile and non-textile anti-inflammation, Int. J. Biol. Macromol. 150 (2020) 1061–1071, https://doi.
applications, Polymers 14 (4) (2022), https://doi.org/10.3390/polym14040659. org/10.1016/j.ijbiomac.2019.10.111.
[66] S. Franz, S. Rammelt, D. Scharnweber, J.C. Simon, Immune responses to implants [89] A. Kurioka, M. Yamazaki, Purification and identification of flavonoids from the
- a review of the implications for the design of immunomodulatory biomaterials, yellow green cocoon shell (Sasamayu) of the silkworm, Bombyx mori, Biosci.
Biomaterials 32 (28) (2011) 6692–6709, https://doi.org/10.1016/j. Biotechnol. Biochem. 66 (6) (2002) 1396–1399, https://doi.org/10.1271/
biomaterials.2011.05.078. bbb.66.1396.
[67] B.N. Brown, S.F. Badylak, Expanded applications, shifting paradigms and an [90] M. Schurink, W.J. van Berkel, H.J. Wichers, C.G. Boeriu, Novel peptides with
improved understanding of host-biomaterial interactions, Acta Biomater. 9 (2) tyrosinase inhibitory activity, Peptides 28 (3) (2007) 485–495, https://doi.org/
(2013) 4948–4955, https://doi.org/10.1016/j.actbio.2012.10.025. 10.1016/j.peptides.2006.11.023.
[68] C.G. Uragoda, P.N. Wijekoon, Asthma in silk workers, J. Soc. Occup. Med. 41 (3) [91] T. Chlapanidas, S. Farago, G. Lucconi, S. Perteghella, M. Galuzzi, M. Mantelli, M.
(1991) 140–142, https://doi.org/10.1093/occmed/41.3.140. A. Avanzini, M.C. Tosca, M. Marazzi, D. Vigo, M.L. Torre, M. Faustini, Sericins
[69] J.C. Celedon, L.J. Palmer, X. Xu, B. Wang, Z. Fang, S.T. Weiss, Sensitization to silk exhibit ROS-scavenging, anti-tyrosinase, anti-elastase, and in vitro
and childhood asthma in rural China, Pediatrics 107 (5) (2001) E80, https://doi. immunomodulatory activities, Int. J. Biol. Macromol. 58 (2013) 47–56, https://
org/10.1542/peds.107.5.e80. doi.org/10.1016/j.ijbiomac.2013.03.054.
[70] S. Das, G. Natarajan, Silk fiber composites in biomedical applications, Mater. [92] J.P. Kumar, B.B. Mandal, Antioxidant potential of mulberry and non-mulberry
Biomed. Eng. (2019) 309–338, https://doi.org/10.1016/B978-0-12-816872- silk sericin and its implications in biomedicine, Free Radic. Biol. Med. 108 (2017)
1.00011-X. 803–818, https://doi.org/10.1016/j.freeradbiomed.2017.05.002.
[71] D. Chouhan, B.B. Mandal, Silk biomaterials in wound healing and skin [93] H. Sies, Biochemie des oxidativen Stress, Angew. Chem. 98 (12) (1986)
regeneration therapeutics: from bench to bedside, Acta Biomater. 103 (2020) 1061–1075, https://doi.org/10.1002/ange.19860981203.
24–51, https://doi.org/10.1016/j.actbio.2019.11.050. [94] J. Kaur, R. Rajkhowa, T. Tsuzuki, K. Millington, J. Zhang, X. Wang,
[72] A.Z. Siavashani, J. Mohammadi, M. Rottmar, B. Senturk, J. Nourmohammadi, Photoprotection by silk cocoons, Biomacromolecules 14 (10) (2013) 3660–3667,
B. Sadeghi, L. Huber, K. Maniura-Weber, Silk fibroin/sericin 3D sponges: the https://doi.org/10.1021/bm401023h.
effect of sericin on structural and biological properties of fibroin, Int. J. Biol. [95] A. Rosena, T. Koobkokkruad, W. Eaknai, P. Bunwatcharaphansakun,
Macromol. 153 (2020) 317–326, https://doi.org/10.1016/j. R. Maniratanachote, S. Aueviriyavit, Protective effect of Thai silk extracts on
ijbiomac.2020.02.316. drug-induced phototoxicity in human epidermal A431 cells and a reconstructed
[73] B.O. Ode Boni, B.M. Bakadia, A.R. Osi, Z. Shi, H. Chen, M. Gauthier, G. Yang, human epidermis model, J. Photochem. Photobiol., B 188 (2018) 50–59, https://
Immune response to silk sericin-fibroin composites: potential immunogenic doi.org/10.1016/j.jphotobiol.2018.08.022.
elements and alternatives for immunomodulation, Macromol. Biosci. 22 (1) [96] T. Isobe, Y. Ikebata, T. Onitsuka, L.T. Do, Y. Sato, M. Taniguchi, T. Otoi,
(2022), e2100292, https://doi.org/10.1002/mabi.202100292. Cryopreservation for bovine embryos in serum-free freezing medium containing
[74] B.B. Mandal, A.S. Priya, S.C. Kundu, Novel silk sericin/gelatin 3-D scaffolds and silk protein sericin, Cryobiology 67 (2) (2013) 184–187, https://doi.org/
2-D films: fabrication and characterization for potential tissue engineering 10.1016/j.cryobiol.2013.06.010.

16
J. Liu et al. Biomaterials 287 (2022) 121638

[97] M. Hosoe, Y. Inaba, Y. Hashiyada, K. Imai, K. Kajitani, Y. Hasegawa, M. Irie, [119] B. Kundu, S.C. Kundu, Silk sericin/polyacrylamide in situ forming hydrogels for
H. Teramoto, T. Takahashi, S. Niimura, Effect of supplemented sericin on the dermal reconstruction, Biomaterials 33 (30) (2012) 7456–7467, https://doi.org/
development, cell number, cryosurvival and number of lipid droplets in cultured 10.1016/j.biomaterials.2012.06.091.
bovine embryos, Anim. Sci. J. 88 (2) (2017) 241–247, https://doi.org/10.1111/ [120] L. Shi, N. Yang, H. Zhang, L. Chen, L. Tao, Y. Wei, H. Liu, Y. Luo, A novel poly
asj.12628. (gamma-glutamic acid)/silk-sericin hydrogel for wound dressing: synthesis,
[98] P. Kumar, D. Kumar, P. Sikka, P. Singh, Sericin supplementation improves semen characterization and biological evaluation, Mater. Sci. Eng., C 48 (2015)
freezability of buffalo bulls by minimizing oxidative stress during 533–540, https://doi.org/10.1016/j.msec.2014.12.047.
cryopreservation, Anim. Reprod. Sci. 152 (2015) 26–31, https://doi.org/ [121] R. Xue, Y. Liu, Q. Zhang, C. Liang, H. Qin, P. Liu, K. Wang, X. Zhang, L. Chen,
10.1016/j.anireprosci.2014.11.015. Y. Wei, Shape changes and interaction mechanism of Escherichia coli cells treated
[99] T.T. Cao, Y.Q. Zhang, The potential of silk sericin protein as a serum substitute or with sericin and use of a sericin-based hydrogel for wound healing, Appl. Environ.
an additive in cell culture and cryopreservation, Amino Acids 49 (6) (2017) Microbiol. 82 (15) (2016) 4663–4672, https://doi.org/10.1128/AEM.00643-16.
1029–1039, https://doi.org/10.1007/s00726-017-2396-3. [122] C. Yang, R. Xue, Q. Zhang, S. Yang, P. Liu, L. Chen, K. Wang, X. Zhang, Y. Wei,
[100] F. Aghaz, M. Khazaei, A. Vaisi-Raygani, M. Bakhtiyari, Cryoprotective effect of Nanoclay cross-linked semi-IPN silk sericin/poly(NIPAm/LMSH) nanocomposite
sericin supplementation in freezing and thawing media on the outcome of hydrogel: an outstanding antibacterial wound dressing, Mater. Sci. Eng., C 81
cryopreservation in human sperm, Aging Male 23 (5) (2020) 469–476, https:// (2017) 303–313, https://doi.org/10.1016/j.msec.2017.08.008.
doi.org/10.1080/13685538.2018.1529156. [123] C. Qi, Y. Deng, L. Xu, C. Yang, Y. Zhu, G. Wang, Z. Wang, L. Wang, A sericin/
[101] C. Song, D. Liu, S. Yang, L. Cheng, E. Xing, Z. Chen, Sericin enhances the insulin- graphene oxide composite scaffold as a biomimetic extracellular matrix for
PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model, Exp. Ther. structural and functional repair of calvarial bone, Theranostics 10 (2) (2020)
Med. 16 (4) (2018) 3345–3352, https://doi.org/10.3892/etm.2018.6615. 741–756, https://doi.org/10.7150/thno.39502.
[102] R.I. Kunz, A.N. Capelassi, A.C.P. Alegre-Maller, M.L. Bonfleur, L.F.C. Ribeiro, R. [124] T. Siritienthong, J. Ratanavaraporn, P. Aramwit, Development of ethyl alcohol-
M. Costa, M.R.M. Natali, Sericin as treatment of obesity: morphophysiological precipitated silk sericin/polyvinyl alcohol scaffolds for accelerated healing of full-
effects in obese mice fed with high-fat diet, Einstein (Sao Paulo) 18 (2020), thickness wounds, Int. J. Pharm. 439 (1–2) (2012) 175–186, https://doi.org/
eAO4876, https://doi.org/10.31744/einstein_journal/2020AO4876. 10.1016/j.ijpharm.2012.09.043.
[103] M. Yang, N. Mandal, Y. Shuai, G. Zhou, S. Min, L. Zhu, Mineralization and [125] S. Dinescu, B. Galateanu, M. Albu, A. Lungu, E. Radu, A. Hermenean,
biocompatibility of Antheraea pernyi (A. pernyi) silk sericin film for potential M. Costache, Biocompatibility assessment of novel collagen-sericin scaffolds
bone tissue engineering, Bio Med. Mater. Eng. 24 (1) (2014) 815–824, https:// improved with hyaluronic Acid and chondroitin sulfate for cartilage regeneration,
doi.org/10.3233/BME-130873. BioMed Res. Int. 2013 (2013), 598056, https://doi.org/10.1155/2013/598056.
[104] O. Akturk, A. Tezcaner, H. Bilgili, M.S. Deveci, M.R. Gecit, D. Keskin, Evaluation [126] S. Nayak, S.C. Kundu, Sericin-carboxymethyl cellulose porous matrices as cellular
of sericin/collagen membranes as prospective wound dressing biomaterial, wound dressing material, J. Biomed. Mater. Res. 102 (6) (2014) 1928–1940,
J. Biosci. Bioeng. 112 (3) (2011) 279–288, https://doi.org/10.1016/j. https://doi.org/10.1002/jbm.a.34865.
jbiosc.2011.05.014. [127] M. Yang, Y. Shuai, C. Zhang, Y. Chen, L. Zhu, C. Mao, H. OuYang, Biomimetic
[105] L. Lamboni, Y. Li, J. Liu, G. Yang, Silk sericin-functionalized bacterial cellulose as nucleation of hydroxyapatite crystals mediated by Antheraea pernyi silk sericin
a potential wound-healing biomaterial, Biomacromolecules 17 (9) (2016) promotes osteogenic differentiation of human bone marrow derived
3076–3084, https://doi.org/10.1021/acs.biomac.6b00995. mesenchymal stem cells, Biomacromolecules 15 (4) (2014) 1185–1193, https://
[106] H.P. Zhang, X.Y. Wang, S.J. Min, M. Mandal, M.Y. Yang, L.J. Zhu, doi.org/10.1021/bm401740x.
Hydroxyapatite/sericin composite film prepared through mineralization of [128] Z. Jiayao, Z. Guanshan, Z. Jinchi, C. Yuyin, Z. Yongqiang, Antheraea pernyi silk
flexible ethanol-treated sericin film with simulated body fluids, Ceram. Int. 40 (1) sericin mediating biomimetic nucleation and growth of hydroxylapatite crystals
(2014) 985–991, https://doi.org/10.1016/j.ceramint.2013.06.095. promoting bone matrix formation, Microsc. Res. Tech. 80 (3) (2017) 305–311,
[107] R. Wang, J. Li, W. Chen, T. Xu, S. Yun, Z. Xu, Z. Xu, T. Sato, B. Chi, H. Xu, https://doi.org/10.1002/jemt.22793.
A biomimetic mussel-inspired ε-Poly-l-lysine hydrogel with robust tissue-anchor [129] P. Aramwit, J. Ratanavaraporn, S. T, Improvement of physical and wound
and anti-infection capacity, Adv. Funct. Mater. 27 (8) (2017), 1604894, https:// adhesion properties of silk sericin and polyvinyl alcohol dressing using glycerin,
doi.org/10.1002/adfm.201604894. Adv. Skin Wound Care 28 (8) (2015) 10, https://doi.org/10.1097/01.
[108] Y. Wang, R. Cai, G. Tao, P. Wang, H. Zuo, P. Zhao, A. Umar, H. He, A novel ASW.0000467304.77196.b9.
AgNPs/sericin/agar film with enhanced mechanical property and antibacterial [130] J. Wang, X. Li, Y. Song, Q. Su, X. Xiaohalati, W. Yang, L. Xu, B. Cai, G. Wang,
capability, Molecules 23 (7) (2018) 1821, https://doi.org/10.3390/ Z. Wang, L. Wang, Injectable silk sericin scaffolds with programmable shape-
molecules23071821. memory property and neuro-differentiation-promoting activity for individualized
[109] R. Cai, T. Gang, H. He, K. Song, H. Zuo, W. Jiang, Y.J.M. Wang, One-step synthesis brain repair of severe ischemic stroke, Bioact. Mater. 6 (7) (2021) 1988–1999,
of silver nanoparticles on polydopamine-coated sericin/polyvinyl alcohol https://doi.org/10.1016/j.bioactmat.2020.12.017.
composite films for potential antimicrobial applications, Molecules 22 (5) (2017) [131] T. Siritientong, A. Angspatt, J. Ratanavaraporn, P. Aramwit, Clinical potential of a
721, https://doi.org/10.3390/molecules22050721. silk sericin-releasing bioactive wound dressing for the treatment of split-thickness
[110] L. Liu, R. Cai, Y. Wang, G. Tao, L. Ai, P. Wang, M. Yang, H. Zuo, P. Zhao, H. He, skin graft donor sites, Pharm. Res. (N. Y.) 31 (1) (2014) 104–116, https://doi.org/
Polydopamine-assisted silver nanoparticle self-assembly on sericin/agar film for 10.1007/s11095-013-1136-y.
potential wound dressing application, Int. J. Mol. Sci. 19 (10) (2018) 2875, [132] S. Napavichayanun, R. Yamdech, P. Aramwit, The safety and efficacy of bacterial
https://doi.org/10.3390/ijms19102875. nanocellulose wound dressing incorporating sericin and polyhexamethylene
[111] W. Li, Z. Huang, R. Cai, W. Yang, H. He, Y. Wang, Rational design of Ag/ZnO biguanide: in vitro, in vivo and clinical studies, Arch. Dermatol. Res. 308 (2)
hybrid nanoparticles on sericin/agarose composite film for enhanced (2016) 123–132, https://doi.org/10.1007/s00403-016-1621-3.
antimicrobial applications, Int. J. Mol. Sci. 22 (1) (2020) 105, https://doi.org/ [133] Clinical trials. http://clinicaltrials.gov/ct2/results?term=sericin, 2022. (Accessed
10.3390/ijms22010105. 3 May 2022).
[112] P. Aramwit, S. Palapinyo, T. Srichana, S. Chottanapund, P. Muangman, Silk [134] B.J. Parker, D.I. Rhodes, C.M. O’Brien, A.E. Rodda, N.R. Cameron, Nerve
sericin ameliorates wound healing and its clinical efficacy in burn wounds, Arch. guidance conduit development for primary treatment of peripheral nerve
Dermatol. Res. 305 (7) (2013) 585–594, https://doi.org/10.1007/s00403-013- transection injuries: a commercial perspective, Acta Biomater. 135 (3) (2021)
1371-4. 64–86, https://doi.org/10.1016/j.actbio.2021.08.052.
[113] P. Aramwit, O. Keongamaroon, T. Siritientong, N. Bang, O. Supasyndh, Sericin [135] M. Lin, Z. Zheng, L. Yang, M. Luo, L. Fu, B. Lin, C. Xu, A high-performance,
cream reduces pruritus in hemodialysis patients: a randomized, double-blind, sensitive, wearable multifunctional sensor based on rubber/CNT for human
placebo-controlled experimental study, BMC Nephrol. 13 (2012) 119, https://doi. motion and skin temperature detection, Adv. Mater. 34 (1) (2022), e2107309,
org/10.1186/1471-2369-13-119. https://doi.org/10.1002/adma.202107309.
[114] J.W. Kim, Y.Y. Jo, J.Y. Kim, J.h. Oh, B.E. Yang, S.G. Kim, Clinical study for silk [136] X. Liang, H. Li, J. Dou, Q. Wang, W. He, C. Wang, D. Li, J.M. Lin, Y. Zhang, Stable
mat application into extraction socket: a split-mouth, randomized clinical trial, and biocompatible carbon nanotube ink mediated by silk protein for printed
Appl. Sci. 9 (6) (2019), https://doi.org/10.3390/app9061208. electronics, Adv. Mater. 32 (31) (2020), e2000165, https://doi.org/10.1002/
[115] Y. Song, C. Zhang, J. Zhang, N. Sun, K. Huang, H. Li, Z. Wang, K. Huang, L. Wang, adma.202000165.
An injectable silk sericin hydrogel promotes cardiac functional recovery after [137] H. Ma, J. Li, J. Zhou, Q. Luo, W. Wu, Z. Mao, W. Ma, Screen-printed carbon black/
ischemic myocardial infarction, Acta Biomater. 41 (2016) 210–223, https://doi. recycled Sericin@Fabrics for wearable sensors to monitor sweat loss, ACS Appl.
org/10.1016/j.actbio.2016.05.039. Mater. Interfaces 14 (9) (2022) 11813–11819, https://doi.org/10.1021/
[116] M. Yang, Y. Wang, T. Gang, R. Cai, W. Peng, L. Liu, L. Ai, H. Zuo, P. Zhao, U.J. acsami.1c23341.
N. Ahmad, Fabrication of sericin/agrose gel loaded lysozyme and its potential in [138] Y. An, C. Luo, D. Yao, S. Wen, P. Zheng, S. Chi, Y. Yang, J. Chang, Y. Deng,
wound dressing application, Nanomaterials 8 (4) (2018) 235, https://doi.org/ C. Wang, Natural cocoons enabling flexible and stable fabric lithium-sulfur full
10.3390/nano8040235. batteries, Nano-Micro Lett. 13 (1) (2021) 84, https://doi.org/10.1007/s40820-
[117] S. Sapru, A.K. Ghosh, S.C. Kundu, Non-immunogenic, porous and antibacterial 021-00609-3.
chitosan and Antheraea mylitta silk sericin hydrogels as potential dermal [139] Q. Duan, Y. Lu, Silk sericin as a green adhesive to fabricate a textile strain sensor
substitute, Carbohydr. Polym. 167 (2017) 196–209, https://doi.org/10.1016/j. with excellent electromagnetic shielding performance, ACS Appl. Mater.
carbpol.2017.02.098. Interfaces 13 (24) (2021) 28832–28842, https://doi.org/10.1021/
[118] Y. Zhang, J. Liu, L. Huang, Z. Wang, L. Wang, Design and performance of a acsami.1c05671.
sericin-alginate interpenetrating network hydrogel for cell and drug delivery, Sci. [140] H. Wang, F. Meng, Y. Cai, L. Zheng, Y. Li, Y. Liu, Y. Jiang, X. Wang, X. Chen,
Rep. 5 (2015), 12374, https://doi.org/10.1038/srep12374. Sericin for resistance switching device with multilevel nonvolatile memory, Adv.
Mater. 25 (38) (2013) 5498–5503, https://doi.org/10.1002/adma.201301983.

17
J. Liu et al. Biomaterials 287 (2022) 121638

[141] H.P. Felgueiras, M.T.P. Amorim, Functionalization of electrospun polymeric [165] J. Liu, Q.L. Li, J.X. Zhang, L. Huang, C. Qi, L.M. Xu, X.X. Liu, G.B. Wang, L. Wang,
wound dressings with antimicrobial peptides, Colloids Surf., B 156 (2017) Z. Wang, Safe and effective reversal of cancer multidrug resistance using sericin-
133–148, https://doi.org/10.1016/j.colsurfb.2017.05.001. coated mesoporous silica nanoparticles for lysosome-targeting delivery in mice,
[142] D. Chouhan, N. Dey, N. Bhardwaj, B.B. Mandal, Emerging and innovative Small 13 (9) (2017), 1602567, https://doi.org/10.1002/smll.201602567.
approaches for wound healing and skin regeneration: current status and [166] J. Kanoujia, M. Singh, P. Singh, S.A. Saraf, Novel genipin crosslinked atorvastatin
advances, Biomaterials 216 (2019), 119267, https://doi.org/10.1016/j. loaded sericin nanoparticles for their enhanced antihyperlipidemic activity,
biomaterials.2019.119267. Mater. Sci. Eng., C 69 (2016) 967–976, https://doi.org/10.1016/j.
[143] Y.J. Son, J.W. Tse, Y. Zhou, W. Mao, E.K.F. Yim, H.S. Yoo, Biomaterials and msec.2016.08.011.
controlled release strategy for epithelial wound healing, Biomater. Sci. 7 (11) [167] K. Suktham, T. Koobkokkruad, T. Wutikhun, S. Surassmo, Efficiency of
(2019) 4444–4471, https://doi.org/10.1039/c9bm00456d. resveratrol-loaded sericin nanoparticles: promising bionanocarriers for drug
[144] R. Wang, J. Li, W. Chen, T. Xu, S. Yun, Z. Xu, Z. Xu, T. Sato, B. Chi, H. Xu, delivery, Int. J. Pharm. 537 (1–2) (2018) 48–56, https://doi.org/10.1016/j.
A biomimetic mussel-inspired ε-Poly-l-lysine hydrogel with robust tissue-anchor ijpharm.2017.12.015.
and anti-infection capacity, Adv. Funct. Mater. 27 (8) (2017), https://doi.org/ [168] O.I. Parisi, M. Fiorillo, L. Scrivano, M.S. Sinicropi, V. Dolce, D. Iacopetta, F. Puoci,
10.1002/adfm.201604894. A.R. Cappello, Sericin/Poly(ethylcyanoacrylate) nanospheres by interfacial
[145] R. Cai, G. Tao, H. He, K. Song, H. Zuo, W. Jiang, Y. Wang, One-step synthesis of polymerization for enhanced bioefficacy of fenofibrate: in vitro and in vivo
silver nanoparticles on polydopamine-coated sericin/polyvinyl alcohol composite studies, Biomacromolecules 16 (10) (2015) 3126–3133, https://doi.org/
films for potential antimicrobial applications, Molecules 22 (5) (2017), https:// 10.1021/acs.biomac.5b00746.
doi.org/10.3390/molecules22050721. [169] W. Guo, L. Deng, J. Yu, Z. Chen, Y. Woo, H. Liu, T. Li, T. Lin, H. Chen, M. Zhao,
[146] L. Liu, R. Cai, Y. Wang, G. Tao, L. Ai, P. Wang, M. Yang, H. Zuo, P. Zhao, H. He, L. Zhang, G. Li, Y. Hu, Sericin nanomicelles with enhanced cellular uptake and
Polydopamine-assisted silver nanoparticle self-assembly on sericin/agar film for pH-triggered release of doxorubicin reverse cancer drug resistance, Drug Deliv. 25
potential wound dressing application, Int. J. Mol. Sci. 19 (10) (2018), https://doi. (1) (2018) 1103–1116, https://doi.org/10.1080/10717544.2018.1469686.
org/10.3390/ijms19102875. [170] W. Guo, L. Deng, Z. Chen, Z. Chen, J. Yu, H. Liu, T. Li, T. Lin, H. Chen, M. Zhao,
[147] T.E. Brown, K.S. Anseth, Spatiotemporal hydrogel biomaterials for regenerative L. Zhang, G. Li, Y. Hu, Vitamin B12-conjugated sericin micelles for targeting
medicine, Chem. Soc. Rev. 46 (21) (2017) 6532–6552, https://doi.org/10.1039/ CD320-overexpressed gastric cancer and reversing drug resistance, Nanomedicine
c7cs00445a. 14 (3) (2019) 353–370, https://doi.org/10.2217/nnm-2018-0321.
[148] S. Jasmine, B.B. Mandal, Types and properties of non-mulberry silk biomaterials [171] K. Boonpavanitchakul, L.K. Bast, N. Bruns, R. Magaraphan, Silk sericin-
for tissue engineering applications, Silk Biomater. Tissue Eng. Regen. Med. (2014) polylactide protein-polymer conjugates as biodegradable amphiphilic materials
275–298, https://doi.org/10.1533/9780857097064.2.275. and their application in drug release systems, Bioconjugate Chem. 31 (10) (2020)
[149] J.J. Zhao, D.C. Liu, Y.H. Yu, H. Tang, Development of gelatin-silk sericin 2312–2324, https://doi.org/10.1021/acs.bioconjchem.0c00399.
incorporated with poly(vinyl alcohol) hydrogel-based nanocomposite for [172] L. Huang, K. Tao, J. Liu, C. Qi, L. Xu, P. Chang, J. Gao, X. Shuai, G. Wang,
articular cartilage defects in rat knee Joint repair, J. Biomed. Nanotechnol. 17 Z. Wang, L. Wang, Design and fabrication of multifunctional sericin nanoparticles
(2021) 242–252, https://doi.org/10.1166/jbn.2021.3024. for tumor targeting and pH-responsive subcellular delivery of cancer
[150] M. Yang, Y. Wang, G. Tao, R. Cai, P. Wang, L. Liu, L. Ai, H. Zuo, P. Zhao, A. Umar, chemotherapy drugs, ACS Appl. Mater. Interfaces 8 (10) (2016) 6577–6585,
C. Mao, H. He, Fabrication of sericin/agrose gel loaded lysozyme and its potential https://doi.org/10.1021/acsami.5b11617.
in wound dressing application, Nanomaterials 8 (4) (2018), https://doi.org/ [173] H. He, G. Tao, Y. Wang, R. Cai, P. Guo, L. Chen, H. Zuo, P. Zhao, Q. Xia, In situ
10.3390/nano8040235. green synthesis and characterization of sericin-silver nanoparticle composite with
[151] G. Tao, R. Cai, Y. Wang, H. Zuo, H. He, Fabrication of antibacterial sericin based effective antibacterial activity and good biocompatibility, Mater. Sci. Eng., C 80
hydrogel as an injectable and mouldable wound dressing, Mater. Sci. Eng., C 119 (2017) 509–516, https://doi.org/10.1016/j.msec.2017.06.015.
(2021), 111597, https://doi.org/10.1016/j.msec.2020.111597. [174] J. Wang, S. Yang, C. Li, Y. Miao, L. Zhu, C. Mao, M. Yang, Nucleation and
[152] M. Zhang, D. Wang, N. Ji, S. Lee, G. Wang, Y. Zheng, X. Zhang, L. Yang, Z. Qin, assembly of silica into protein-based nanocomposites as effective anticancer drug
Y. Yang, Bioinspired design of sericin/chitosan/Ag@MOF/GO hydrogels for carriers using self-assembled silk protein nanostructures as biotemplates, ACS
efficiently combating resistant bacteria, rapid hemostasis, and wound healing, Appl. Mater. Interfaces 9 (27) (2017) 22259–22267, https://doi.org/10.1021/
Polymers (Basel) 13 (16) (2021) 2812, https://doi.org/10.3390/polym13162812. acsami.7b05664.
[153] Y. Zhang, R. Jiang, A. Fang, Y. Zhao, T. Wu, X. Cao, P. Liang, D. Xia, G. Zhang, [175] W. Chaisabai, A. Khamhaengpol, S. Siri, Sericins of mulberry and non-mulberry
A highly transparent, elastic, injectable sericin hydrogel induced by ultrasound, silkworms for eco-friendly synthesis of silver nanoparticles, Artif. Cell Nanomed.
Polym. Test. 77 (2019), 105890, https://doi.org/10.1016/j. Biotechnol. 46 (3) (2018) 536–543, https://doi.org/10.1080/
polymertesting.2019.05.006. 21691401.2017.1328686.
[154] Y. Zhu, H. Liu, S. Qin, C. Yang, Q. Lv, Z. Wang, L. Wang, Antibacterial sericin [176] D. Tian, H. Xu, B. Xiao, X. Zhou, X. Liu, Z. Zhou, H.K. Patra, N. Slater, J. Tang,
cryogels promote hemostasis by facilitating the activation of coagulation pathway Y. Shen, Single-step formulation of levodopa-based nanotheranostics - strategy for
and platelets, Adv. Healthc. Mater. (2022), e2102717, https://doi.org/10.1002/ ultra-sensitive high longitudinal relaxivity MRI guided switchable therapeutics,
adhm.202102717. Biomater. Sci. 8 (6) (2020) 1615–1621, https://doi.org/10.1039/c9bm01799b.
[155] S. Abdulghani, G.R. Mitchell, Biomaterials for in situ tissue regeneration: a [177] O. Akturk, Z. Gun Gok, O. Erdemli, M. Yigitoglu, One-pot facile synthesis of silk
review, Biomolecules 9 (11) (2019) 750, https://doi.org/10.3390/biom9110750. sericin-capped gold nanoparticles by UVC radiation: investigation of stability,
[156] W.H. De Jong, P.J. Borm, Drug delivery and nanoparticles:applications and biocompatibility, and antibacterial activity, J. Biomed. Mater. Res. 107 (12)
hazards, Int. J. Nanomed. 3 (2) (2008) 133–149, https://doi.org/10.2147/ijn. (2019) 2667–2679, https://doi.org/10.1002/jbm.a.36771.
s596. [178] L. Deng, W. Guo, G. Li, Y. Hu, L.M. Zhang, Hydrophobic IR780 loaded sericin
[157] M.J. Mitchell, M.M. Billingsley, R.M. Haley, M.E. Wechsler, N.A. Peppas, nanomicelles for phototherapy with enhanced antitumor efficiency, Int. J. Pharm.
R. Langer, Engineering precision nanoparticles for drug delivery, Nat. Rev. Drug 566 (2019) 549–556, https://doi.org/10.1016/j.ijpharm.2019.05.075.
Discov. 20 (2) (2021) 101–124, https://doi.org/10.1038/s41573-020-0090-8. [179] M. Arjama, S. Mehnath, M. Rajan, M. Jeyaraj, Sericin/RBA embedded gellan gum
[158] A.A. Saei, M. Yazdani, S.E. Lohse, Z. Bakhtiary, V. Serpooshan, M. Ghavami, based smart nanosystem for pH responsive drug delivery, Int. J. Biol. Macromol.
M. Asadian, S. Mashaghi, E.C. Dreaden, A. Mashaghi, M. Mahmoudi, Nanoparticle 120 (Pt B) (2018) 1561–1571, https://doi.org/10.1016/j.ijbiomac.2018.09.146.
surface functionality dictates cellular and systemic toxicity, Chem. Mater. 29 (16) [180] S. Mehnath, M.A. Ayisha Sithika, M. Arjama, M. Rajan, R. Amarnath Praphakar,
(2017) 6578–6595, https://doi.org/10.1021/acs.chemmater.7b01979. M. Jeyaraj, Sericin-chitosan doped maleate gellan gum nanocomposites for
[159] J. Lazarovits, Y.Y. Chen, E.A. Sykes, W.C. Chan, Nanoparticle-blood interactions: effective cell damage in Mycobacterium tuberculosis, Int. J. Biol. Macromol. 122
the implications on solid tumour targeting, Chem. Commun. 51 (14) (2015) (2019) 174–184, https://doi.org/10.1016/j.ijbiomac.2018.10.167.
2756–2767, https://doi.org/10.1039/c4cc07644c. [181] E. Hadipour-Goudarzi, M. Montazer, M. Latifi, A.A. Aghaji, Electrospinning of
[160] T. Limongi, M. Canta, L. Racca, A. Ancona, S. Tritta, V. Vighetto, V. Cauda, chitosan/sericin/PVA nanofibers incorporated with in situ synthesis of nano
Improving dispersal of therapeutic nanoparticles in the human body, silver, Carbohydr. Polym. 113 (2014) 231–239, https://doi.org/10.1016/j.
Nanomedicine 14 (7) (2019) 797–801, https://doi.org/10.2217/nnm-2019-0070. carbpol.2014.06.082.
[161] D. Zhang, J. Qiu, L. Shi, Y. Liu, B. Pan, B. Xing, The mechanisms and [182] S. Doakhan, M. Montazer, A. Rashidi, R. Moniri, M.B. Moghadam, Influence of
environmental implications of engineered nanoparticles dispersion, Sci. Total sericin/TiO(2) nanocomposite on cotton fabric: part 1. Enhanced antibacterial
Environ. 722 (2020), 137781, https://doi.org/10.1016/j.scitotenv.2020.137781. effect, Carbohydr. Polym. 94 (2) (2013) 737–748, https://doi.org/10.1016/j.
[162] M. Jahanshahi, E. Kowsari, V. Haddadi-Asl, M. Khoobi, J.H. Lee, F.B. Kadumudi, carbpol.2013.01.023.
S. Talebian, N. Kamaly, M. Mehrali, Sericin grafted multifunctional curcumin [183] T. Khampieng, P. Aramwit, P. Supaphol, Silk sericin loaded alginate
loaded fluorinated graphene oxide nanomedicines with charge switching nanoparticles: preparation and anti-inflammatory efficacy, Int. J. Biol. Macromol.
properties for effective cancer cell targeting, Int. J. Pharm. 572 (2019), 118791, 80 (2015) 636–643, https://doi.org/10.1016/j.ijbiomac.2015.07.018.
https://doi.org/10.1016/j.ijpharm.2019.118791. [184] N. Nagai, Y. Iwai, S. Deguchi, H. Otake, K. Kanai, N. Okamoto, Y. Shimomura,
[163] D. Hu, T. Li, Z. Xu, D. Liu, M. Yang, L. Zhu, Self-stabilized silk sericin-based Therapeutic potential of a combination of magnesium hydroxide nanoparticles
nanoparticles: in vivo biocompatibility and reduced doxorubicin-induced toxicity, and sericin for epithelial corneal wound healing, Nanomaterials 9 (5) (2019) 768,
Acta Biomater. 74 (2018) 385–396, https://doi.org/10.1016/j. https://doi.org/10.3390/nano9050768.
actbio.2018.05.024. [185] T. Chlapanidas, S. Perteghella, F. Leoni, S. Farago, M. Marazzi, D. Rossi,
[164] X. Li, S. Hou, J. Chen, C.E. He, Y.E. Gao, Y. Lu, D. Jia, X. Ma, P. Xue, Y. Kang, E. Martino, R. Gaggeri, S. Collina, TNF-alpha blocker effect of naringenin-loaded
Z. Xu, Engineering silk sericin decorated zeolitic imidazolate framework-8 sericin microparticles that are potentially useful in the treatment of psoriasis, Int.
nanoplatform to enhance chemotherapy, Colloids Surf., B 200 (2021), 111594, J. Mol. Sci. 15 (8) (2014) 13624–13636, https://doi.org/10.3390/
https://doi.org/10.1016/j.colsurfb.2021.111594. ijms150813624.

18
J. Liu et al. Biomaterials 287 (2022) 121638

[186] Z. Liu, Y. Cai, Y. Jia, L. Liu, X. Kong, S.C. Kundu, J. Yao, One-step synthesis of [190] N.E. Kurland, T. Dey, C. Wang, S.C. Kundu, V.K. Yadavalli, Silk protein
natural silk sericin-based microcapsules with bionic structures, Macromol. Rapid lithography as a route to fabricate sericin microarchitectures, Adv. Mater. 26 (26)
Commun. 35 (19) (2014) 1668–1672, https://doi.org/10.1002/marc.201400304. (2014) 4431–4437, https://doi.org/10.1002/adma.201400777.
[187] E. Bari, C.R. Arciola, B. Vigani, B. Crivelli, P. Moro, G. Marrubini, M. Sorrenti, [191] S. Nayak, S. Dey, S.C. Kundu, Silk sericin-alginate-chitosan microcapsules:
L. Catenacci, G. Bruni, T. Chlapanidas, E. Lucarelli, S. Perteghella, M.L. Torre, In hepatocytes encapsulation for enhanced cellular functions, Int. J. Biol. Macromol.
vitro effectiveness of microspheres based on silk sericin and chlorella vulgaris or 65 (2014) 258–266, https://doi.org/10.1016/j.ijbiomac.2014.01.042.
arthrospira platensis for wound healing applications, Materials 10 (9) (2017) 983, [192] C.S. Chen, F. Zeng, X. Xiao, Z. Wang, X.L. Li, R.W. Tan, W.Q. Liu, Y.S. Zhang, Z.
https://doi.org/10.3390/ma10090983. D. She, S.J. Li, Three-dimensionally printed silk-sericin-based hydrogel scaffold: a
[188] W. Li, X. Su, Q. Zhong, Z. Liu, Y. Cai, J. Yao, Influence of reaction conditions on promising visualized dressing material for real-time monitoring of wounds, ACS
the self-assembly of the natural silk sericin protein, Microsc. Res. Tech. 80 (3) Appl. Mater. Interfaces 10 (40) (2018) 33879–33890, https://doi.org/10.1021/
(2017) 298–304, https://doi.org/10.1002/jemt.22666. acsami.8b10072.
[189] J. Liu, Y. Deng, D. Fu, Y. Yuan, Q. Li, L. Shi, G. Wang, Z. Wang, L. Wang, Sericin [193] Y. Deng, C. Yang, Y. Zhu, W. Liu, H. Li, L. Wang, W. Chen, Z. Wang, L. Wang,
microparticles enveloped with metal-organic networks as a pulmonary targeting Lamprey-teeth-inspired oriented antibacterial sericin microneedles for infected
delivery system for intra-tracheally treating metastatic lung cancer, Bioact. Mater. wound healing improvement, Nano Lett. 22 (7) (2022) 2702–2711, https://doi.
6 (1) (2021) 273–284, https://doi.org/10.1016/j.bioactmat.2020.08.006. org/10.1021/acs.nanolett.1c04573.

19

You might also like