Next Article in Journal
Pharmaceutical Potential of Remedial Plants and Helminths for Treating Inflammatory Bowel Disease
Previous Article in Journal
Critical Appraisal and Future Challenges of Artificial Intelligence and Anticancer Drug Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Silkworm Cocoon: Dual Functions as a Traditional Chinese Medicine and the Raw Material of Promising Biocompatible Carriers

by
Zhijie Tian
1,2,†,
Chuncao Zhao
2,†,
Ting Huang
2,
Lining Yu
2,
Yijie Sun
3,
Yian Tao
2,
Yunfeng Cao
2,
Ruofei Du
4,
Wenhui Lin
1,* and
Jia Zeng
2,*
1
School of Chemistry & Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
2
NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 200032, China
3
Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
4
Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2024, 17(7), 817; https://doi.org/10.3390/ph17070817
Submission received: 31 May 2024 / Revised: 17 June 2024 / Accepted: 19 June 2024 / Published: 21 June 2024
(This article belongs to the Section Natural Products)

Abstract

:
The silkworm cocoon (SC), both as a traditional Chinese medicine and as the raw material for biocompatible carriers, has been extensively used in the medical and biomedical fields. This review elaborates on the multiple functions of SC, with an in-depth analysis of its chemical composition, biological activities, as well as its applications in modern medicine. The primary chemical components of SC include silk fibroin (SF), silk sericin (SS), and other flavonoid-like bioactive compounds demonstrating various biological effects. These include hypoglycemic, cardioprotective, hypolipidemic, anti-inflammatory, antioxidant, and antimicrobial actions, which highlight its potential therapeutic benefits. Furthermore, the review explores the applications of silk-derived materials in drug delivery systems, tissue engineering, regenerative medicine, and in vitro diagnostics. It also highlights the progression of SC from laboratory research to clinical trials, emphasizing the safety and efficacy of SC-based materials across multiple medical domains. Moreover, we discuss the market products developed from silk proteins, illustrating the transition from traditional uses to contemporary medical applications. This review provides support in understanding the current research status of SC and the further development and application of its derived products.

1. Introduction

The silkworm cocoon (SC) is the dry shell of Bombyx mori L., which belongs to the Saturniidae family. SC is widely used in the silk industry, agriculture, biological materials, medicine, and other fields. In medicinal applications, SC was first mentioned in the famous medical book Compendium of Materia Medica and has been used as a traditional medicine for hundreds of years in China for its diverse therapeutic effects [1,2,3]. SC has pharmacological effects to collect astringency, stop bleeding, quench thirst, and detoxify boils. Hence, it is used for the treatment of hematemesis, bloody stools, metrorrhagia, excessive urination, and the pathogenesis of carbuncle and pus.
Modern research indicates SC contains major chemical components such as silk sericin (SS) and silk fibroin (SF) [4,5]. These components endow SC with significant biological activities, not only enhancing the traditional health benefits, but also providing foundational properties for innovative therapeutic techniques. The therapeutic actions of SS and SF are extensive, including antidiabetic effects [6,7], cardioprotective properties [8,9,10], and lipid-lowering capabilities [11]. Concurrently, the ability to modulate inflammatory responses [12,13,14] and resist oxidative stress [15,16,17,18] broadens their application in disease prevention and treatment. Furthermore, the inherent antimicrobial [4,19,20] and antiviral [21] properties of these proteins enhance their effectiveness in developing cutting-edge medical interventions.
With advancements in biotechnology, the application of SC has expanded into the modern biomedical field, and SC has been proven to be a promising biocompatible carrier [22,23]. Therefore, it is considered not only to be a key component of traditional medicine but also a crucial material in contemporary medical innovation. Presently, its applications primarily manifest in areas such as drug delivery systems [24,25,26], tissue engineering [27,28,29,30], and regenerative medicine [30,31]. These studies leverage the biodegradability and excellent biocompatibility of SC to develop new medical materials and treatment methods, showcasing its potential as a natural resource in the contemporary medical domain.
Considering these developments, in this study, we deeply examine the dual functions of SC in both laboratory research and clinical studies by providing an in-depth analysis of the chemical composition and biological activity of SC and highlighting its emerging applications in biomedicine. The aim of this review is to explore how SC can be cleverly redesigned for future medical science.

2. Chemical Composition

The chemical composition of SC primarily consists of SF and SS. SF forms the structural framework of SC, and SS acts as an adhesive that bonds the fibrous components within the SC together. In addition to these primary components, SC contains bioactive substances such as flavonoids, peptides, and calcium oxalate crystals, as shown in Table 1.

2.1. Silk Fibroin

SF is the principal protein, and approximately 70% the amount of SC is characterized by a hierarchical structure of a light chain (approximately 26 kDa) and a heavy chain (approximately 350 kDa) bonded through disulfide links. The primary structure of the heavy chain consists of 12 large repeating hydrophobic domains of amino acids, such as glycine, alanine, serine, valine, and tyrosine, separated by 11 short hydrophilic regions, while the light chain features hydrophilic amino acids, such as glutamic acid, lysine, and aspartic acid [5,32]. Approximately 5.3% of SF is composed of tyrosine, while 0.2% of SF is lysine residues. Both have functional groups that can react with epoxides. The ability of SF to form β-sheet structures via physical cross-linking allows the silk materials to exhibit a range of multifunctional properties that can be finely adjusted [33]. Owing to its low immunogenicity and fibrous structure resembling collagen I, SF is one of the most attractive natural biomaterials for biomedical applications [34].

2.2. Silk Sericin

SS, which accounts for approximately 30% of the SC, is a glycoprotein adhesive containing 18 different amino acids primarily organized in the form of polar side chains, such as hydroxyl, acidic, and basic amino acids, SS forms three primary peptide complexes with molecular weights of 150, 180–250, and 400 kDa. These functional groups enable SS to easily cross-link, blend, and copolymerize with other polymers, thereby producing improved biocompatible materials with enhanced properties [35,36].

2.3. Flavonoids

Flavonoids exist in the cocoons of different silkworm species. Seven flavonoid compounds were reportedly isolated, purified, and identified from yellow–green SC, and their aglycones were isolated and purified from the ethanol extract of SC [37]. The presence of flavonoids in the SC helps to provide antioxidant protection to the SC itself [38].

2.4. Other Components

Many types of outdoor-reared SC contain abundant calcium oxalate monohydrate crystals, primarily comprising oxalic acid and calcium ions, with a chemical formula of CaC2O4·H2O [39]. The formation of these crystals may be related to the metabolic substances and environmental conditions within the silkworm, making them a common component of the SC [40].
In addition to primary proteins like SF and SS, SC is composed of peptides with different sequences and functions. These peptides are small molecular protein fragments composed of amino acids and are mainly found in the degradation products of SF or SS, which could play roles in regulating biological processes within the silk gland, contributing to the microenvironment of the SF and ensuring the silkworm’s development and transformation into a moth [41].

3. Biological Activities

SC exhibits a wide range of pharmacological effects, such as lowering blood glucose and lipid levels, heart protection, anti-inflammatory, antioxidative, and anti-infection properties. Moreover, it shows potential in cancer therapy. Its unique chemical composition and elastic properties facilitate wound healing and provide pain relief, making it highly applicable in the medical field. The biological activities and mechanisms of SC are summarized in Table 2.

3.1. Hypoglycemic Action

In traditional medicine, SC is known as an antidiabetic agent that was widely used in ancient formulas. Modern medicine has also confirmed that the primary components of SC are ideal candidates for the prevention and treatment of diabetes. SF, SS, peptides, or their hydrolysates present α-glucosidase inhibitory activity. Additionally, other hypoglycemic mechanisms of these compounds include the inhibition of the expression of intestinal glucose transporters, the promotion of the regeneration of pancreatic β-cells, and the activation of the insulin-like signaling pathway [42]. The ethanolic extract from the green cocoon layer of the silkworm has been demonstrated to improve glucose metabolism by reducing inflammatory reactions, enhancing the antioxidant capacity and insulin sensitivity, and regulating the balance between glycolysis and gluconeogenesis [43].
Diabetes can also cause many disorders such as nephropathy, neuropathy, and retinopathy. SC has significant therapeutic effects on a variety of complications caused by diabetes. The ethanolic extract is rich in quercetin and kaempferol, and the glucosides from SC reduced blood glucose levels and improved body weights in diabetic mice, and it may have a potential therapeutic application in the treatment of diabetic nephropathy. The ethanolic extract inhibits the expressions of renal tumor necrosis factor-alpha, monocyte chemoattractant protein-1, fibronectin, and P38 mitogen-activated protein kinase. Furthermore, it significantly elevates the levels of superoxide dismutase and glutathione peroxidase in diabetic mice [39].
There is also evidence that diabetes affects men’s sexual function through changes in the process of spermatogenesis or the endocrine changes affecting it; it affects erectile dysfunction and decreases libido, it can lessen sperm DNA damage, and it affects the hypothalamic–pituitary–gonadal axis [44]. A recent study suggested that a hydroalcoholic extract of SC decreased the effects of diabetes on the hypothalamic–pituitary–gonadal axis. It increased the level of sex hormone secretion in a dose-dependent manner in diabetic rats by affecting the activity of the pituitary–gonadal axis. Additionally, the dose-dependent administration of the aqueous extract of SC can improve testicular tissue injuries in type I diabetic rats, thus protecting fertility [45]. These findings provide evidence of SC as a potential therapeutic approach for the treatment of diabetes and insulin resistance.

3.2. Cardioprotective Effect

In terms of cardiac protection, SS plays a critical role in myocardial injury, as the protective properties of amino acids have just been recognized as the fundamental reason for enhancing cardiac protection [46]. In an isoproterenol-induced rat model, SS showed a decrease in edema, the dilation of capillaries, scar formation, and yellowing of the heart.
The intraventricular septum thickness, right ventricular wall thickness, and left ventricular wall thickness decreased dramatically after SS treatment. SS also significantly increased the non-enzymatic antioxidant markers in the serum and heart tissue, including glutathione, vitamin E, and vitamin C. The results were the same for the enzymatic antioxidant marker, mitochondrial enzymes, and protein. SS reduced the synthesis of collagen in myocardiocytes, thus reducing the incidence of fibrosis, and the synthesis of collagens. In summary, it showed potent cardio-protective properties by decreasing inflammatory reactions and oxidative stress, thereby leading to improved myocardial activity and reduced cardiac damage after myocardial ischemia [9].
Mitochondrial abnormalities in myocardial cells are one of the causes of heart failure [47]. SS might improve the dysmorphic mitochondrial structure, metabolism, and energy production of cardiac mitochondria under hypercholesterolaemic conditions. The mechanisms included the upregulation of optic atrophy 1 and the reduction of NADH–ubiquinone oxidoreductase 75-kDa subunit expression, as well as an improvement in mitochondrial energy production by upregulating acetyl-CoA acetyltransferase and the NADH dehydrogenase 1a subcomplex subunit 10 expression [48].
Furthermore, the cardioprotective activity of the ethanolic extract of SC has also been revealed. It significantly prevented isoproterenol-induced myocardial damage and hypertrophy and decreased the levels of the myocardial enzyme markers [8]. The cardioprotective effect of a formulation combined with the methanol extract of SC, flaxseed oil, and coenzyme Q10 against doxorubicin induced myocardial toxicity, and this was evaluated in rats. The test formulation lowered the increase in heart weight due to hypertrophy while significantly reducing the serum levels of aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, creatinine, triglyceride, low-density lipoprotein cholesterol, and very-low-density lipoprotein cholesterol and triglyceride. It also increased the levels of high-density lipoprotein cholesterol and antioxidant parameters, such as superoxide dismutase and tissue glutathione as a catalase. These results strongly indicated the cardioprotective effect of the SC-based formulations, which might be further developed as good alternatives for the treatment of heart-related diseases [49].

3.3. Hypolipidemic Activity

It has been documented in Unani medicine that SC has a protective role in hyperlipidemia that is related to its antioxidant compounds [50]. After four weeks of treatment, the crude extract of SC successfully lowered the rise of serum lipid levels caused by cholesterol-rich foods in New Zealand white rabbits and reduced the size of atherosclerotic plaques. The researchers concluded that an extract of SC could probably inhibit lipid implantation in the injured arterial wall due to its lipid lowering and antioxidant property [51].
In addition, previous studies showed that combining the SS extracted from SC and a high-fat diet intake has been associated with hypolipidemic effects [52]; therefore, SS is further considered a potential obesity prevention approach. Obesity, especially cases caused by high-fat and/or high-calorie diets, has been shown to affect intestinal wall components [53]. A high-fat diet was induced in a C57BL/6 obese mice model, and although SS treatment was not able to reverse plasma and biometric changes promoted by obesity, it restored the jejunal morphometry, including a reduction in the intestinal wall thickness and villus height and an increase in the crypt depth. Furthermore, SS increased lipid excretion in the feces of obese mice, signifying its prospective antiobesity effects [11].

3.4. Anti-Inflammatory Effect

Carbonized SC has been widely used for many inflammation-related diseases. SC-based carbon dots (CDs) that were prepared using a high-temperature pyrolysis method exhibited significant anti-inflammatory bioactivity that was comparable in efficacy to established treatments like dexamethasone. CDs are a class of carbon-based nanomaterials known for their luminescent properties and zero-dimensional structure, composed solely of carbon atoms and enriched with various organic functional groups on their surface [54]. The underlying mechanism of the anti-inflammatory action of SC-CDs appears to be their capacity to inhibit interleukin-6 and tumor necrosis factor-α [3], which are central to the initiation and maintenance of inflammatory responses in the body. This modulation by SC-CDs highlights a promising therapeutic pathway for managing inflammatory conditions [55].
SS has a positive effect on the treatment of chronic inflammatory diseases [56]. It can effectively reduce the thickness of the epidermis in psoriatic skin conditions, akin to the action of known anti-inflammatory drugs such as betamethasone and calcitriol. This is primarily due to its ability to regulate key inflammatory pathways, including the downregulation of the C–C motif chemokine 20 and the JAK-signal transducers and activators of the transcription signaling pathway, thus reducing the production of inflammatory cytokines and alleviating the overall inflammatory response. In conclusion, the multiple actions of SS underscore its potential application in treating psoriatic and other skin diseases, particularly in regulating epidermal cell proliferation and the dynamics of immune responses [12].

3.5. Antioxidant Effect

The ethanolic extract from the green cocoons, rich in flavonoids, has been reported to have excellent antioxidant properties. The half-maximal inhibitory concentration values for 2,2-diphenyl-1-picrylhydrazyl and 1,2′-azino-bis(3-ethylbenzthiazoline-6-sulphonicacid) tests were found to be 296.95 ± 13.24 μg/mL and 94.31 ± 9.13 μg/mL, respectively. Moreover, this extract was found to reduce the level of reactive oxygen species and oxidative stress in L02 cells induced by high glucose levels [57].
In a further study of the antioxidant capacity, three fractions were isolated from SC, i.e., crude SS, purified SS, and the methanolic extract. In the Trolox equivalent antioxidant capacity assay, the methanolic extract exhibited a higher antioxidant capacity with approximately twice the Trolox equivalents than the other two fractions. Murine retinal photoreceptor cells are the cell line that is most vulnerable to oxidants and play an essential role in retinopathies that are primarily caused by oxidative stress. When cultured with the murine retinal photoreceptor cells, compared with crude SS and the purified SS fractions, the methanolic extract improved the cell viability by nearly 10 times at a concentration of 1 mg/mL and displayed maximum cell activity at a concentration of 5 mg/mL. This result indicated that this fraction significantly protected the cultured cells exposed to hydrogen peroxide at different doses [58].
Furthermore, SC increased the total collagen content in human dermal fibroblast cells that were exposed to ultraviolet A1. This was because SC primarily comprises SS and associated secondary metabolites such as polyphenols and flavonoids, which may be able to partially prevent ultraviolet radiation-induced photoaging [59].

3.6. Antiviral and Antimicrobial Effects

The 95% ethanol extract of SC showed a high potential activity against the herpes simplex virus (HSV)-1 and HSV-2. HSV-2-infected HeLa cells treated with the 95% ethanol extract of SC drastically reduced cell death and prevented inflammation by reducing the production of inflammatory cytokine genes. The bioactive components of this portion of the extract were further shown to be gallic acid, flavonoids, and xanthophyll, and they played important roles in the antiviral activity. This study indicated that the SC extract has potential as a therapeutic treatment for herpes simplex virus (HSV) infection [21].
The underlying mechanism of the antimicrobial effects of SC was also investigated. SC contains a large number of proteins that inhibit the growth of fungi and bacteria, including protease inhibitors and seroins [60]. BmSPI51 is the most abundant SC protease inhibitor, and it has been reported to play a part in the antimicrobial role of SC. It strongly suppressed the sporular growth of three fungal species, i.e., Candida albicans, Beauveria bassiana, and Saccharomyces cerevisiae. According to the in vitro inhibition experiments, its defense mechanisms revealed that BmSPI51 inhibited fungal growth by binding to the cell wall polysaccharides, mannan, and β-glucan or preventing fungi from obtaining nutrients and retarding the rate of budding [61].
Table 2. The biological activities and mechanisms of silkworm cocoon.
Table 2. The biological activities and mechanisms of silkworm cocoon.
Biological ActivitiesSampleAnimalModelDoseResultsMechanismsRefs.
hypoglycemic
effect
fibroin, sericinsilkworm
/in vivo
diets containing glucose or sucrose5% added to the dietsexhibited postprandial
antihyperglycemic activity
inhibition of the expression of intestinal glucose transporters, promotion of the regeneration of pancreatic β cells, or activation of the insulin-like signaling pathway[42]
ethanolic
extract from
the green
cocoon sericin layer of silkworm
ICR mice
/in vivo
fed with a high-fat diet and
injected with
streptozotocin
150, 250, and 350 mg/kgameliorated glucose
metabolism and
regulated the balance
between glycolysis
and gluconeogenesis
reduction of the levels of NF-κB, IL-6, and TNF-α; enhancement of the expression levels of IR, IRS, PI3K, p-Akt, and p-GSK3β involved in insulin signalling; activation of AMPK and GLUT4; reduction of the levels of G6pase and PEPCK; improvement of the GK level[43]
flavonoid-rich ethanolic extract from silkworm green
cocoon
ICR mice
/in vivo
induced by
high-fat and
streptozotocin
150, 250, and 350 mg/kgregulated the glucose
level and body weight
and improved renal
dysfunction
inhibition of the TNF-α-p38 MAP kinase signaling pathway[39]
hydroalcoholic extract of silk cocoonWistar rats
/in vivo
induced by
streptozotocin
200, 400, and 800 mg/kgdecreased prolactin and inhibin; increased leptin, IGF-2, activin A, insulin, LH, testosterone, FSH, and GnRH levels; improved gonadal weight, the diameter of tunica albuginea, and seminiferous tubules as well as increased the numbers of spermatocytes and Sertoli–Leydig cellsNA[44]
cardioprotective
effect
sericinWistar rats
/in vivo
isoproterenol induced cardiac toxicity and hypertrophy500 and 1000 mg/kgsignificantly increased the non-enzymatic antioxidant
markers in serum and heart tissue; significantly decreased the myocyte size
prevented the myocardial tissue from enzymatic leakage from the cell sites; reduced the synthesis of collagen in myocardiocytes, thus reducing the incidence of fibrosis; reduction of fibrosis and synthesis of collagens contributed to the protective effect against hypertrophy; decreased
inflammatory reactions and oxidative stress, which led to improved myocardial activity, and reduced cardiac damage after myocardial ischemia
[9]
sericinWistar rats
/in vivo
cholesterol diet-induced hypercholesterolaemia model1000 mg/kgimproved cardiac muscle contraction under hypercholesterolaemia, restored the cardiac mitochondrial structure, increased mitochondrial fusion in the heart, and inhibited the progression of apoptosis at the last stage of dysmorphic mitochondriaupregulation of OPA1 and reduction of NADH-ubiquin-one oxidoreductase 75 kDa subunit expression; improvement of mitochondrial energy production by upregulating acetyl-CoA acetyltransferase and NADH dehydrogenase 1a subcomplex subunit 10 expression[48]
ethanolic extract of silk cocoonsAlbino Wistar rats
/in vivo
isoprenaline-induced myocardial infarction250 and 500 mg/kgsignificantly prevented myocardial damage and hypertrophy, and decreased the levels of various cardiac enzymesNA[8]
an emulsion formulation composed of methanol extract of silk cocoons, flaxseed oil, and coenzyme Q10Sprague Dawley rats
/in vivo
doxorubicin induced myocardial toxicity500 mg/kg methanol extract of silk cocoons, 1.8 mL/kg flaxseed oil, and 5 mg/kg coenzyme Q10significantly prevented the increase in serum levels of AST, ALT, LDH, and creatinine and the lipid profile, increased the levels of HDL, SOD, GSH, and CAT in heart tissue, and lowered the increase in heart weight due to hypertrophymay be mainly due to the high protein content of sericin, flavonoids, and n-3 fatty acids that have potential free radical scavenging and antioxidant activities (the author speculated; needs to be experimentally confirmed)[49]
hypolipidemic
effect
1% NaCl
solution extract of silk cocoons
New
Zealand
white
rabbits
/in vivo
cholesterol powder mixed with coconut oil50 mg/
100 g
reduced the levels of total cholesterol, triglycerides, and low-density lipoprotein, as well as the size of atherosclerotic plaque in the aorta; increased the high-density lipoprotein
level and body weight
probably inhibited the second step of lipid implantation in the injured arterial wall by its lipid-lowering and antioxidant
properties (the author speculated; needs to be experimentally confirmed)
[51]
sericinC57BL/6
mice
/in vivo
fed with fat-rich diets1000
mg/kg
increased lipid excretion
in feces and restored intestinal wall morphometry in obese
mice
NA[11]
anti-inflammatory effectsilkworm cocoon-derived carbon dotsC57 black mice and Kunming mice
/in vivo
(1) dimethylbenzene-induced ear oedema;
(2) vascular permeability induced by acetic acid;
(3) lipopolysaccharide- induced sepsis model
0.35, 0.7, and 1.4 mg/kgsignificantly lowered the percentage inflammation at the doses of 0.7 and 1.4 mg/kg, and the plasma extravasation
of the test groups was similar to that of the dexamethasone group
inhibition of the expressions of IL-6 and TNF-α[3]
sericinSprague–Dawley rats
/in vivo
imiquimod-induced skin psoriasis2.5, 5, and 10% sericin cream applied topically10% sericin had the desired effect of improving skin psoriasis, similar to that of betamethasone and calcitriol treatmentsreduction in cytokine production of Th17 cells by interfering with the JAK-STAT signaling pathway; modulation of immune response via upregulation of galectin-3 and downregulation of sphingosine-1-phosphate lyase1[12]
antioxidant effectethanolic extract of the green cocoonsin vitroDPPH and ABTS assayin DPPH test:
IC50 = 296.95 ± 13.24 μg/mL; in ABTS test: IC50 = 94.31 ± 9.13 μg/mL
showed excellent antioxidationNA[57]
diazo cocoon extractsin vitroDPPH and ABTS assayNAexhibited high antioxidant activitiesNA[58]
silk sericin and associated secondary metabolites (polyphenols and flavonoids)in vitrohuman dermal fibroblast cellsNAthe human dermal fibroblast cells treated with silk sericin exposed to UVA1 showed a significant increase in total collagen contentupregulates the expression of MMP-1 in human dermal fibroblast cells along with MMP-3, resulting in the degradation of collagen, and leads
to the loss of the structural integrity of the skin
[59]
antiviral and antimicrobial effect95% ethanol extract of silk cocoonin vitroHSV-1 and HSV-2NAthe inactivation of HSV-1 and HSV-2drastically reduced HSV-induced cell death and prevented inflammation by reducing the production of inflammatory cytokine genes[21]
silkworm cocoonin vitrothree different species of fungi: Candida albicans, Beauveria bassiana, and Saccharomyces cerevisiaeNAstrongly suppressed the sporular growth of the three fungal speciesBmSPI51 attaches to mannan and β-D-glucan on the surface of fungal cells, thus inhibiting fungal growth[61]
NA: not available.

3.7. Other Effects

SC has other pharmacological activities that include antitumor and analgesic effects. Its primary component, SS, has an effect against colon cancer cell lines like SW480, where it regulates cellular processes, such as apoptosis, by increasing caspase-3 activity and reducing Bcl-2 expression, highlighting its therapeutic potential in tumor treatment [62]. Moreover, the unique strength and elasticity of SC make it well suited for treating tendon strains and aiding wound healing. It also possesses pain-relieving properties. Although this effect has been observed in tendinopathy models, further research is required to reveal its mechanisms of action [63].

4. Practical Applications from Laboratories to Clinics and Markets

As has been mentioned previously, SF, SS, and their related derivative materials are the primary bioactive components of SC, and they also act as biodegradable carriers with good compatibility. They have been widely used in drug delivery systems, tissue engineering, regenerative medicine, and in vitro diagnostics. These applications offer new treatment options for a variety of medical conditions. Nevertheless, most studies are still in the laboratory investigation stages, and only a few products have entered clinical trials and even the market. The current studies and the marketed products are primarily based on SF. The safety and efficacy of SF-based products have been confirmed through clinical studies, and a few of them have been approved by the Food and Drug Administration (FDA). The continuous development of SF bodes well for the broad prospects of silk-based materials in biomedical applications and markets.

4.1. Laboratory Investigations

The wide range of applications of SF and its derived materials has demonstrated significant innovation in the laboratory research stage, and these materials provide advanced solutions for future clinical applications and medical diagnostics.

4.1.1. Application in Drug Delivery Systems

SF has been used in drug delivery systems to act as a promising carrier of films [64,65], nanoparticles [66,67], hydrogels [68], and microneedles [69,70,71,72] for diverse therapeutic attempts. The schematic diagram is depicted in Figure 1.
SF films loaded with honeysuckle flower extract induced apoptosis in HeLa cells, signifying its potential use as a promising material for cancer therapy [64]. SF films also act as a sustained release delivery system for insulin-like growth factor-1 that significantly accelerates wound healing in diabetic mice. The zero-order release kinetics of SF films ensure the continuous presence of insulin-like growth factor-1 at the wound site, thus enhancing reepithelialization and granulation tissue formation [65].
The integration of SF into nanoparticle-based drug delivery systems offers a promising approach to enhancing the efficacy and safety of various therapeutic agents. To overcome the poor bioavailability of curcumin, SF nanoparticles have been engineered to encapsulate curcumin, leading to improved cellular uptake and cytotoxicity against cancer cells while sparing healthy cells [66]. Baicalein, which is a flavonoid with anti-inflammatory properties, has been combined with SF to enhance its therapeutic effects. The resulting SF–baicalein complex demonstrated significant anti-inflammatory activity in a zebrafish model, indicating potential for developing new anti-inflammatory drugs with reduced side effects [67]. These systems have shown the particular promise of SF in cancer therapy and anti-inflammatory applications, with potential for further expansion into other areas of pharmacotherapy.
SF hydrogels, with their unique properties, show promise as a carrier for protein drugs, offering a sustainable and effective solution for drug delivery systems. SF hydrogels have demonstrated good drug-loading capacities, with a cumulative release of 80% bovine serum albumin within 12 h. Additionally, they simultaneously exhibit excellent stability and biocompatibility, with a storage modulus that increases significantly with concentration. The degradation behavior of SF hydrogels in various media further confirmed their potential for broad applications in biomedicine [68].
SF microneedles have garnered considerable interest due to their biocompatibility, mechanical strength, and tunable degradation properties. They are typically prepared using a process that involves the degumming of SC, the dissolution of silk fibers, and the subsequent solidification into a microneedle form. SF microneedles have been successfully applied for the transdermal delivery of insulin, demonstrating the potential to provide a painless and effective alternative for diabetic patients [69]. Additionally, their use in vaccine delivery has shown promising results, with the ability to elicit robust immune responses against various pathogens. Figure 2 shows the preparation process for vaccine-coated SF microneedles [70]. The ability of SF microneedles to protect labile biomolecules and control release rates makes them suitable for a wide range of therapeutic applications [71,72], and this represents a significant advancement in the field of transdermal drug delivery and positions them as a versatile platform for the delivery of diverse therapeutic agents.

4.1.2. Application in Tissue Engineering

The exceptional multifunctionality of SF has established it as a significant material in the field of biomedical tissue engineering [73,74]. It is widely used to construct human scaffolds and develop bone hydrogels, making it a leading candidate material for tissue engineering (as summarized in Figure 3).
SF-based scaffolds are commonly recognized for their superior biocompatibility and tunable mechanical properties. With the use of special catalysts, the precise control of the cryogelation process and catalytic cross-linking reactions has promoted the development of new SF scaffolds that surpass the structural and physical properties of traditionally prepared scaffolds, demonstrating better flexibility, elasticity, and cellular biological performance [75]. Furthermore, the combination of SF with hyaluronic acid [76] and silver nanoparticles [77] has been shown to be particularly useful in bone tissue engineering. By adjusting the proportions and cross-linking strategies of these materials, the developed composite scaffolds have shown significant advantages in promoting cell proliferation and biocompatibility and are especially suitable for cartilage and bone tissue engineering [78,79]. Furthermore, the use of 3D-printing technology to construct complex three-dimensional structures required for tissue engineering with bio-inks made from these composite materials (as shown in Figure 4 [80]) can result in implants and scaffold structures suitable for more complex structural and functional applications [80,81]. It can also simulate the complex geometric shapes of natural tissues and organs [82], which is crucial for tissue engineering applications.
The development of SF bone hydrogels represents a significant advancement in tissue engineering. These hydrogels provide an ideal three-dimensional growth framework for cells by mimicking the natural environment of the extracellular matrix, and they feature an appropriate water content, controllable degradation rates, and good mechanical stability [83]. In particular, by improving the SF structure by combining physical and enzymatic crosslinking strategies, an SF matrix composite hydrogel with enhanced strength and elasticity was obtained. This development also improves the shortcomings of traditional SF hydrogels with uneven structures and poor mechanical properties [84].

4.1.3. Application in Regenerative Medicine

The application of SF encompasses numerous aspects of regenerative medicine, including bone regeneration, vascular regeneration, and soft tissue regeneration, which benefit from the ability of SF to be designed into various forms, such as porous scaffolds and fibrous nets, to meet diverse medical needs [85] (as summarized in Figure 5).
In bone regeneration, an innovative study successfully developed a biological composite thin film by combining hydroxyapatite with SF [86] that enhanced bone conductivity and biocompatibility. It also showed significant potential in promoting bone tissue repair and regeneration, even with slightly reduced tensile strength, offering new material options.
The high design flexibility of SF also plays a crucial role in the domain of vascular regeneration and soft tissue regeneration. Researchers have constructed vascular grafts with excellent blood compatibility and good in-body tolerance using advanced purification, processing, and functionalization techniques. These SF vascular grafts not only mimic the mechanical properties of natural arteries, thus facilitating the rapid restoration of the endothelial cell layer, but also promote vascular remodeling by modulating local inflammatory responses [87]. They significantly reduce the tendency for thrombus formation and show a lower thromboprotein adsorption rate than traditional polytetrafluoroethylene materials, a key to maintaining the long-term patency of vascular grafts [88]. In addition, a new type of “bio-origami” material has been developed by integrating SS into biopolymer films with the aim to promote skin regeneration. These films that utilize different concentrations of SS exhibit outstanding physicochemical properties, mechanical strength, and antioxidant capacities [89].

4.1.4. Application in In Vitro Diagnosis

SC and its derived materials have shown extensive potential in the field of in vitro diagnosis, and the latest research progress in biosensing platforms, cell cultures, signal molecule monitoring, blood type identification, and biocompatibility improvements has shown its value in improving diagnostic accuracy, convenience, and biomedical applications [90,91,92,93,94,95] (as summarized in Figure 6).
Gold nanoparticles (Au NPs) have important applications in biological imaging due to their unique physicochemical properties. Through the regulatory role of SF as a carrier, a biocompatible Au NP with uniform size and good dispersion was prepared. The SF fibers played multiple roles in the formation of the Au NPs, including as a reactive substrate, reducing agent, and modifier. These Au NPs not only exhibit excellent performance in biological imaging but also serve as efficient contrast agents to improve the accuracy of medical imaging [90].
A 3D cell-adhesive sensing matrix was designed and fabricated from SC-derived hierarchical carbon fiber networks assembled with the iron porphyrin of hemin. The matrix, which provided excellent biocompatibility for cell adhesion and long-term culture, was capable of highly sensitive and selective monitoring of cell-released nitric oxide molecules that could be used for live-cell assays and to explore physiological processes in complex biological systems. This is of great significance for understanding cellular behavior and disease mechanisms [91].
A versatile and thermally stable immunosensing platform was investigated using natural SC membranes as the substrate material. The platform utilized the intrinsic properties of the SC membrane for the directional immobilization of biomolecules through immunoaffinity recognition. This not only improved the sensitivity and selectivity of the sensor but also enhanced the stability and reliability of the sensor by maintaining the biological activity of antibodies. The immunosensor showed significant detection capabilities and excellent stability for immobilized antibodies, making it suitable for immunoassays and potential applications in resource-limited settings [92]. Correspondingly, the natural SC membranes were also used to develop a rapid and reliable immunoassay for ABO and RhD blood group typing. The assay was simple, requiring only a few pipetting steps and observable results within 30 s, making it suitable for point-of-care testing. The accuracy of the assay was validated against the gold-standard tube test, and it showed a 100% match, suggesting its effectiveness for blood typing [93].
Another study explored the biofunctionalization of biomimetic silk biomaterials with recombinantly expressed domain V of the human basement membrane proteoglycan perlecan for blood-compatible surfaces. The recombinantly expressed domain V was covalently immobilized on SF using plasma immersion ion implantation, a method that does not rely on specific amino acids in the silk protein chain. The biomimetic silk biomaterials demonstrated blood compatibility and could be a promising platform for developing blood-contacting devices. This provides new strategies for the development of vascular transplantation and blood contact devices [94].
A visually detectable H2O2 sensing system was created based on a gold nanozyme-SF hydrogel hybrid. The hydrogel exhibited a fast response and high sensitivity for H2O2 detection, with excellent stability and selectivity. The system has significant potential for the clinical diagnosis of H2O2, showcasing compatibility with biological tissues and minimal cytotoxicity [95].
The application of SC and its derived materials in in vitro diagnoses has demonstrated its multifunctionality and biocompatibility, providing new possibilities for developing new diagnostic tools and improving disease diagnosis efficiency.

4.2. Clinical Studies

Apart from the favorable laboratory research results mentioned above, several products based on SF have entered different stages of clinical research. By searching the website of www.clinicaltrials.gov launched by the U.S. National Institutes of Health [96], we summarized the information of these clinical trials that can be seen in Table 3. Among these clinical trials, three trials have already been completed, while the rest are still under recruitment or have an unknown status.
NanoSilk Cosmo is a novel SF-based product developed with the aim of addressing the biophysical parameters associated with skin aging. This product has been the subject of clinical trials and research studies to evaluate its safety and efficacy for human facial skin, particularly for improving skin resiliency and hydration. The results of the study suggested that NanoSilk Cosmo is safe for use on human facial skin. Furthermore, it has been observed to improve skin resiliency and hydration, which are essential factors in maintaining youthful-looking skin. This indicates it may offer benefits in the management of skin aging, providing a potential new option for individuals seeking to improve the appearance and health of their skin [97].
The efficacy and safety of wound dressings are critical factors in the management of split-thickness skin graft donor sites. A wound dressing containing SF with a bioactive coating layer was developed as an alternative to traditional medicated paraffin gauze dressings. Clinical studies have revealed that SF-based wound dressings can accelerate the healing process, reduce pain associated with wound care, and improve the overall cosmetic outcome compared to traditional dressings such as medicated paraffin gauze. It been shown that these dressings reduce the time to complete wound closure, decrease the frequency of dressing changes, and minimize the risk of complications such as infection and scarring. The use of a wound dressing containing SF with a bioactive coating layer represents a promising advancement in the treatment of split-thickness skin graft donor sites [96].

4.3. Products on the Market

In recent years, with the significant progress in the application of SF in the medical field, multiple SF-based products have been successfully launched domestically and internationally, and they are summarized in Table 4. The most representative products are Silk Voice®, SERI®, Tympasil®, and Sidaiyi®.
Developed by Sofregen Medical, Inc., Silk Voice® is the first and only natural silk protein injectable for tissue bulking. The FDA cleared it for vocal fold medialization and vocal fold insufficiency that may be improved by injection of a soft tissue bulking agent [98]. It comprises porous bioabsorbable silk particles suspended in an isotonic, aqueous formulation of cross-linked, high-molecular-weight hyaluronic acid. This unique composition enables the product to be biocompatible and easily integrated into the body’s tissues, with the silk particles gradually being absorbed over time as they promote the patient’s own tissue growth [99]. The development and approval of Silk Voice® represent a milestone in the application of silk-based biomaterials in medical treatments. Another FDA-approved SF-based product, SERI®, was also developed by Sofregen Inc. It is a medical device used for abdominal wall reconstruction and plastic surgery, and it has shown high satisfaction and minimal complications in clinical trials [100].
Other countries have also approved the use of SF in vivo by their regulating authorities. Tympasil® is a medical device produced by Daewoong-Bio, South Korea. This product is a thin, transparent patch made from SF that is designed for the repair of acute tympanic membrane perforations [101], offering a noninvasive and effective treatment option for patients. In clinical studies, Tympasil® has demonstrated promising results in treating patients with chronic tympanic membrane perforations. The patch is softened by immersion in warm saline and then carefully placed over the perforation site. This process allows the patch to conform to the shape of the eardrum, promoting healing and regeneration of the tissue [102]. In China, SF has been approved for use as a clinical wound dressing, with the brand name of Sidaiyi®. It is designed as a two-layered system, featuring an SF sponge attached to a silicone membrane. This unique construction is intended to provide an environment conducive to wound healing, offering protection from external contaminants while simultaneously promoting the regeneration of skin tissue [103]. It represents a significant advancement in the field of regenerative medicine and wound care. Moreover, other SF-related products have been approved in succession. For example, absorbable SF repair film is used for oral dental implant repair, and it can promote the growth of alveolar bone and prevent soft tissue invasion. It has become the first domestically approved Class III implantable SF medical device product.

5. Safety

The collective findings from the current studies underscore the potential of SF and SS as safe and effective materials for various applications. The safety assessment of SS was studied using a series of rigorous tests that included bacterial reverse mutation tests, mammalian erythrocyte micronucleus tests, and a 90-day subchronic toxicity study in Sprague–Dawley rats. SS exhibited no genotoxicity or mutagenicity, and its no-observed-adverse-effect level was determined to be 1 g/kg/day, suggesting a low toxicity profile [104].
Another study examined the toxicological assessment and potential allergy risks associated with SF. A comprehensive battery of tests, such as the Ames test, the in vivo mouse erythrocyte micronucleus test, and 28-day oral toxicity studies, were conducted to evaluate the safety of SF for consumption. An in vitro pepsin digestion assay was performed to assess the allergenicity of SF [105]. The studies concluded that SF does not raise concerns regarding mutagenicity, genotoxicity, toxicity, or allergenicity, making it a promising material for food preservation applications.
The cytotoxicity, biocompatibility, and cell adhesion kinetics of SF were further explored. It was found to be immunologically inert, invoking minimal immune responses. This was evaluated by measuring the nitric oxide and factor-alpha production in murine peritoneal macrophages and RAW 264.7 murine macrophages, and it showed a comparable stimulation as collagen. Cell viability studies and flow cytometric analyses indicated that the SF matrices supported cell growth and proliferation comparable to collagen, making it suitable for long-term culture applications [106].

6. Conclusions and Future Perspectives

Researchers in the field of medical bioengineering are especially interested in SC due to its multifunctionality, morphological flexibility, and excellent biocompatibility. The dual functionality of SC both as a traditional Chinese medicine and the raw material of biocompatible carriers presents an intriguing avenue for its further exploration and utilization.
Nevertheless, the research and application of SC is still facing a raft of issues and challenges: (1) Existing research on the pharmacological effects and clinical applications of SC remains in the initial phases, necessitating more in-depth studies on its mechanisms and therapeutic efficacy. (2) Traditional silk biomaterial preparation involves dissolution and reshaping, a complex and lengthy process that calls for the development of simpler and more sustainable methods. (3) SC, as a potential wound dressing material, inherently possesses certain antimicrobial properties. However, it may need to be improved or combined with other materials to enhance the antimicrobial effectiveness to meet higher medical application standards. (4) SC is a natural material with unique geometric characteristics and can be used as an excellent model for biomimetic design and applications. Nevertheless, effectively translating these characteristics into practical applications remains challenging. (5) More comprehensive research is required on the mechanical behavior of SC-derived materials, especially their performance under different strain rates and microstructures, to optimize their use in various engineering applications. (6) Wild SC is a potential alternative for SC resources, but effective utilization of these resources and overcoming challenges related to silk extraction and quality compared to domesticated silkworms remain issues that need to be addressed.
To summarize, as our understanding of SC and its primary components deepens, we believe that its latent value and application scope will continue to be explored and expanded. This will bring more surprises and breakthroughs to future medical and life science research. Notably, the research and application of SC encompass a wide range of issues that require interdisciplinary collaboration, while existing challenges should be overcome through innovative approaches.

Author Contributions

Z.T., C.Z. and T.H. wrote and edited the original draft. L.Y. and Y.S. analyzed and interpretated the data. Y.T. and Y.C. prepared the graphics and tables. R.D. and W.L. supervised the writing. J.Z. supervised, reviewed, and finalized the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Shanghai Municipal Commission of Science and Technology (No. 23S21901100), Innovation Promotion Program of NHC and Shanghai Key Labs, SIBPT (No. CX2023-05), and Expenditure Budget Program of Shanghai University of Traditional Chinese Medicine (No. 2021LK003).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data were used for the research described in the paper.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

SC: silkworm cocoon; SS, silk sericin; SF, silk fibroin; CDs, carbon dots; HSV, herpes simplex virus; Au NPs, gold nanoparticles; FDA, Food and Drug Administration.

References

  1. Zhang, L.; Zhang, L.; Li, Y.; Guo, X.-F.; Liu, X.-S. Biotransformation effect of Bombyx mori L. may play an important role in treating diabetic nephropathy. Chin. J. Integr. Med. 2016, 22, 872–879. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, X.; Chen, Y.; Zhang, J.; Zhou, L.; Zhang, Y.; Gu, X. Clinical observation of silkworm cocoon shell decoction combined with salmeterol in the treatment of bronchial asthma. J. Clin. Pulm. Med. 2018, 23, 404–408. [Google Scholar]
  3. Wang, X.; Zhang, Y.; Kong, H.; Cheng, J.; Zhang, M.; Sun, Z.; Wang, S.; Liu, J.; Qu, H.; Zhao, Y. Novel mulberry silkworm cocoon-derived carbon dots and their anti-inflammatory properties. Artif. Cells Nanomed. Biotechnol. 2020, 48, 68–76. [Google Scholar] [CrossRef] [PubMed]
  4. Dong, Z.; Xia, Q.; Zhao, P. Antimicrobial components in the cocoon silk of silkworm, Bombyx mori. Int. J. Biol. Macromol. 2023, 224, 68–78. [Google Scholar] [CrossRef] [PubMed]
  5. DeBari, M.K.; King, C.I., III; Altgold, T.A.; Abbott, R.D. Silk fibroin as a green material. ACS Biomater. Sci. Eng. 2021, 7, 3530–3544. [Google Scholar] [CrossRef] [PubMed]
  6. Cong, L.; Cao, G.; Renyu, X.; Zhonghua, P.; Xiaojian, Z.; Zhou, W.; Gong, C. Reducing blood glucose level in TIDM mice by orally administering the silk glands of transgenic hIGF-I silkworms. Biochem. Biophys. Res. Commun. 2011, 410, 721–725. [Google Scholar] [CrossRef] [PubMed]
  7. Lee, H.J.; Choi, J.W.; Ra, K.S.; Kim, J.-M.; Suh, H.J. Novel tripeptides with α-glucosidase inhibitory activity isolated from silk cocoon hydrolysate. J. Agric. Food Chem. 2011, 59, 11522–11525. [Google Scholar] [CrossRef] [PubMed]
  8. Srivastav, R.K.; Siddiqui, H.H.; Mahmood, T.; Ahsan, F. Evaluation of cardioprotective effect of silk cocoon (Abresham) on isoprenaline-induced myocardial infarction in rats. Avicenna J. Phytomed. 2013, 3, 216–223. [Google Scholar] [PubMed]
  9. Ahsan, F.; Mahmood, T.; Wani, T.A.; Zargar, S.; Siddiqui, M.H.; Usmani, S.; Shamim, A.; Wahajuddin, M. Effectual Endeavors of silk protein sericin against isoproterenol induced cardiac toxicity and hypertrophy in wistar rats. Life 2022, 12, 1063. [Google Scholar] [CrossRef]
  10. Shariq, M.; Mahmood, T.; Kushwaha, P.; Parveen, S.; Shamim, A.; Ahsan, F.; Wani, T.A.; Zargar, S.; Wasim, R.; Wahajuddin, M. Fabrication of nanoformulation containing carvedilol and silk protein sericin against doxorubicin induced cardiac damage in rats. Pharmaceuticals 2023, 16, 561. [Google Scholar] [CrossRef]
  11. Kunz, R.I.; Capelassi, A.N.; Alegre-Maller, A.C.P.; Bonfleur, M.L.; Ribeiro, L.d.F.C.; Costa, R.M.; Natali, M.R.M. Sericin as treatment of obesity: Morphophysiological effects in obese mice fed with high-fat diet. Einstein (Sao Paulo) 2020, 18, eAO4876. [Google Scholar] [CrossRef] [PubMed]
  12. Rujimongkon, K.; Ampawong, S.; Reamtong, O.; Buaban, T.; Aramwit, P. The therapeutic effects of Bombyx mori sericin on rat skin psoriasis through modulated epidermal immunity and attenuated cell proliferation. J. Tradit. Complement. Med. 2021, 11, 587–597. [Google Scholar] [CrossRef] [PubMed]
  13. Ritprajak, P.; Sirithanakorn, C.; NY Nguyen, T.N.; Sereemaspun, A.; Aramwit, P. Biosynthetic sericin 1-like protein skews dendritic cells to tolerogenic-like phenotype. Biotechnol. Appl. Biochem. 2021, 68, 1508–1517. [Google Scholar] [CrossRef] [PubMed]
  14. Tuentam, K.; Aramwit, P.; Reamtong, O.; Supasai, S.; Chaisri, U.; Fongsodsri, K.; Yamdech, R.; Tirawanchai, N.; Sukphopetch, P.; Ampawong, S. Sericin-based poly(Vinyl) alcohol relieves plaque and epidermal pesions in Psoriasis; a chance for dressing development in a specific area. Int. J. Mol. Sci. 2022, 24, 145. [Google Scholar] [CrossRef]
  15. Seyedaghamiri, F.; Farajdokht, F.; Vatandoust, S.M.; Mahmoudi, J.; Khabbaz, A.; Sadigh-Eteghad, S. Sericin modulates learning and memory behaviors by tuning of antioxidant, inflammatory, and apoptotic markers in the hippocampus of aged mice. Mol. Biol. Rep. 2021, 48, 1371–1382. [Google Scholar] [CrossRef] [PubMed]
  16. Dash, R.; Acharya, C.; Bindu, P.C.; Kundu, S.C. Antioxidant potential of silk protein sericin against hydrogen peroxide-induced oxidative stress in skin fibroblasts. BMB Rep. 2008, 41, 236–241. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, F.; Wang, R.Y.; Zhong, D.B.; Zhao, P.; Xia, Q.Y. Highly efficient expression of human extracellular superoxide dismutase (rhEcSOD) with ultraviolet-B-induced damage-resistance activity in transgenic silkworm cocoons. Insect Sci. 2023, 30, 1–15. [Google Scholar] [CrossRef] [PubMed]
  18. Mumtaz, S.; Ali, S.; Pervaiz, A.; Awan, U.A.; Nauroze, T.; Kanwal, L.; Summer, M.; Mumtaz, S.; Mughal, T.A.; Tahir, H.M. Toxicological effects of dimethlybenzeneanthracene in Balb C mice and pharmacological intervention by silk sericin–conjugated silver nanoparticles. Sci. Prog. 2024, 107, 1–32. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, Y.; Tang, M.; Dong, Z.; Zhao, D.; An, L.; Zhu, H.; Xia, Q.; Zhao, P. Synthesis, secretion, and antifungal mechanism of a phosphatidylethanolamine-binding protein from the silk gland of the silkworm Bombyx mori. Int. J. Biol. Macromol. 2020, 149, 1000–1007. [Google Scholar] [CrossRef]
  20. Deng, Y.; Yang, C.; Zhu, Y.; Liu, W.; Li, H.; Wang, L.; Chen, W.; Wang, Z.; Wang, L. Lamprey-teeth-inspired oriented antibacterial sericin microneedles for infected wound healing improvement. Nano Lett. 2022, 22, 2702–2711. [Google Scholar] [CrossRef]
  21. Jantakee, K.; Prangkio, P.; Panya, A.; Tragoolpua, Y. Anti-herpes simplex virus efficacy of silk cocoon, silkworm pupa and non-sericin extracts. Antibiotics 2021, 10, 1553. [Google Scholar] [CrossRef]
  22. Tan, H.; Ji, Y.; Lei, H.; Wang, F.; Dong, H.; Yang, S.; Zhou, H.; Deng, H.; Chen, S.; Kaplan, D.L.; et al. Large-scale and cost-effective production of recombinant human serum albumin (rHSA) in transgenic Bombyx mori cocoons. Int. J. Biol. Macromol. 2023, 245, 125527. [Google Scholar] [CrossRef] [PubMed]
  23. Lian, A.A.; Yamaji, Y.; Kajiwara, K.; Takaki, K.; Mori, H.; Liew, M.W.O.; Kotani, E.; Maruta, R. A bioengineering approach for the development of fibroblast growth factor-7-functionalized sericin biomaterial applicable for the cultivation of keratinocytes. Int. J. Mol. Sci. 2022, 23, 9953. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, H.; Xiang, P.; Yu, B.; Sun, Z.; Li, X.; Qv, A.; Sohail, M.; Li, Y. Research progress of novel drug delivery systems of Chinese medicine monomers based on natural silk fibroin: A mini-review. Curr. Drug Deliv. 2023, 20, 211–222. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, S.; Yang, Q.; Wang, R.; Tian, C.; Ji, Y.; Tan, H.; Zhao, P.; Kaplan, D.L.; Wang, F.; Xia, Q. Genetically engineered pH-responsive silk sericin nanospheres with efficient therapeutic effect on ulcerative colitis. Acta Biomater. 2022, 144, 81–95. [Google Scholar] [CrossRef] [PubMed]
  26. Xu, S.; Tan, H.; Yang, Q.; Wang, R.; Tian, C.; Ji, Y.; Zhao, P.; Xia, Q.; Wang, F. Fabrication of a silk sericin hydrogel system delivering human lactoferrin using genetically engineered silk with improved bioavailability to alleviate chemotherapy-induced immunosuppression. ACS Appl. Mater. Interfaces 2021, 13, 45175–45190. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, J.; Shi, L.; Deng, Y.; Zou, M.; Cai, B.; Song, Y.; Wang, Z.; Wang, L. Silk sericin-based materials for biomedical applications. Biomaterials 2022, 287, 121638. [Google Scholar] [CrossRef] [PubMed]
  28. Silva, A.S.; Costa, E.C.; Reis, S.; Spencer, C.; Calhelha, R.C.; Miguel, S.P.; Ribeiro, M.P.; Barros, L.; Vaz, J.A.; Coutinho, P. Silk sericin: A promising sustainable biomaterial for biomedical and pharmaceutical applications. Polymers 2022, 14, 4931. [Google Scholar] [CrossRef] [PubMed]
  29. Luetchford, K.A.; Chaudhuri, J.B.; De Bank, P.A. Silk fibroin/gelatin microcarriers as scaffolds for bone tissue engineering. Mater. Sci. Eng. C Mater Biol Appl 2020, 106, 110116. [Google Scholar] [CrossRef]
  30. Wang, J.; Liu, H.; Shi, X.; Qin, S.; Liu, J.; Lv, Q.; Liu, J.; Li, Q.; Wang, Z.; Wang, L. Development and application of an advanced biomedical material-silk sericin. Adv. Mater. 2024, 36, e2311593. [Google Scholar] [CrossRef]
  31. Chuysinuan, P.; Nooeaid, P.; Thanyacharoen, T.; Techasakul, S.; Pavasant, P.; Kanjanamekanant, K. Injectable eggshell-derived hydroxyapatite-incorporated fibroin-alginate composite hydrogel for bone tissue engineering. Int. J. Biol. Macromol. 2021, 193, 799–808. [Google Scholar] [CrossRef] [PubMed]
  32. Santi, S.; Mancini, I.; Dirè, S.; Callone, E.; Speranza, G.; Pugno, N.; Migliaresi, C.; Motta, A. A bio-inspired multifunctionalized silk fibroin. ACS Biomater. Sci. Eng. 2021, 7, 507–516. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, K.; Yazawa, K.; Tsuchiya, K.; Numata, K.; Guan, J. Molecular Interactions and toughening mechanisms in silk fibroin-epoxy resin blend films. Biomacromolecules 2019, 20, 2295–2304. [Google Scholar] [CrossRef] [PubMed]
  34. Yao, D.; Li, M.; Wang, T.; Sun, F.; Su, C.; Shi, T. Viscoelastic silk fibroin hydrogels with tunable strength. ACS Biomater. Sci. Eng. 2021, 7, 636–647. [Google Scholar] [CrossRef] [PubMed]
  35. Baptista-Silva, S.; Borges, S.; Costa-Pinto, A.R.; Costa, R.; Amorim, M.; Dias, J.R.; Ramos, Ó.; Alves, P.; Granja, P.L.; Soares, R.; et al. In situ forming silk sericin-based hydrogel: A novel wound healing biomaterial. ACS Biomater. Sci. Eng. 2021, 7, 1573–1586. [Google Scholar] [CrossRef] [PubMed]
  36. Manesa, K.C.; Kebede, T.G.; Dube, S.; Nindi, M.M. Fabrication and characterization of sericin-PVA composite films from Gonometa postica, Gonometa rufobrunnea, and Argema mimosae: Potentially applicable in biomaterials. ACS Omega 2022, 7, 19328–19336. [Google Scholar] [CrossRef] [PubMed]
  37. Kurioka, A.; Yamazaki, M. Purification and identification of flavonoids from the yellow green cocoon shell (Sasamayu) of the silkworm, Bombyx mori. Biosci. Biotechnol. Biochem. 2002, 66, 1396–1399. [Google Scholar] [CrossRef] [PubMed]
  38. Bungthong, C.; Siriamornpun, S. Changes in amino acid profiles and bioactive compounds of Thai silk cocoons as affected by water extraction. Molecules 2021, 26, 2033. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, H.-Y.; Zhao, J.-G.; Wei, Z.-G.; Zhang, Y.-Q. The renal protection of flavonoid-rich ethanolic extract from silkworm green cocoon involves in inhibiting TNF-α-p38 MAP kinase signalling pathway in type 2 diabetic mice. Biomed. Pharmacother. 2019, 118, 109379. [Google Scholar] [CrossRef]
  40. Kaur, J.; Rajkhowa, R.; Tsuzuki, T.; Wang, X. Crystals in Antheraea assamensis silkworm cocoon: Their removal, recovery and roles. Mater. Des. 2015, 88, 236–244. [Google Scholar] [CrossRef]
  41. Wang, Z.; Guo, P.; Wang, Q.; Zhang, Y.; Xu, H.; Zhao, P. Overexpression of Gloverin2 in the Bombyx mori silk gland enhances cocoon/silk antimicrobial activity. Dev. Comp. Immunol. 2019, 98, 6–12. [Google Scholar] [CrossRef] [PubMed]
  42. Aznar-Cervantes, S.D.; Monteagudo Santesteban, B.; Cenis, J.L. Products of sericulture and their hypoglycemic action evaluated by using the silkworm, Bombyx mori (Lepidoptera: Bombycidae), as a model. Insects 2021, 12, 1059. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, J.-G.; Wang, H.-Y.; Wei, Z.-G.; Zhang, Y.-Q. Therapeutic effects of ethanolic extract from the green cocoon shell of silkworm Bombyx mori on type 2 diabetic mice and its hypoglycaemic mechanism. Toxicol. Res. 2019, 8, 407–420. [Google Scholar] [CrossRef] [PubMed]
  44. Jangir, R.N.; Jain, G.C. Diabetes mellitus induced impairment of male reproductive functions: A review. Curr. Diabetes Rev. 2014, 10, 147–157. [Google Scholar] [CrossRef] [PubMed]
  45. Ahi, S.; Ebrahimi, F.; Abedi, H.A.; Kargar Jahromi, H.; Zarei, S. The effects of hydroalcoholic extract of silk cocoon on hypothalamic-pituitary–gonadal axis in streptozotocin-induced diabetic male rats. Autoimmune Dis. 2022, 2022, 7916159. [Google Scholar] [CrossRef] [PubMed]
  46. Ahsan, F.; Ansari, T.M.; Usmani, S.; Bagga, P. An insight on silk protein sericin: From processing to biomedical application. Drug Res. 2018, 68, 317–327. [Google Scholar] [CrossRef] [PubMed]
  47. Zhou, B.; Tian, R. Mitochondrial dysfunction in pathophysiology of heart failure. J. Clin. Investig. 2018, 128, 3716–3726. [Google Scholar] [CrossRef] [PubMed]
  48. Rujimongkon, K.; Ampawong, S.; Isarangkul, D.; Reamtong, O.; Aramwit, P. Sericin-mediated improvement of dysmorphic cardiac mitochondria from hypercholesterolaemia is associated with maintaining mitochondrial dynamics, energy production, and mitochondrial structure. Pharm. Biol. 2022, 60, 708–721. [Google Scholar] [CrossRef] [PubMed]
  49. Tarique, M.; Badruddeen; Ahsan, F.; Akhtar, J.; Khan, M.I.; Khalid, M. Formulation development and pharmacological evaluation of fixed dose combination of Bombyx mori coccon shell extract, Flaxseed oil and coenzyme Q10 against doxorubicin induced cardiomyopathy in rats. Orient. Pharm. Exp. Med. 2019, 19, 469–483. [Google Scholar] [CrossRef]
  50. Nasiruddin, M.; Ahmad, N.; Tajuddin. Effect of an unani formulation on lipid profile in rat. Indian J. Pharmacol. 2006, 38, 56–57. [Google Scholar] [CrossRef]
  51. Ali, M.M.; Arumugam, S.B. Effect of crude extract of Bombyx mori coccoons in hyperlipidemia and atherosclerosis. J. Ayurveda Integr. Med. 2011, 2, 72–78. [Google Scholar] [CrossRef] [PubMed]
  52. Okazaki, Y.; Kakehi, S.; Xu, Y.; Tsujimoto, K.; Sasaki, M.; Ogawa, H.; Kato, N. Consumption of sericin reduces serum lipids, ameliorates glucose tolerance and elevates serum adiponectin in rats fed a high-fat diet. Biosci. Biotechnol. Biochem. 2010, 74, 1534–1538. [Google Scholar] [CrossRef] [PubMed]
  53. Soares, A.; Beraldi, E.J.; Ferreira, P.E.B.; Bazotte, R.B.; Buttow, N.C. Intestinal and neuronal myenteric adaptations in the small intestine induced by a high-fat diet in mice. BMC Gastroenterol. 2015, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  54. Jia, X.; Pei, M.; Zhao, X.; Tian, K.; Zhou, T.; Liu, P. PEGylated oxidized alginate-DOX prodrug conjugate nanoparticles cross-linked with fluorescent carbon dots for tumor theranostics. ACS Biomater. Sci. Eng. 2016, 2, 1641–1648. [Google Scholar] [CrossRef] [PubMed]
  55. Matsumoto, H.; Ogura, H.; Shimizu, K.; Ikeda, M.; Hirose, T.; Matsuura, H.; Kang, S.; Takahashi, K.; Tanaka, T.; Shimazu, T. The clinical importance of a cytokine network in the acute phase of sepsis. Sci. Rep. 2018, 8, 13995. [Google Scholar] [CrossRef] [PubMed]
  56. Aramwit, P.; Towiwat, P.; Srichana, T. Anti-inflammatory potential of silk sericin. Nat. Prod. Commun. 2013, 8, 501–504. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, H.-Y.; Zhao, J.-G.; Zhang, Y.-Q. The flavonoid-rich ethanolic extract from the green cocoon shell of silkworm has excellent antioxidation, glucosidase inhibition, and cell protective effects in vitro. Food Nutr. Res. 2020, 64, 1637. [Google Scholar] [CrossRef]
  58. Tsvetkova, M.; Hristova-Avakumova, N.; Atanasova, L.; Panayotov, M.; Hadjimitova, V. Effect of extraction conditions on the antioxidant activity of diazo cocoon extracts effect of extraction conditions on the antioxidant activity of diazo cocoon extracts. In Proceedings of the 10th Jubilee International Conference of the Balkan Physical Union, Sofia, Bulgaria, 26–30 August 2018. [Google Scholar]
  59. Kumar, J.P.; Mandal, B.B. Inhibitory role of silk cocoon extract against elastase, hyaluronidase and UV radiation-induced matrix metalloproteinase expression in human dermal fibroblasts and keratinocytes. Photochem. Photobiol. Sci. 2019, 18, 1259–1274. [Google Scholar] [CrossRef] [PubMed]
  60. Li, Y.; Zhao, P.; Liu, H.; Guo, X.; He, H.; Zhu, R.; Xiang, Z.; Xia, Q. TIL-type protease inhibitors may be used as targeted resistance factors to enhance silkworm defenses against invasive fungi. Insect Biochem. Mol. Biol. 2015, 57, 11–19. [Google Scholar] [CrossRef]
  61. Zhang, X.; Guo, K.; Dong, Z.; Chen, Z.; Zhu, H.; Zhang, Y.; Xia, Q.; Zhao, P. Kunitz-type protease inhibitor BmSPI51 plays an antifungal role in the silkworm cocoon. Insect Biochem. Mol. Biol. 2020, 116, 103258. [Google Scholar] [CrossRef]
  62. Kaewkorn, W.; Limpeanchob, N.; Tiyaboonchai, W.; Pongcharoen, S.; Sutheerawattananonda, M. Effects of silk sericin on the proliferation and apoptosis of colon cancer cells. Biol. Res. 2012, 45, 45–50. [Google Scholar] [CrossRef] [PubMed]
  63. Micheli, L.; Parisio, C.; Lucarini, E.; Carrino, D.; Ciampi, C.; Toti, A.; Ferrara, V.; Pacini, A.; Ghelardini, C.; Di Cesare Mannelli, L. Restorative and pain-relieving effects of fibroin in preclinical models of tendinopathy. Biomed. Pharmacother. 2022, 148, 112693. [Google Scholar] [CrossRef] [PubMed]
  64. Li, C.; Yang, M.; Zhu, L.; Zhu, Y. Honeysuckle flowers extract loaded Bombyx mori silk fibroin films for inducing apoptosis of HeLa cells. Microsc. Res. Tech. 2017, 80, 1297–1303. [Google Scholar] [CrossRef]
  65. Lin, M.-J.; Lu, M.-C.; Chang, H.-Y. Sustained Release of Insulin-Like Growth Factor-1 from Bombyx mori L. Silk fibroin delivery for diabetic wound therapy. Int. J. Mol. Sci. 2021, 22, 6267. [Google Scholar] [CrossRef]
  66. Montalbán, M.G.; Coburn, J.M.; Lozano-Pérez, A.A.; Cenis, J.L.; Víllora, G.; Kaplan, D.L. Production of curcumin-loaded silk fibroin nanoparticles for cancer therapy. Nanomaterials 2018, 8, 126. [Google Scholar] [CrossRef] [PubMed]
  67. Holanda, F.H.; Ribeiro, A.N.; Sánchez-Ortiz, B.L.; de Souza, G.C.; Borges, S.F.; Ferreira, A.M.; Florentino, A.C.; Yoshioka, S.A.; Moraes, L.S.; Carvalho, J.C.T.; et al. Anti-inflammatory potential of baicalein combined with silk fibroin protein in a zebrafish model (Danio rerio). Biotechnol. Lett. 2023, 45, 235–253. [Google Scholar] [CrossRef]
  68. Liu, J.; Sun, H.; Peng, Y.; Chen, L.; Xu, W.; Shao, R. Preparation and Characterization of natural silk fibroin hydrogel for protein drug delivery. Molecules 2022, 27, 3418. [Google Scholar] [CrossRef]
  69. Wang, S.; Zhu, M.; Zhao, L.; Kuang, D.; Kundu, S.C.; Lu, S. Insulin-loaded silk fibroin microneedles as sustained release system. ACS Biomater. Sci. Eng. 2019, 5, 1887–1894. [Google Scholar] [CrossRef]
  70. Stinson, J.A.; Raja, W.K.; Lee, S.; Kim, H.B.; Diwan, I.; Tutunjian, S.; Panilaitis, B.; Omenetto, F.G.; Tzipori, S.; Kaplan, D.L. Silk fibroin microneedles for transdermal vaccine delivery. ACS Biomater. Sci. Eng. 2017, 3, 360–369. [Google Scholar] [CrossRef]
  71. Wang, Z.; Yang, Z.; Jiang, J.; Shi, Z.; Mao, Y.; Qin, N.; Tao, T.H. Silk microneedle patch capable of on-demand multidrug delivery to the brain for glioblastoma treatment. Adv. Mater. 2022, 34, 2106606. [Google Scholar] [CrossRef]
  72. Yin, Z.; Kuang, D.; Wang, S.; Zheng, Z.; Yadavalli, V.K.; Lu, S. Swellable silk fibroin microneedles for transdermal drug delivery. Int. J. Biol. Macromol. 2018, 106, 48–56. [Google Scholar] [CrossRef] [PubMed]
  73. Lujerdean, C.; Baci, G.-M.; Cucu, A.-A.; Dezmirean, D.S. The contribution of silk fibroin in biomedical engineering. Insects 2022, 13, 286. [Google Scholar] [CrossRef] [PubMed]
  74. Bucciarelli, A.; Motta, A. Use of Bombyx mori silk fibroin in tissue engineering: From cocoons to medical devices, challenges, and future perspectives. Mater. Sci. Eng. C 2022, 139, 212982. [Google Scholar] [CrossRef] [PubMed]
  75. Mao, Z.; Bi, X.; Ye, F.; Shu, X.; Sun, L.; Guan, J.; Ritchie, R.O.; Wu, S. Controlled Cryogelation and catalytic cross-linking yields highly elastic and robust silk fibroin scaffolds. ACS Biomater. Sci. Eng. 2020, 6, 4512–4522. [Google Scholar] [CrossRef] [PubMed]
  76. Saleem, M.; Rasheed, S.; Yougen, C. Silk fibroin/hydroxyapatite scaffold: A highly compatible material for bone regeneration. Sci. Technol. Adv. Mater. 2020, 21, 242–266. [Google Scholar] [CrossRef] [PubMed]
  77. Mejia, M.L.; Moncada, M.E.; Ossa-Orozco, C.P. Poly (vinyl alcohol)/Silk Fibroin/Ag NPs composite nanofibers for bone tissue engineering. In Proceedings of the 2021 43rd Annual International Conference of the IEEE Engineering in Medicine & Biology Society (EMBC), Guadalajara, Mexico, 1–5 November 2021; pp. 1176–1180. [Google Scholar] [CrossRef] [PubMed]
  78. Jiang, P.; Ran, J.; Yan, P.; Zheng, L.; Shen, X.; Tong, H. Rational design of a high-strength bone scaffold platform based on in situ hybridization of bacterial cellulose/nano-hydroxyapatite framework and silk fibroin reinforcing phase. J. Biomater. Sci. Polym. Ed. 2018, 29, 107–124. [Google Scholar] [CrossRef] [PubMed]
  79. Asadpour, S.; Kargozar, S.; Moradi, L.; Ai, A.; Nosrati, H.; Ai, J. Natural biomacromolecule based composite scaffolds from silk fibroin, gelatin and chitosan toward tissue engineering applications. Int. J. Biol. Macromol. 2020, 154, 1285–1294. [Google Scholar] [CrossRef]
  80. Bon, S.B.; Chiesa, I.; Degli Esposti, M.; Morselli, D.; Fabbri, P.; De Maria, C.; Morabito, A.; Coletta, R.; Calamai, M.; Pavone, F.S.; et al. Carbon nanotubes/regenerated silk composite as a three-dimensional printable bio-adhesive ink with self-powering properties. ACS Appl. Mater. Interfaces 2021, 13, 21007–21017. [Google Scholar] [CrossRef]
  81. Chakraborty, J.; Mu, X.; Pramanick, A.; Kaplan, D.L.; Ghosh, S. Recent advances in bioprinting using silk protein-based bioinks. Biomaterials 2022, 287, 121672. [Google Scholar] [CrossRef]
  82. Mu, X.; Gonzalez-Obeso, C.; Xia, Z.; Sahoo, J.K.; Li, G.; Cebe, P.; Zhang, Y.S.; Kaplan, D.L. 3D printing of monolithic proteinaceous cantilevers using regenerated silk fibroin. Molecules 2022, 27, 2148. [Google Scholar] [CrossRef] [PubMed]
  83. Sanz-Fraile, H.; Amoros, S.; Mendizabal, I.; Galvez-Monton, C.; Prat-Vidal, C.; Bayes-Genis, A.; Navajas, D.; Farre, R.; Otero, J. Silk-reinforced collagen hydrogels with raised multiscale stiffness for mesenchymal cells 3D culture. Tissue Eng. Part A 2020, 26, 358–370. [Google Scholar] [CrossRef] [PubMed]
  84. Qu, X.; Yan, L.; Liu, S.; Tan, Y.; Xiao, J.; Cao, Y.; Chen, K.; Xiao, W.; Li, B.; Liao, X. Preparation of silk fibroin/hyaluronic acid hydrogels with enhanced mechanical performance by a combination of physical and enzymatic crosslinking. J. Biomater. Sci. Polym. Ed. 2021, 32, 1635–1653. [Google Scholar] [CrossRef] [PubMed]
  85. Khosropanah, M.H.; Vaghasloo, M.A.; Shakibaei, M.; Mueller, A.L.; Kajbafzadeh, A.M.; Amani, L.; Haririan, I.; Azimzadeh, A.; Hassannejad, Z.; Zolbin, M.M. Biomedical applications of silkworm (Bombyx mori) proteins in regenerative medicine (a narrative review). J. Tissue Eng. Regen. Med. 2022, 16, 91–109. [Google Scholar] [CrossRef] [PubMed]
  86. Manjubala, I.; Akila, R.; Priya, G.; Anitha, R.; Madhan, B.; Narendrakumar, U. Fabrication of biocomposite sheets from silk cocoons for tissue engineering applications. Int. J. Nanotechnol. 2021, 18, 561–571. [Google Scholar] [CrossRef]
  87. Lam, Y.T.; Tan, R.P.; Michael, P.L.; Lau, K.; Yang, N.; Rnjak-Kovacina, J.; Wise, S.G. Bioengineering silk into blood vessels. Biochem. Soc. Trans. 2021, 49, 2271–2286. [Google Scholar] [CrossRef] [PubMed]
  88. Gupta, P.; Lorentz, K.L.; Haskett, D.G.; Cunnane, E.M.; Ramaswamy, A.K.; Weinbaum, J.S.; Vorp, D.A.; Mandal, B.B. Bioresorbable silk grafts for small diameter vascular tissue engineering applications: In vitro and in vivo functional analysis. Acta Biomater. 2020, 105, 146–158. [Google Scholar] [CrossRef] [PubMed]
  89. Balcão, V.M.; Harada, L.K.; Jorge, L.R., Jr.; Oliveira, J.M.; Tubino, M.; Vila, M.M.D.C. Structural and functional stabilization of sericin from Bombyx mori cocoons in a biopolysaccharide film: Bioorigami for skin regeneration. J. Braz. Chem. Soc. 2020, 31, 833–848. [Google Scholar] [CrossRef]
  90. Yang, C.; Chen, S.; Su, H.; Zhang, H.; Tang, J.; Guo, C.; Song, F.; Zhang, W.; Gu, J.; Liu, Q. Biocompatible, small-sized and well-dispersed gold nanoparticles regulated by silk fibroin fiber from Bombyx mori cocoons. Front. Mater. Sci. 2019, 13, 126–132. [Google Scholar] [CrossRef]
  91. Hu, F.X.; Xie, X.; Wang, D.; Bin Yang, H.; Gu, Y.; Chen, B.; Zhang, C.; Rao, Q.; Li, Q.; Guo, C. Three-dimensional cell-adhesive matrix of silk cocoon derived carbon fiber assembled with iron-porphyrin for monitoring cell released signal molecules. Sens. Actuators B Chem. 2021, 334, 129594. [Google Scholar] [CrossRef]
  92. Wang, H.; Duan, S.; Chen, Y.; Liu, H.; Tian, J.; Wu, F.; Du, Z.; Tang, L.; Li, Y.; Ding, S. Study on a natural silk cocoon membrane-based versatile and stable immunosensing platform via directional immunoaffinity recognition. ACS Omega 2022, 7, 35297–35304. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, H.; Duan, S.; Wang, M.; Wei, S.; Chen, Y.; Chen, W.; Li, Y.; Ding, S. Silk cocoon membrane-based immunosensing assay for red blood cell antigen typing. Sens. Actuators B Chem. 2020, 320, 128376. [Google Scholar] [CrossRef]
  94. Lau, K.; Waterhouse, A.; Akhavan, B.; Gao, L.; Na Kim, H.; Tang, F.; Whitelock, J.M.; Bilek, M.M.; Lord, M.S.; Rnjak-Kovacina, J. Biomimetic silk biomaterials: Perlecan-functionalized silk fibroin for use in blood-contacting devices. Acta Biomater. 2021, 132, 162–175. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, Z.-L.; Shen, Y.-H.; Sun, X.; Li, Z.-H.; Wang, X.-Y.; Zhao, Z. High biocompatible AuNCs-silk fibroin hydrogel system for visual detection of H2O2. Microchem. J. 2020, 157, 105036. [Google Scholar] [CrossRef]
  96. Available online: www.clinicaltrials.gov (accessed on 13 November 2023).
  97. Louiselle, A.E.; Niemiec, S.; Azeltine, M.; Mundra, L.; French, B.; Zgheib, C.; Liechty, K.W. Evaluation of skin care concerns and patient’s perception of the effect of NanoSilk Cream on facial skin. J. Cosmet. Dermatol. 2022, 21, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
  98. Egan, G.; Phuagkhaopong, S.; Matthew, S.A.L.; Connolly, P.; Seib, F.P. Impact of silk hydrogel secondary structure on hydrogel formation, silk leaching and in vitro response. Sci. Rep. 2022, 12, 3729. [Google Scholar] [CrossRef] [PubMed]
  99. Brown, J.E.; Gulka, C.P.; Giordano, J.E.M.; Montero, M.P.; Hoang, A.; Carroll, T.L. Injectable silk protein microparticle-based fillers: A novel material for potential use in glottic insufficiency. J. Voice 2019, 33, 773–780. [Google Scholar] [CrossRef] [PubMed]
  100. Kijanska, M.; Marmaras, A.; Hegglin, A.; Kurtcuoglu, V.; Giovanoli, P.; Lindenblatt, N. In vivo characterization of the integration and vascularization of a silk-derived surgical scaffold. JPRAS J. Plast. Reconstr. Aesthetic Surg. 2016, 69, 1141–1150. [Google Scholar] [CrossRef] [PubMed]
  101. Lee, J.H.; Kim, D.-K.; Park, H.S.; Jeong, J.Y.; Yeon, Y.K.; Kumar, V.; Bae, S.H.; Lee, J.M.; Moon, B.M.; Park, C.H. A prospective cohort study of the silk fibroin patch in chronic tympanic membrane perforation. Laryngoscope 2016, 126, 2798–2803. [Google Scholar] [CrossRef]
  102. Lee, J.H.; Lee, J.S.; Kim, D.-K.; Park, C.H.; Lee, H.R. Clinical outcomes of silk patch in acute tympanic membrane perforation. Clin. Exp. Otorhinolaryngol. 2015, 8, 117–122. [Google Scholar] [CrossRef]
  103. Mazurek, Ł.; Szudzik, M.; Rybka, M.; Konop, M. Silk fibroin biomaterials and their beneficial role in skin wound healing. Biomolecules 2022, 12, 1852. [Google Scholar] [CrossRef] [PubMed]
  104. Yigit, S.; Hallaj, N.S.; Sugarman, J.L.; Chong, L.C.; Roman, S.E.; Abu-Taleb, L.M.; Goodman, R.E.; Johnson, P.E.; Behrens, A.M. Toxicological assessment and food allergy of silk fibroin derived from Bombyx mori cocoons. Food Chem. Toxicol. 2021, 151, 112117. [Google Scholar] [CrossRef] [PubMed]
  105. Qin, H.; Zhang, J.; Yang, H.; Yao, S.; He, L.; Liang, H.; Wang, Y.; Chen, H.; Zhao, P.; Qin, G. Safety assessment of water-extract sericin from silkworm (Bombyx mori) cocoons using different model approaches. BioMed Res. Int. 2020, 2020, 9689386. [Google Scholar] [CrossRef] [PubMed]
  106. Acharya, C.; Ghosh, S.K.; Kundu, S.C. Silk fibroin protein from mulberry and non-mulberry silkworms: Cytotoxicity, biocompatibility and kinetics of L929 murine fibroblast adhesion. J. Mater. Sci. Mater. Med. 2008, 19, 2827–2836. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The application of SC-derived materials in drug delivery systems.
Figure 1. The application of SC-derived materials in drug delivery systems.
Pharmaceuticals 17 00817 g001
Figure 2. The preparation process for vaccine-coated silk fibroin microneedles. (A) Silk microneedles were demolded, annealed with water, and trimmed to create the final uncoated constructs. (B) For uniform coating of silk microneedles with vaccine-containing solution, the devices were pre-wetted with water droplets.
Figure 2. The preparation process for vaccine-coated silk fibroin microneedles. (A) Silk microneedles were demolded, annealed with water, and trimmed to create the final uncoated constructs. (B) For uniform coating of silk microneedles with vaccine-containing solution, the devices were pre-wetted with water droplets.
Pharmaceuticals 17 00817 g002
Figure 3. The application of SC-derived materials in tissue engineering.
Figure 3. The application of SC-derived materials in tissue engineering.
Pharmaceuticals 17 00817 g003
Figure 4. Schematic diagram of a solvent-free method for preparing a bio-ink of SF extracted from SC.
Figure 4. Schematic diagram of a solvent-free method for preparing a bio-ink of SF extracted from SC.
Pharmaceuticals 17 00817 g004
Figure 5. The application of SC-derived materials in regenerative medicine.
Figure 5. The application of SC-derived materials in regenerative medicine.
Pharmaceuticals 17 00817 g005
Figure 6. The application of SC-derived materials in in vitro diagnosis.
Figure 6. The application of SC-derived materials in in vitro diagnosis.
Pharmaceuticals 17 00817 g006
Table 1. Main bioactive compounds extracted from SC.
Table 1. Main bioactive compounds extracted from SC.
Main CompoundExtraction MethodContent
silk fibroinsoda boiling process (degumming)70–80%
silk sericinhydrothermal method;
chemical method;
enzymatic method
20–30%
flavonoidssolvent extraction process<1%
calcium oxalateacid dissolution method
other peptidesenzymolysis method
Table 3. Clinical trials of silk fibroin products [96].
Table 3. Clinical trials of silk fibroin products [96].
Official TitleSponsorEnrollmentObjectiveDosageStudy PhaseConditionLocationCurrent Status
A Comparative Evaluation of Subgingivally Delivered Chlorhexidine, Silk Fibroin and Combination of Fibroin and Chlorhexidine as Local Drug Delivery in Periodontitis—A Randomized Control TrialKrishnadevaraya College of Dental Sciences & Hospital15 (Estimated)To evaluate the effect of silk fibroin as a drug delivery system while simultaneously assessing the efficacy of silk fibroin in comparison to chlorhexidineFilmsPhase 1Periodontal PocketNot providedNot Yet Recruiting
Efficacy and Safety of Wound Dressing Containing Silk Fibroin With Bioactive Coating Layer Versus Medicated Paraffin Gauze Dressing in the Treatment of Split-thickness Skin Graft Donor SitesChulalongkorn University29
(Actual)
To compare wound dressing containing silk fibroin with bioactive coating layer with Bactigras®, with regard to healing time, patients’ pain intensity, skin’s transepidermal water loss after healing and evidence of infection in the treatment of split-thickness skin graft donor sitesFilmsPhase 1
Phase 2
Impaired Wound Healing;
Infection of Skin Donor Site;
Late Complication From Skin Graft;
Intractable Pain
ThailandCompleted
Manufacturing, Characterization and Evaluation of the Effect of Silk Fibroin Membranes, Loaded or Not With Neurotensins on Open Wounds in the Palate: Randomized Clinical StudyUniversidade Estadual Paulista Júlio de Mesquita Filho66 (Estimated)To manufacture and characterize silk fibroin membranes loaded or not with neurotensin and to evaluate clinical, patient-centered, and immunological parameters to determine the effect of using these membranes on open wounds on the human palateFilmsNot ApplicableWound Healing;
Palate Wound
BrazilRecruiting
A New Drug Delivery System—Silk Fibroin Film Loaded or Not With Insulin on Palatal Mucosa Wound Healing: in Vitro Study and a Randomized Clinical TrialUniversidade Estadual Paulista Júlio de Mesquita Filho75 (Estimated)To evaluate the effect of silk fibroin films loaded or not with insulin in the repair of palatal mucosa open woundsFilmsNot ApplicableWound Healing;
Palate Wound
BrazilUnknown
A Pilot Study to Evaluate the Reconstruction of Digital Nerve Defects in Humans Using an Implanted
Silk Nerve Guide
Silk Biomaterials srl4
(Actual)
To ascertain the feasibility and safety of the procedure using SilkBridge—a biocompatible silk fibroin-based scaffold—for the regeneration of sensory nerve fibersScaffoldNot ApplicablePeripheral Nerve Injury Digital Nerve HandSwitzerlandUnknown
NanoSilk Cosmo: Evaluation of a Novel Silk Complex on Biophysical Parameters Related to Skin AgingUniversity of Colorado, Denver46
(Actual)
To evaluate a novel silk complex on biophysical parameters related to skin aging including skin resilience, elasticity, and hydrationNanosolutionNot ApplicableAgingUnited StatesCompleted
Multi-center, Randomized, Active-controlled, Single-blind, Parallel Two-group Trial of HQ® Matrix Soft Tissue Mesh and ULTRAPRO® Partially Absorbable Lightweight Mesh for the Treatment of Inguinal HerniaZhejiang Xingyue Biotechnology Co., Ltd.144 (Estimated)To evaluate the safety and effectiveness of HQ® Matrix Soft Tissue Mesh for the Treatment of Inguinal HerniaScaffoldNot ApplicableInguinal HerniaChinaUnknown
Pilot Evaluation of Cosmetic Outcome and Surgical Site Infection Rates of Coated VICRYL* Plus Antibacterial (Polyglactin 910) Suture Compared to Chinese Silk in Scheduled Breast Cancer SurgeryEthicon, Inc.101
(Actual)
To evaluate the cosmetic outcome and surgical site infection in approximately 100 patients from 6 centers in China undergoing scheduled modified radical mastectomy for breast cancerSilk SuturePhase 4Breast CancerChinaCompleted
Table 4. Silk fibroin products on the market.
Table 4. Silk fibroin products on the market.
Product NameMain CompositionsIndicationsApproval YearNation
Silk Voice®A silk fibroin injectionTo treat vocal cord-mediated and vocal cord dysfunction.2018USA
SERI®Surgical stent device based on silk fibroinAbdominal wall reconstruction and plastic surgery.UnknownUSA
Tympasil®A silk fibroin patchTo treat ear drum perforations.UnknownKorea
Antibacterial wound dressing patchA silk fibroin patchPostoperative incisions, skin surface abrasions, and ulcer coverage.2020China
Functional healing wound dressing patchSilk fibroin, surface coated with a composite silicon-based powderPromoting healing, repair, and coverage of postoperative wounds, abrasions, and non-healing wounds.2021China
Absorbable silk fibroin repair filmComposed of silk fibroin, glycerol, and waterUsed in conjunction with bone meal as a physical barrier for preserving the extraction site of adult patients after tooth extraction.2022China
Silk fibroin film dressingSilk fibroin with an amino acid content of ≥90%For skin area coverage.2020China
Silk fibroin vaginal packing gelGel consists of silk fibroin, carbomer, triethanolamine, sodium ethyl paraben, glycerin, and purified waterTo block HPV infection in the reproductive tract and prevent cervical lesions caused by HPV infection. To improve the vaginal microenvironment, alleviate itching, pain, congestion, edema, increased secretion, purulent discharge symptoms caused by chronic cervicitis, and reduce the surface of cervical erosion.2023China
Silk fibroin hydrogel dressingGel composed of silk fibroin and purified waterCoverage and care of non-chronic wounds after laser surgery.2022China
Light guide gelComposed of carbomer 940, glycerol, silk fibroin, pentanediol, sodium hydroxide, and purified waterFor thermal insulation and light guidance during photon therapy, in conjunction with photon therapy equipment.2022China
Liquid dressingA solution composed of silk fibroin, sodium alginate, sodium carboxymethyl cellulose, glycerol, and purified waterFor the care of superficial wounds and surrounding skin such as small wounds, abrasions, and cuts.2021China
Liquid wound dressingA solution composed of silk fibroin, sodium chloride, glycerol, and carbomerFor the care of superficial wounds and surrounding skin such as small wounds, abrasions, and cuts.2020China
Liquid dressingA solution composed of silk fibroin, glycerol, sodium benzoate, potassium sorbate, and carboxymethyl celluloseFor the care of superficial wounds and surrounding skin such as small wounds, abrasions, and cuts.2020China
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tian, Z.; Zhao, C.; Huang, T.; Yu, L.; Sun, Y.; Tao, Y.; Cao, Y.; Du, R.; Lin, W.; Zeng, J. Silkworm Cocoon: Dual Functions as a Traditional Chinese Medicine and the Raw Material of Promising Biocompatible Carriers. Pharmaceuticals 2024, 17, 817. https://doi.org/10.3390/ph17070817

AMA Style

Tian Z, Zhao C, Huang T, Yu L, Sun Y, Tao Y, Cao Y, Du R, Lin W, Zeng J. Silkworm Cocoon: Dual Functions as a Traditional Chinese Medicine and the Raw Material of Promising Biocompatible Carriers. Pharmaceuticals. 2024; 17(7):817. https://doi.org/10.3390/ph17070817

Chicago/Turabian Style

Tian, Zhijie, Chuncao Zhao, Ting Huang, Lining Yu, Yijie Sun, Yian Tao, Yunfeng Cao, Ruofei Du, Wenhui Lin, and Jia Zeng. 2024. "Silkworm Cocoon: Dual Functions as a Traditional Chinese Medicine and the Raw Material of Promising Biocompatible Carriers" Pharmaceuticals 17, no. 7: 817. https://doi.org/10.3390/ph17070817

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop