Next Article in Journal
Design and Synthesis of (Z)-2-(Benzylamino)-5-benzylidenethiazol-4(5H)-one Derivatives as Tyrosinase Inhibitors and Their Anti-Melanogenic and Antioxidant Effects
Next Article in Special Issue
Improvement of the Chemical Reactivity of Michael Acceptor of Ethacrynic Acid Correlates with Antiproliferative Activities
Previous Article in Journal
ISIDORE, a Probe for In Situ Trace Metal Speciation Based on the Donnan Membrane Technique and Electrochemical Detection Part 2: Cd and Pb Measurements during the Accumulation Time of the Donnan Membrane Technique
Previous Article in Special Issue
Beta-Caryophyllene Enhances the Anti-Tumor Activity of Cisplatin in Lung Cancer Cell Lines through Regulating Cell Cycle and Apoptosis Signaling Molecules
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sericultural By-Products: The Potential for Alternative Therapy in Cancer Drug Design

by
Gabriela-Maria Baci
*,
Ecaterina-Daniela Baciu
,
Alexandra-Antonia Cucu
,
Adriana-Sebastiana Muscă
,
Alexandru Ioan Giurgiu
,
Adela Ramona Moise
,
Marius Zăhan
and
Daniel Severus Dezmirean
*
Faculty of Animal Science and Biotechnology, University of Animal Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(2), 850; https://doi.org/10.3390/molecules28020850
Submission received: 24 November 2022 / Revised: 10 January 2023 / Accepted: 11 January 2023 / Published: 14 January 2023
(This article belongs to the Special Issue Molecular Targets and Mechanisms of Action of Anti-cancer Agents)

Abstract

:
Major progress has been made in cancer research; however, cancer remains one of the most important health-related burdens. Sericulture importance is no longer limited to the textile industry, but its by-products, such as silk fibroin or mulberry, exhibit great impact in the cancer research area. Fibroin, the pivotal compound that is found in silk, owns superior biocompatibility and biodegradability, representing one of the most important biomaterials. Numerous studies have reported its successful use as a drug delivery system, and it is currently used to develop three-dimensional tumor models that lead to a better understanding of cancer biology and play a great role in the development of novel antitumoral strategies. Moreover, sericin’s cytotoxic effect on various tumoral cell lines has been reported, but also, it has been used as a nanocarrier for target therapeutic agents. On the other hand, mulberry compounds include various bioactive elements that are well known for their antitumoral activities, such as polyphenols or anthocyanins. In this review, the latest progress of using sericultural by-products in cancer therapy is discussed by highlighting their notable impact in developing novel effective drug strategies.

1. Introduction

Worldwide, cancer is one of the major health and economic burdens. Even if significant progress has been made in cancer research and there have been described new treatments, nevertheless, cancer continues to be one of the leading causes of death [1,2,3,4]. With the purpose to overcome specific limitations of the current cancer treatments, there have been numerous biomaterials developed, such as polymers, ceramics, or biologically derived materials. Among these, the most used in the process of fighting cancer, due to their high biocompatibility, are biologically derived materials [5]. Using biomaterials to combat cancer, numerous studies have shown great progress in reducing chemotherapeutics’ toxicity, expanding its half-life, and also in improving anticancer drugs’ solubility [6]. Moreover, in numerous experiments, biomaterials are used to develop tumor microenvironment models to elucidate certain cancer mechanisms that would lead to the development of novel therapies [7].
Another promising strategy to prevent and to treat cancer is the use of specific plants and plant-derived products as anticancer agents. This strategy also includes the exploitation of the chemoprotective activity of a wide range of plants to increase the cancer patient’s life quality [8]. The most important plant-derived compound that is currently used as a first-line treatment in different types of cancer is paclitaxel, which is extracted from Taxus chinensis [9,10,11]. Furthermore, this strategy has brought into the spotlight a broad range of plants that anticancer activity has been certified, for instance: Curcuma longa, Artemisia annua, Peganum harmala [10], and Morus alba [12,13], just to mention a few.
Nowadays, one of the most important agro-based industries is sericulture. The main activity of sericulture (Bombyx mori) is the rearing of silkworms in order to obtain raw silk. For example, the rearing of silkworms (Antheraea pernyi) in China is to obtain pupae and moths for food and raw silk for textiles [14]. However, sericulture (B. mori) also involves the cultivation of food plants to provide silkworms’ nourishment source. Furthermore, it includes silk yarn extraction and a wide range of post-cocoon activities [15,16]. There are several species of silkworms that have distinct nutritional requirements and produce different types of silk. By far, among the silkworm species, B. mori exhibits the greatest importance, not only in terms of the silk industry but it displays an extraordinary impact in the life science fields [15,17,18,19]. Along with Apis mellifera and Drosophila melanogaster [20], the silkworm, B. mori, represents one of the most important insects that exhibits considerable interest for the scientific community, being extensively studied in worldwide research laboratories [21]. Its potential in life science areas began to be massively exploited in 2004 when its draft genome sequence was published [22]. One of the most important roles of B. mori in the scientific fields is that of a model organism and bioreactor. It represents a valuable model organism for various human diseases due to the fact that its genetic information is well documented [23]. In addition, the silkworm’s genome contains a meaningful percent of human ortholog genes, and there are various mutant strains described [21].
However, the scientific community’s interest in silkworms is also highlighted by the key role of silk fibroin (SF) as a pivotal biomaterial [24]. Fibroin is the main compound of the silk produced by B. mori. It possesses certain properties, such as superior biocompatibility, biodegradability, and remarkable mechanical features, that have led to the development of an extensive variety of SF-based biomaterials [25,26,27,28]. There have been various types of SF biomaterials described [29], such as hydrogels [30,31,32,33], scaffolds [34,35,36,37], and sponges [38,39,40,41]. On the other hand, numerous studies have reported the antitumoral effect of sericin and its impact as a nanocarrier biomaterial for anticancer therapeutic agents [42].
B. mori’s unique nourishment source is represented by mulberry leaves [15,18,43,44]. Nevertheless, due to its complex composition since ancient times, it has been recognized as a significant medicinal plant, and nowadays, mulberry represents one of the pivotal functional foods. Among mulberry ingredients, the most notable elements, which are responsible for a broad spectrum of bioactivities, are the phenolic compounds [45]. There are numerous epidemiologic studies that have certified mulberry plant’s medicinal value—more specifically, its anticancer, chemoprotective, antioxidative, cardioprotective, anti-obesity, and anti-inflammatory activities [45,46,47,48,49,50,51,52,53,54].
Nutrition directly influences larval growth, such as the economic traits, including filament length and fibroin content. It is well documented that the leaf quality and its nutritional value impact silk production [55]. Nowadays, by keeping in mind the leading role of SF as a biomaterial in cancer research and beyond, it is of keen interest to enhance the fibroin content. Considering this, the scientific community paid considerable attention to supplementing silkworms’ traditional food. For instance, Thangapandiyan and Dharanipriya (2019) [56] investigated the influence of the synergistic effect of spirulina and silver nanoparticles on a wide range of B. mori’s economic traits, including the SF and sericin contents. Their results showed that the chosen agents positively impacted the cocoon’s characters. In other studies, superior quality silk fiber was obtained by supplementing mulberry leaves with pollen [57], honey [58], lactic acid [59], palm sugar [60], micronutrients [61], or the Amway protein [62].
Keeping in mind the significant need of developing new strategies to combat cancer, it is of great importance to point at the progress and perspectives of the functional aspects of sericulture in cancer therapy. Herein, we discuss the latest progress that has been made in developing fibroin-based biomaterials to overcome certain limits of chemotherapeutic drugs but also in using this type of biomaterial to mimic the tumor microenvironment. Furthermore, we summarize the perspective of exploiting mulberry’s anticancer activity to develop novel cancer drug designs.

2. Sericultural By-Products

In recent years, the concept of sericulture has not been limited anymore to the silk industry. The great medicinal potential of silkworms (Figure 1), SF, and mulberry plants have brought sericulture to researchers’ attention [63].

2.1. Silk Fibroin as a Functional Biomaterial

One of the most appreciated naturally derived polymers, among cellulose and chitosan, is silk. Numerous insects produce this biopolymer; however, the most explored is the silk produced by the silkworm B. mori [64]. Owning extraordinary properties such as superior mechanical strength, flexibility, and breathability, it is one of the most important luxury fibers [65]. Moreover, the broad range of qualities have brought silk to researchers’ attention, and other great features of silk have been documented—more specifically, its great biocompatibility, versatility, and biodegradability. Based on these features, silk has been used as a surgical suture since ancient times [66,67]. With reference to the structural composition of B. mori’s silk, it includes two major players, namely fibroin and sericin. In the silk thread, there are two fibroin fibers, while sericin represents a glue-like protein for them [64]. Fibroin is responsible for silk’s superior properties; in opposition, in most cases, sericin is removed by certain thermos–chemical treatments [68]. At this time, fibroin is being explored by a wide range of researchers that develop various SF-based biomaterials, such as hydrogels, nanoparticles, scaffolds, microtubes, biofilms, and sponges (Figure 2), which exhibit great importance for the life science fields [69,70].
Gastrointestinal disorders are one of the most common chronic conditions; accordingly, there is an extensive need to prevent these diseases [71]. In this direction, one of the most approached strategies is represented by probiotics. In the past years, the use of probiotics met rapid enlargement, even though their history began centuries ago. Probiotics consist of living microorganisms that exhibit various benefits for human health [72,73], especially for certain gastrointestinal disorders. In addition, the health-promoting properties are not limited to gastrointestinal diseases, and it has been certified that probiotics are beneficial to prevent and to treat oral cavity and skin disorders [74]. However, specific tissues and organs exhibit certain limitations when it comes to probiotics’ viability and activity; specifically, there are disturbing factors such as oxidative stress or pH. The most used strategy to overcome the viability and activity limitations is the probiotics’ encapsulation by using biomaterials [75,76].
Recently, Kwon et al. (2021) [27] used SF as a coating biomaterial for various probiotics. By using the Coeuret’s method, the authors isolated the target bacterial from newborn infant feces, fermented fruits, and raw milk. Regarding the SF-coating preparation, the silk threads were firstly degummed to remove sericin, and subsequently, a treatment with two agents (Protmax and T100) was applied to enhance SF’s solubility but also its absorption rate. Primarily, the authors investigated if the addition of water-soluble calcium to SF-coated bacterial strains impacted its survival ratio. Additionally, to evaluate the effects of using SF as a coating biomaterial, the gastrointestinal conditions were stimulated to analyze the acid and bile tolerance. The SF-coated probiotics were exposed to simulated gastric fluid, which included HCl and NaOH for pH adjustments and 0.5% oxgall. The indirect method of evaluating the intestinal adhesion of probiotics implies the determination of cell surface hydrophobicity. Moreover, the researchers directly investigated probiotics’ intestinal adherence by using a human colorectal adenocarcinoma cell line. By evaluating the impact of several different concentrations of water-soluble calcium on a bacterial strain’s survival rate, their results revealed that the highest survival ratio was observed when adding 0.1% of this component. Successfully, by using SF as coating biomaterial for probiotics, the survival rate was positively impacted under a simulated gastrointestinal environment. When simulating the gastric fluid, the target parameter was enhanced by 13.3–31.3%, and regarding the simulated intestinal fluid, the bacterial survival rate was increased by 4.8–23.5%. Their data showed that SF coating led to a significant increase of the cell surface hydrophobicity; notably, the highest increase observed was up to 71.7%. Furthermore, when evaluating the cell adhesion capacity, the authors reported that it has been enhanced up to 36%. Their data highlight the positive effects of using SF as a coating biomaterial on probiotic bacterial stability. In the specialized literature, there are a broad range of recent studies that have reported the successful use of SF as a biomaterial for various purposes (Table 1).

2.2. Therapuetic Potential of Mulberry

In the healthcare system, a great number of plants are engaged as curative agents and functional foods, mainly due to the great number of bioactive compounds that are found in plants [98]. Furthermore, these organisms have received considerable attention, because various parts of the plants can be exploited for medical purposes, the most notable being the leaves, fruits, seeds, and roots. In addition, another great advantage when it comes to therapeutic herbs is their low cost [99,100].
Among all powerful therapeutic plants, such as Allium sativum [101], Citrus limon [102], and Vaccinium corymbosum [103], one of the richest plants in bioactive components is the mulberry (Morus spp.). A generous spectrum of recent studies highlighted the beneficial effects of various parts of the mulberry on human health [104,105]. Each botanical part of the mulberry tree exhibits certain therapeutic effects—more specifically, the mulberry fruits, leaves, roots, and twigs [106]. As for mulberry’s composition, it includes a great content of carbohydrates (7.8–9%) and proteins (5–1.4%) [104] but also lipids, fibers, vitamins, and minerals [107]. In the matter of the functional compounds, in Figure 3 are listed the main bioactive elements found in mulberries that are associated with powerful therapeutic effects. Notably, the chemical composition of the mulberry depends on the mulberry variety, climate, and soil condition [104].
Oxidative stress is one of the major threats in cellular damage being responsible for aging, cardiovascular diseases, cancer, and so on [108]. In this direction, a certain group of plants’ secondary metabolites, namely the phenolic compounds, have gained a lot of attention by playing a key role as antioxidative agents. These secondary metabolites are extensively present in various plant foods, such as apples, cranberries, and pears [109,110]. Another plant that exhibits a great number of phenolic compounds is the mulberry. Numerous studies have investigated the phenolic profiles of numerous varieties of the mulberry and confirmed its antioxidative activities [111,112,113,114].
In a recent study, Hu et al. (2021) [115] investigated wherever the mulberry growth period impacts the number of flavonoids in leaves. The authors also explored the influence of several distinct drying methods on the flavonoid levels. Moreover, the flavonoid content was correlated with the antioxidative activity of mulberry leaves. Their data revealed that the flavonoids’ levels and the antioxidative effect are impacted by both variables. When it comes to the mulberry growth time, the highest flavonoid accumulation was registered in two situations, namely in the leaves that grew for a long period of time and in the leaves that were not exposed long term to environmental stress. As for the evaluated drying methods, namely the sun, air, oven, and freeze drying, their results showed that, by using the first two methods, the highest flavonoid content was registered. On the other hand, by using air-drying and freeze-drying methods, the antioxidative effects of the flavonoids were better retained.
Jaruchotikamol and Pannangpetch (2013) [116] analyzed the cytoprotective action of M. alba leaf extract. Firstly, in vitro, the mulberry leaves extract exhibited a scavenging action on a certain free radical, namely 1, 1-diphenyl-2-picrylhydrazyl (DPPH). The authors revealed that the antioxidative effect of mulberry leaves depends on the concentration. However, the authors used vitamin C as a positive control and showed that, even if mulberry extract possesses certain antioxidative effects, vitamin C displays more powerful antioxidative activity. Additionally, by using 2, 2′-azobis (2-amidinopropane) dihydrochloride (AAPH), they induced hemolysis in order to investigate the protective action of the mulberry extract on erythrocytes. Positively, the target extract registered protective action against hemolysis induced by reactive oxygen species. After confirming the therapeutic effect of the mulberry extract in vitro, the authors further investigated it in vivo, in rats, by inducing a gastric mucosal lesion through ischemia–reperfusion (I/R). For three days, the rats received oral doses of mulberry extract of different quantities: 0.25, 0.50, 1, and 2 g/kg/day, respectively. Interestingly, by administering 0.25 and 0.50 g/kg/day doses, the target product exposes great protective action against gastric mucosal injury. To the contrary, when higher doses were administered, the protective action of the investigated extract was absent.
In Table 2 are listed various recent studies that certified mulberry’s health-promoting potential.

3. Fibroin’s Impact in Anticancer Therapy

3.1. Fibroin-Based Target Delivery Systems

Nowadays, tremendous progress has been made in overcoming the limitations of conventional cancer therapies. The most used therapeutic modalities in cancer are chemotherapy and radiotherapy. It is well documented that these treatments include a broad range of negative side effects, such as systemic toxicity and psychological problems, and also, the tumor cells develop high drug resistance [125]. One great strategy to overcome both the drug resistance and negative side effects is the use of certain drug delivery systems [126].
In order to develop a target delivery system, Mottaghitalab et al. (2017) [127] used loaded SF nanoparticles to target lung tumors. The prepared SF nanoparticles were loaded with gemcitabine and SP5-52 peptide. The last compound was conjugated due to its ability to conduct the anticancer drug carriers, and by adding it, a target delivery system was obtained. The prepared SF nanoparticles were acquired by using the prepared SF solution to dissolve 10 mL of DMSO. After it was centrifuged, the precipitate nanoparticle aggregates were evaluated to determine its size, morphology, and surface charge by using both dynamic light scattering and scanning electron microscopy. The peptide Sp5-52 was covalently attached to the loaded SF nanoparticles by using two elements, namely 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide and N-hydroxysuccinimide. Following the addition of 25 mg of the obtained nanoparticles and 2.5 mg of gemcitabine into 10 mL DMSO, the solution was sonicated and dialyzed against water. UV spectroscopy was used to determine the quantity of the loaded drug. The same technique was used to examine the gemcitabine release profile. Additionally, the toxicity of the loaded nanoparticles was tested both in vitro and in vivo by using lung carcinoma cells and a normal lung epithelial cell line and mice, respectively. Regarding the in vivo experiments, the authors induced lung tumors in mice. Their data revealed that, by adding SP5-52 peptide to the SF gemcitabine-loaded nanoparticles, superior cytotoxicity and cellular uptake was observed, compared with the single drug-loaded nanoparticles and control group. Furthermore, by using this delivery strategy, in the lung tissue, a greater accumulation of gemcitabine was recorded. The in vivo evaluation showed that the targeted delivery system led to higher mice survival rates and a lower mortality percent. No metastasis was observed in mice after the target treatment was applied. This study underlines the efficacy of using SF as a drug delivery system.
Zare-Zardini et al. (2021) [128] developed two SF-based biomaterials as drug delivery approaches, specifically micro- and nanoparticles. The fibroin carriers were loaded with doxorubicin, and its biological activity was assessed in this formulation. The cytotoxicity of the coated therapeutic agent was confirmed by using three different cell lines, specifically breast cancer, normal, and bone cancer cell lines. Two techniques, staining with PI/annexin V and flow cytometry, respectively, were used in order to determine the induced apoptosis, and the results revealed that, by using both developed doxorubicin delivery systems, notable levels of apoptosis were observed. Additionally, the authors evaluated the p53 gene expression by using real-time PCR, and their data showed that the doxorubicin-loaded fibroin nanoparticles led to a remarkable increase in the p53 expression level in both tumoral cell lines; on the other hand, a notable decrease was observed in the healthy cell line. Employing the doxorubicin microparticles in normal and breast cancer cell lines, an increased level of expression of p53 was observed; however, in the osteogenic sarcoma cell line, the expression level decreased. These strategies exhibit great potential as new antitumoral treatments.
The impact of using SF as a drug delivery platform was reported by Wu et al. (2018) [129]. The authors targeted the cancer stem cells that exhibit chemotherapy resistance. It has been certified that salinomycin, a veterinary antibiotic, is effective against cancer stem cell proliferation. Keeping in mind that salinomycin poisoning is lethal, the authors used SF nanoparticles to achieve drug-controlled delivery in order to administer salinomycin in a safer and more efficient manner. Several studies demonstrated that this specific antibiotic sensitized paclitaxel, a well-known chemotherapeutic agent [130,131]. Furthermore, they also prepared paclitaxel-loaded nanoparticles to develop a codelivery system for anticancer treatment. After preparing the SF salinomycin and paclitaxel-loaded nanoparticles, the authors fabricated a SF-based hydrogen and incorporated the nanoparticles into it. Prior to SF gelation, the loaded nanoparticles were dispersed into it. The hydrogel’s biodegradability and its injectable properties were maintained. Additionally, when analyzing the drug distribution, the authors observed that, by incorporating drug-loaded nanoparticles into the hydrogel, the distribution was more homogeneous compared to the hydrogel, which did not have incorporated nanoparticles but uncoated drugs. In terms of tumor growth inhibition, by comparing the codelivery strategy with the single drug treatment in the murine hepatic carcinoma tumor model, the first mentioned therapy resulted in superior tumor growth inhibition. In addition, the developed fibroin-based strategy increased the salinomycin tolerance.
In a novel study, Suyamud et al. (2021) [132] used SF as a coating biomaterial for doxorubicin-loaded liposomes. SF was chosen for liposomal coating to prolong doxorubicin’s circulation time but also to improve its long-term efficacy. The main objectives of this study were to evaluate the encapsulation efficiency, the anticancer effect, the cytotoxicity in healthy cells, and, also, the doxorubicin release profile. The authors examined the anticancer activity by using two different breast cancer cell lines. Their data showed that, by using fibroin as the coating biomaterial, the anticancer effect was diminished; however, the author achieved doxorubicin long-term doxorubicin release. Moreover, by using SF, the cytotoxicity of doxorubicin on healthy cells was reduced.
In another study [133], SF was used as a nanocarrier for curcumin, a well-known phenolic pigment that exhibits extraordinary biological activities, including an antitumoral effect. Even if it displays great antitumoral activity, there are several major impediments regarding its bioavailability, including rapid intestinal metabolism and low water solubility. Accordingly, it is of great importance to use nanocarriers to foster the delivery of curcumin. The loaded SF-based nanoparticles were developed by using two techniques, specifically coprecipitation and physical adsorption. The cytotoxic effect of formulated nanoparticles was evaluated on normal, neuroblastoma, and hepatocellular carcinoma cell lines. The in vitro toxicity experiments confirmed the antitumoral effect of curcumin-loaded SF nanoparticles on cancer cell lines but not on the normal cell lines. This study confirmed the chemotherapeutic and chemopreventive potential of curcumin but also the feasibility of using SF as a nanocarrier. In Table 3 are recorded the novel studies that used various fibroin-based carriers for a cancer therapeutic approach.

3.2. Fibroin-Based Tumor Models

One of the most important strategies that leads to a better understanding and plays a great role in fighting cancer is the development of three-dimensional (3D) tumor models. Its production overcomes the limitations of using animal models that cannot mimic numerous human aspects, including the immune system, stromal interactions, and metabolism [153]. By using these tools, numerous cancer aspects can be studied, such as cancer cell differentiation, proliferation, angiogenesis, drug resistance, immunosurveillance, testing new therapeutics, and so on [154]. There are numerous biomaterials that are currently being used for developing these bioengineered platforms, including alginate, chitosan, SF, agarose, and fibrinogen [155]. Due to its biocompatibility and biodegradability, fibroin is one of the most used biomaterials for the development of 3D tumor models [156].
Buhome et al. (2022) [157] developed SF gelatin/hyaluronic acid/heparan sulfate scaffolds (SF-GHH) that served as 3D models in order to facilitate and accelerate the investigation of the role played by cancer stem cells in cholangiocarcinoma. In this study, the authors used the developed scaffold to examine the cancer cell morphology and proliferation. The tumor microenvironment (SF-GHH) was fabricated in various blending ratios (2:1, 1:1, and 1:2) by lyophilization, and their data showed that, by using a ratio of 1:2, the scaffold could simulate the extracellular matrix. In terms of its physical features, including the optimal porosity, stable beta-sheet, and water uptake characteristics, the best results were observed by using a scaffold with a 350 ± 102 μm pore size. The SF-GHH 3D model supported cell proliferation, cell aggregation, and attachment, which led to spheroid formation. Moreover, the authors investigated the expression levels in the 3D culture of two stemness-related genes, specifically Sox2 and Nanog, but also of four epithelial–mesenchymal transition-related genes: MMP-9, Snail-1, Twist1, and Zeb1. The results showed that, by using a SF-GHH scaffold as a 3D model, the targeted genes were upregulated.
In a recent study, SF-based hydrogels were used as artificial biomimetic 3D matrices. The authors developed rapidly responsive hydrogels, keeping in mind the great impact of proteins’ conformational transition on cell behavior and tumor suppression. By using the horseradish peroxidase and hydrogen peroxide crosslinking method, the researchers obtained rapidly responsive SF-based hydrogels that, under physiological conditions, exhibited conformational changes; specifically, random coil SF were converted in β-sheet SF hydrogels. A very interesting characteristic of the SF-based developed hydrogel was the conformation influence on hydrogels’ opaque morphology; more specifically, a transparent appearance was observed during the random coil conformation and was opaque during the β-sheet form. For the in vitro evaluation, the hydrogels were used for human neuronal glioblastoma cell encapsulation, showing that the transparent hydrogels stimulate cell viability while the opaque SF hydrogels induced cell apoptosis. These findings exhibit major progress on investigating programed cancer cells death [158].
Dondajewska et al. (2017) [159] used SF to develop porous scaffolds to investigate breast tumor biology but also to analyze the antitumoral activity of doxorubicin. In the developed SF scaffolds were seeded two different cell lines, EMT6 breast cancer and NIH3T3 fibroblast, respectively, in order to study cell–cell interactions but also cell–extracellular matrix relations. Two specific methods, namely lyophilization and salt leaching, were used to develop four distinct types of scaffolds. The developed biomaterials exhibited differences in terms of pore size and shape and also scaffold thickness. By using the salt leaching method, the authors observed spherical pores that were uniformly distributed; on the other hand, by using the lyophilization technique, longitudinal pores were detected. The cell attachment was not impacted by the used technique; however, the cancer line attached slower to the silk scaffold compared to fibroblasts. These results confirm the fact that SF supports cell cultures. Furthermore, their data showed that the cell lines that were grown in 3D cultures were more resistant to the antitumoral activity of doxorubicin compared to its effects in 2D cultures. This underlines the potential of using a SF tumor model to analyze the anticancer drug agent’s ability to penetrate the tumor.
In another novel study (2022) [160], SF and chitosan were associated with developing 3D scaffolds for chemotherapeutic drug screening sensitivity. Two cross-linking elements were used to prepare the target scaffolds—specifically, 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide and sodium tripolyphosphate—and various techniques such as scanning electron microscopy, swelling, or the water absorption ration were used to characterize its properties. A colon cancer cell line (LoVo) was used to investigate scaffold’s impact on cell adhesion and growth. In addition, a breast cancer cell line, MDA-MB-231, was employed for drug sensitivity testing. The authors reported that, by using 3D scaffolds, a superior sensitivity of the drug screening process was observed compared to the results registered when using 2D models.
Another research group used SF and chitosan to develop a valuable platform for mimicking the tumor microenvironment in order to evaluate the activity of certain anticancer drugs. Human tumoral lung cell line (A549) was seeded on a SF/chitosan porous scaffold and displayed good distribution after 24 h, and after 48 h, cancer cells displayed a spherical shape and were also interconnected. Moreover, after 72 h, a high rates of tumor spheres were observed, and their sizes increased. Compared to 2D models, by using 3D SF/chitosan scaffolds, the tumoral cell line had a superior adhesion, distribution, and proliferation rate. Doxorubicin was used to evaluate the drug resistance of target tumoral cells on the porous scaffold. Their results showed that, by using a 3D model, a higher concentration of chemotherapeutic agent was needed for 50% cell death compared to the 2D systems. This underlines the drug incapacity of uniformly diffusing and the necessity of studying the tumor microenvironment in the process of drug design. Their data showed that, by using SF/chitosan scaffolds, various valuable pieces of information can be brought to light regarding the anticancer activity of specific chemotherapeutic agents and tumor drug resistance [161].

4. Sericin in Cancer Research

Although SF is the main protein used in cancer research, various studies have disclosed sericin’s therapeutic effects, including its anti-inflammatory [162,163] and antioxidative [164] activities. Moreover, it has been reported that sericin exhibits a great impact in anticancer research due to its cytotoxic activity, and also, it has been used for nanocarrier formulations in anticancer therapy [165]. In Table 4 are listed the studies that used sericin as a nanocarrier system to deliver target antitumoral agents.
Niu et al. (2021) [178] investigated the antitumoral activity of distinct concentrations of sericin on two triple-negative human breast cancer cell lines, this being one of the most aggressive type of cancers. After applying the sericin treatment, the cell viability was analyzed by using the MTT assay, and the results showed that sericin exhibits an antitumoral effect in a dose-dependent way. Additionally, the colony formation rate and proliferation were suppressed by sericin. Furthermore, flow cytometry was used to investigate the apoptosis and cell cycle progress, and the results revealed that the natural compound led to cell cycle arrest, specifically in the G0/G1 phase. These results were confirmed by assessing the expression level of several target proteins, including cyclin D1, Cdk4, E2F3, P21, and P27. Additionally, the apoptotic rate was evaluated, and it validated the tumoral cell apoptosis caused by sericin. In addition, the induced tumor cell apoptosis was investigated by analyzing the level of expression of several apoptosis-related proteins, specifically Bax, Cyto-C, and Bacl-2. The first two proteins were upregulated, while the last one was downregulated by applying sericin as the treatment, confirming the induced cellular apoptosis. In another study, Kumar et al. (2019) evaluated sericin’s antitumoral activity on three different tumoral cell lines, specifically squamous carcinoma (A431), tongue carcinoma (SAS), and breast adenocarcinoma (MCF-7). By applying 1 mg/mL of silk sericin treatment, the viability of the cancer cells was not affected, but by increasing the concentration by 4 mg/mL, the cell viability was negatively impacted in all tumoral cell lines. Comparing the normal cells’ and tumoral cells’ viability after treating them with silk sericin, the authors revealed that sericin exhibits a greater cytotoxicity to tumoral cell lines than healthy cells [179].

5. Mulberry as an Alternative Anticancer Approach

Numerous studies have certified the health-promoting benefits of the mulberry plant—more specifically, the medicinal value of mulberry leaves, root, bark, or fruits [63]. One of the greatest bioactivities of the mulberry is the antitumoral effect.
Shu et al. (2020) [180] isolated and purified guangsangon E, a phenolic compound that is found in M. alba, in order to evaluate its antitumoral activity against respiratory cancer. Two cancer cell lines were used, specifically a lung cancer (A549) and a nasopharyngeal carcinoma cell line (CNE1), and were treated with distinct concentrations of guangsangon E for 24, 48, and 72 h. In both tumoral cell lines, guangsangon E displayed antitumoral activity and significantly negatively impacted the colony formation. Analyzing the type of cell death induced by the phenolic compound, the authors revealed that, in the treated cells, the ratio of apoptotic cells was 35.4%. Moreover, these results were confirmed after the examination of the expression level of two key players for tumor progression, namely Bax and Bcl-2. Its expression levels were significantly decreased in the cells that received guangsangon E treatment. Furthermore, the impact of the phenolic compound on a specific autophagosome marker (LC3) was assessed. Their data showed that guangsangon E and the ration levels of LC3 were remarkably increased, revealing the influence of this element on enhancing the autophagy flux in tumoral cell lines. The autophagy and apoptosis connection was investigated by analyzing through immunoprecipitation the interaction of the Bcl-2 protein and an antiapoptotic protein named Beclin 1. The interaction of the two elements was diminished by applying the guangsangon E treatment; therefore, the autophagy process was stimulated by dissociating the antiapoptotic protein from Bcl-2. Moreover, the authors exhaustively analyzed the role of autophagy in the antitumoral effect of guangsangon E by using an autophagy inhibitor, chloroquine. Its association with guangsangon E enhanced the tumoral cells’ viability, suggesting the fact that the cell death was caused by autophagy. The next step was the knocking down of an autophagy-related gene, Atg5, and evaluating the viability of genetically modified cells after the treatment was applied. The knockdown of the target gene led to a lower apoptosis rate in the presence of guangsangon E. These results indicate that, through autophagy, this phenolic compound exhibits great antitumoral activity.
One of the most important secondary metabolites produced by mulberry leaves is chalocomoracin. It represents a defense system against fungal germination. It is well documented that it exhibits several bioactivities, including antibacterial and antiviral effects. Han et al. (2018) [181] explored the anticancer activity of chalocomoracin both in vitro and in vivo; in addition, its molecular mechanism was studied. The cytotoxic effect was studied on two human prostate cancer cell lines, LNCaP and PC-3, and one breast cancer cell line, MDA-MB-231. Their data showed that, by treating the cell lines with the target compound, the cell viability was significantly decreased. Interestingly, in two cancer cell lines, PC-3 and MDA-MB-231, the cell death was not triggered by apoptosis but by extensive cytoplasmic vacuolation, this event being distinctive for the paraptosis pathway. Additionally, their findings indicate that chalocomoracin enhanced the endoplasmic reticulum stress but also promoted mitophagy. In addition, the expression level of a key regulator, PINK1, was explored. When mitochondrial damage occurs, high ratios of PINK1 are registered; further, its role in paraptosis was highlighted when the chalocomoracin was applied on the cell lines and PINK1 was upregulated. Additionally, by using nude mice, the impact of chalocomoracin on breast cancer was evaluated in vivo. By administering different doses of the target compound, a significant tumor inhibition was observed up to 54%. The molecular analyses revealed that the treatment also upregulated PINK1 in vivo.
In 2021, Dalkiliç et al. [182] explored in a novel study the cytotoxic potential of M. nigra fruit extract by analyzing its effects on two human cancer lines, specifically MDA-MB-231 (breast cancer) and PC3 (prostate cancer). The authors applied the mulberry extract on the cell lines for three days by using five different concentrations: 1%, 1.33%, 2%, 4%, and 10%, respectively. For measuring the cell viability, the MTT assay was used, and the results revealed that the mulberry exhibits a cytotoxic effect in a concentration-dependent manner. When it comes to the prostate cancer cell line, the cell viability was affected only by applying the mulberry extract of 10% concentration. On the other hand, when applying mulberry extract of lower concentrations, no cytotoxic effect was registered. As for the breast cancer cell line, similar results were registered—the highest concentration led to the most powerful cytotoxic effect; however, lower anticancer activity was reported at 1%, 1.33%, 2%, and 4%, respectively. Moreover, when comparing the anticancer effect of the same concentrations of doxorubicin and mulberry extract, the cytotoxic effect of the fruit extract was higher than the outcome of using the chemotherapy drug.
In another study, the flavonoids were extracted from mulberry leaves (M. alba) to explore its potential as an anticancer treatment. Moreover, in this study, the authors explored the interaction between the flavonoids and DNA. Calf thymus DNA was incubated with various concentrations of the target compounds and analyzed after 45 min by using a spectroscopic analysis. The results showed that the extracted flavonoids inhibited tumor growth through DNA modifications by binding to the sugar–phosphate backbone. However, the in vivo experiments did not confirm the flavonoids’ effect in inhibiting tumor growth but increased the lifespan of cancerous mice [183]. In Table 5 are listed the novel studies that confirmed the antitumoral effects of different compounds that are found in the mulberry.

6. Conclusions

Cancer remains a major public health burden, and its incidence is continually growing, despite the massive effort that is being made by the scientific community. Even if, in the pharmaceutical industry, a wide range of therapeutic agents is available, they face numerous impediments. In this regard, researchers worldwide are constantly searching for natural compounds that could be a source for novel leads in anticancer research. Sericulture’s applicability is no longer limited to the textile industry; it displays a tremendous impact on various science-related fields, including cancer research. The main reason that brought B. mori into the spotlight is the great applicability of SF in the biomaterials area. Numerous studies have used this polymer to overcome certain drawbacks that most of the cancer therapeutic agents exhibit—for instance, drug’s high cytotoxicity on normal cells or its low availability. In this direction, a wide range of researchers have used SF as a drug carrier and demonstrated its feasibility to successfully deliver target agents that exhibited cytotoxicity against various cancer cell lines and achieved long-term release. SF’s applicability in cancer research and therapy is not only due to its role as an anticancer drug delivery strategy, but it is a commonly used biomaterial for the development of 3D tumor models. The 3D models exhibit superior biomimetic properties compared to the 2D models, leading to a better understanding of cancer molecular mechanisms, and play a crucial role in the development of novel anticancer strategies.
Considering the great potential of plants as a rich reservoir of various bioactive elements, tremendous efforts are being made to develop anticancer plant-based therapeutics. Moreover, a wide range of anticancer agents that are frequently used in clinical practice originate from plants, such as irinotecan and paclitaxel. It is well documented that the mulberry composition includes various bioactive elements that make mulberry a great functional food. In this direction, due to its complex composition, numerous studies have certified the anticancer effects of the mulberry and underlined its potential as an anticancer alternative therapy.

Author Contributions

Conceptualization, G.-M.B. and D.S.D.; resources, G.-M.B., E.-D.B., A.-A.C. and D.S.D.; data curation, G.-M.B., A.I.G., A.-S.M., M.Z. and A.R.M.; writing—original draft preparation, G.-M.B., E.-D.B. and A.-A.C.; writing—review and editing, G.-M.B., M.Z. and D.S.D.; visualization, A.I.G., A.-A.C., A.-S.M., A.R.M. and D.S.D.; and supervision, D.S.D. and M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Zhou, B.; Zang, R.; Zhang, M.; Song, P.; Liu, L.; Bie, F.; Peng, Y.; Bai, G.; Gao, S. Worldwide Burden and Epidemiological Trends of Tracheal, Bronchus, and Lung Cancer: A Population-Based Study. EBioMedicine 2022, 78, 103951. [Google Scholar] [CrossRef] [PubMed]
  3. Didkowska, J.; Wojciechowska, U.; Michalek, I.M.; Caetano dos Santos, F.L. Cancer Incidence and Mortality in Poland in 2019. Sci. Rep. 2022, 12, 10875. [Google Scholar] [CrossRef]
  4. Zugazagoitia, J.; Guedes, C.; Ponce, S.; Ferrer, I.; Molina-Pinelo, S.; Paz-Ares, L. Current Challenges in Cancer Treatment. Clin. Ther. 2016, 38, 1551–1566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gammon, J.M.; Dold, N.M.; Jewell, C.M. Improving the Clinical Impact of Biomaterials in Cancer Immunotherapy. Oncotarget 2016, 7, 15421. [Google Scholar] [CrossRef] [PubMed]
  6. Gu, L.; Mooney, D.J. Biomaterials and Emerging Anticancer Therapeutics: Engineering the Microenvironment. Nat. Rev. Cancer 2016, 16, 56–66. [Google Scholar] [CrossRef] [Green Version]
  7. Devarasetty, M.; Forsythe, S.D.; Shelkey, E.; Soker, S. In Vitro Modeling of the Tumor Microenvironment in Tumor Organoids. Tissue Eng. Regen. Med. 2020, 17, 759–771. [Google Scholar] [CrossRef]
  8. Desai, A.G.; Qazi, G.N.; Ganju, R.K.; El-Tamer, M.; Singh, J.; Saxena, A.K.; Bedi, Y.S.; Taneja, S.C.; Bhat, H.K. Medicinal Plants and Cancer Chemoprevention. Curr. Drug Metab. 2008, 9, 581–591. [Google Scholar] [CrossRef] [Green Version]
  9. Siddiqui, A.J.; Jahan, S.; Singh, R.; Saxena, J.; Ashraf, S.A.; Khan, A.; Choudhary, R.K.; Balakrishnan, S.; Badraoui, R.; Bardakci, F.; et al. Plants in Anticancer Drug Discovery: From Molecular Mechanism to Chemoprevention. BioMed Res. Int. 2022, 2022, 5425485. [Google Scholar] [CrossRef]
  10. Iqbal, J.; Abbasi, B.A.; Mahmood, T.; Kanwal, S.; Ali, B.; Shah, S.A.; Khalil, A.T. Plant-Derived Anticancer Agents: A Green Anticancer Approach. Asian Pac. J. Trop. Biomed. 2017, 7, 1129–1150. [Google Scholar] [CrossRef]
  11. Fridlender, M.; Kapulnik, Y.; Koltai, H. Plant Derived Substances with Anti-Cancer Activity: From Folklore to Practice. Front. Plant Sci. 2015, 6, 799. [Google Scholar] [CrossRef] [PubMed]
  12. Eo, H.J.; Park, J.H.; Park, G.H.; Lee, M.H.; Lee, J.R.; Koo, J.S.; Jeong, J.B. Anti-Inflammatory and Anti-Cancer Activity of Mulberry (Morus alba L.) Root Bark. BMC Complement. Altern. Med. 2014, 14, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Chan, E.W.C.; Wong, S.K.; Tangah, J.; Inoue, T.; Chan, H.T. Phenolic Constituents and Anticancer Properties of Morus alba (White Mulberry) Leaves. J. Integr. Med. 2020, 18, 189–195. [Google Scholar] [CrossRef] [PubMed]
  14. Li, W.; Zhang, Z.; Lin, L.; Terenius, O. Antheraea Pernyi (Lepidoptera: Saturniidae) and Its Importance in Sericulture, Food Consumption, and Traditional Chinese Medicine. J. Econom. Entomol. 2017, 110, 1404–1411. [Google Scholar] [CrossRef] [PubMed]
  15. Chauhan, T.P.S.; Tayal, M.K. Mulberry Sericulture. In Industrial Entomology; Springer: Singapore, 2017; pp. 197–263. [Google Scholar] [CrossRef]
  16. Chanotra, S.; Kamlesh Bali, I.; Rk Bali, I.; Corresponding, I.; Kashmir, I.; Bali, K.; Bali, R.K. Sericulture: An Opportunity for the Upliftment of Rural Livelihood. J. Entomol. Zool. Stud. 2019, 7, 1100–1103. [Google Scholar]
  17. Chandan, R. A Review on Genesis, Growth and Development of Bengal Artisanal Silk Industry in India. Integr. J. Res. Arts Humanit. 2022, 2, 27–37. [Google Scholar] [CrossRef]
  18. Bucciarelli, A.; Motta, A. Use of Bombyx mori silk fibroin in tissue engineering: From cocoons to medical devices, challenges, and future perspectives. Biomater. Adv. 2022, 139, 212982. [Google Scholar] [CrossRef]
  19. Wang, Q.; Yang, X. Analysis on the Development of China’s Modern Silk Industry. Asian Soc. Sci. 2022, 18, 27. [Google Scholar] [CrossRef]
  20. Ratiu, A.C.; Neagu, A.; Mihalache, R.M.; Lazar, V.; Ecovoiu, A. Molecular and Bioinformatics Analysis of the Relative Expression Profiles of Dorsal, Toll-1, Relish and Duox Genes in Young versus Old Diutinus Workers of Apis Mellifera. Rom. Biotechnol. Lett. 2016, 21, 11513–11526. [Google Scholar]
  21. Tabunoki, H.; Bono, H.; Ito, K.; Yokoyama, T. Can the Silkworm (Bombyx mori) Be Used as a Human Disease Model? Drug Discov. Ther. 2016, 10, 3–8. [Google Scholar] [CrossRef] [Green Version]
  22. Kawamoto, M.; Jouraku, A.; Toyoda, A.; Yokoi, K.; Minakuchi, Y.; Katsuma, S.; Fujiyama, A.; Kiuchi, T.; Yamamoto, K.; Shimada, T. High-Quality Genome Assembly of the Silkworm, Bombyx mori. Insect Biochem. Mol. Biol. 2019, 107, 53–62. [Google Scholar] [CrossRef] [PubMed]
  23. Baci, G.M.; Cucu, A.A.; Giurgiu, A.I.; Muscă, A.S.; Bagameri, L.; Moise, A.R.; Bobiș, O.; Rațiu, A.C.; Dezmirean, D.S. Advances in Editing Silkworms (Bombyx mori) Genome by Using the Crispr-Cas System. Insects 2022, 13, 28. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, S.; Shah, S.A.M.; Basharat, K.; Qamar, S.A.; Raza, A.; Abdullah, M.; Muhammad, B.; Hafiz, I. Silk-based nano-hydrogels for futuristic biomedical applications. J. Drug Deliv. Sci. Technol. 2022, 72, 103385. [Google Scholar] [CrossRef]
  25. Li, G.; Sun, S. Silk Fibroin-Based Biomaterials for Tissue Engineering Applications. Molecules 2022, 27, 2757. [Google Scholar] [CrossRef]
  26. Sun, W.; Gregory, D.A.; Tomeh, M.A.; Zhao, X. Molecular Sciences Silk Fibroin as a Functional Biomaterial for Tissue Engineering. Int. J. Mol. Sci. 2021, 22, 1499. [Google Scholar] [CrossRef]
  27. Kwon, G.; Heo, B.; Kwon, M.J.; Kim, I.; Chu, J.; Kim, B.Y.; Kim, B.K.; Park, S.S. Effect of Silk Fibroin Biomaterial Coating on Cell Viability and Intestinal Adhesion of Probiotic Bacteria. J. Microbiol. Biotechnol. 2021, 31, 592–600. [Google Scholar] [CrossRef]
  28. Nguyen, T.P.; Nguyen, Q.V.; Nguyen, V.H.; Le, T.H.; Huynh, V.Q.N.; Vo, D.V.N.; Trinh, Q.T.; Kim, S.Y.; van Le, Q. Silk Fibroin F-Based Biomaterials for Biomedical Applications: A Review. Polymers 2019, 11, 1933. [Google Scholar] [CrossRef] [Green Version]
  29. 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]
  30. Yu, R.; Yang, Y.; He, J.; Li, M.; Guo, B. Novel Supramolecular Self-Healing Silk Fibroin-Based Hydrogel via Host–Guest Interaction as Wound Dressing to Enhance Wound Healing. Chem. Eng. J. 2021, 417, 128278. [Google Scholar] [CrossRef]
  31. Zheng, H.; Zuo, B. Functional Silk Fibroin Hydrogels: Preparation, Properties and Applications. J. Mater. Chem. B 2021, 9, 1238–1258. [Google Scholar] [CrossRef]
  32. Pudkon, W.; Laomeephol, C.; Damrongsakkul, S.; Kanokpanont, S.; Ratanavaraporn, J. Comparative Study of Silk Fibroin-based Hydrogels and Their Potential as Material for 3-dimensional (3d) Printing. Molecules 2021, 26, 3887. [Google Scholar] [CrossRef] [PubMed]
  33. Maity, B.; Samanta, S.; Sarkar, S.; Alam, S.; Govindaraju, T. Injectable Silk Fibroin-Based Hydrogel for Sustained Insulin Delivery in Diabetic Rats. ACS Appl. Bio Mater. 2020, 3, 3544–3552. [Google Scholar] [CrossRef] [PubMed]
  34. Varkey, A.; Venugopal, E.; Sugumaran, P.; Janarthanan, G.; Pillai, M.M.; Rajendran, S.; Bhattacharyya, A. Impact of Silk Fibroin-Based Scaffold Structures on Human Osteoblast MG63 Cell Attachment and Proliferation. Int. J. Nanomed. 2015, 10, 43–51. [Google Scholar] [CrossRef] [Green Version]
  35. Nisal, A.; Sayyad, R.; Dhavale, P.; Khude, B.; Deshpande, R.; Mapare, V.; Shukla, S.; Venugopalan, P. Silk Fibroin Micro-Particle Scaffolds with Superior Compression Modulus and Slow Bioresorption for Effective Bone Regeneration. Sci. Rep. 2018, 8, 7235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Parekh, N.; Hushye, C.; Warunkar, S.; sen Gupta, S.; Nisal, A. In Vitro Study of Novel Microparticle Based Silk Fibroin Scaffold with Osteoblast-like Cells for Load-Bearing Osteo-Regenerative Applications. RSC Adv. 2017, 7, 26551–26558. [Google Scholar] [CrossRef] [Green Version]
  37. Li, Z.H.; Ji, S.C.; Wang, Y.Z.; Shen, X.C.; Liang, H. Silk Fibroin-Based Scaffolds for Tissue Engineering. Front. Mater. Sci. 2013, 7, 237–247. [Google Scholar] [CrossRef]
  38. Zhu, H.M.; Liu, W.; Yin, Q.; Li, S.W.; Wang, C.Y.; Song, L.; Qiao, X. Silk Fibroin Sponge Combined with Cell-Derived ECM for Tissue-Engineered 3D Functional Neural Tissues. Sci. China Technol. Sci. 2020, 63, 2113–2122. [Google Scholar] [CrossRef]
  39. Najberg, M.; Haji Mansor, M.; Taillé, T.; Bouré, C.; Molina-Peña, R.; Boury, F.; Cenis, J.L.; Garcion, E.; Alvarez-Lorenzo, C. Aerogel Sponges of Silk Fibroin, Hyaluronic Acid and Heparin for Soft Tissue Engineering: Composition-Properties Relationship. Carbohydr. Polym. 2020, 237, 116107. [Google Scholar] [CrossRef]
  40. Siavashani, A.Z.; Mohammadi, J.; Rottmar, M.; Senturk, B.; Nourmohammadi, J.; Sadeghi, B.; Huber, L.; Maniura-Weber, K. Silk Fibroin/Sericin 3D Sponges: The Effect of Sericin on Structural and Biological Properties of Fibroin. Int. J. Biol. Macromol. 2020, 153, 317–326. [Google Scholar] [CrossRef]
  41. Kambe, Y.; Kojima, K.; Tamada, Y.; Tomita, N.; Kameda, T. Silk Fibroin Sponges with Cell Growth-Promoting Activity Induced by Genetically Fused Basic Fibroblast Growth Factor. J. Biomed. Mater. Res. A 2016, 104, 82–93. [Google Scholar] [CrossRef]
  42. Zhaorigetu, S.; Sasaki, M.; Watanabe, H.; Kato, N. Supplemental Silk Protein, Sericin, Suppresses Colon Tumorigenesis in 1,2-Dimethylhydrazine-Treated Mice by Reducing Oxidative Stress and Cell Proliferation. Biosci. Biotechnol. Biochem. 2001, 65, 2181–2186. [Google Scholar] [CrossRef] [PubMed]
  43. Dong, H.L.; Zhang, S.X.; Tao, H.; Chen, Z.H.; Li, X.; Qiu, J.F.; Cui, W.Z.; Sima, Y.H.; Cui, W.Z.; Xu, S.Q. Metabolomics Differences between Silkworms (Bombyx mori) Reared on Fresh Mulberry (Morus) Leaves or Artificial Diets. Sci. Rep. 2017, 7, 10972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jaiswal, K.K.; Banerjee, I.; Mayookha, V.P. Recent Trends in the Development and Diversification of Sericulture Natural Products for Innovative and Sustainable Applications. Bioresour. Technol. Rep. 2021, 13, 100614. [Google Scholar] [CrossRef]
  45. Kim, H.-B.; Ryu, S.; Baek, J.-S. The Effect of Hot-Melt Extrusion of Mulberry Leaf on the Number of Active Compounds and Antioxidant Activity. Plants 2022, 11, 3019. [Google Scholar] [CrossRef]
  46. de Pádua Lúcio, K.; Rabelo, A.C.S.; Araújo, C.M.; Brandão, G.C.; de Souza, G.H.B.; da Silva, R.G.; de Souza, D.M.S.; Talvani, A.; Bezerra, F.S.; Cruz Calsavara, A.J.; et al. Anti-Inflammatory and Antioxidant Properties of Black Mulberry (Morus nigra L.) in a Model of LPS-Induced Sepsis. Oxid. Med. Cell. Longev. 2018, 2018, 5048031. [Google Scholar] [CrossRef] [Green Version]
  47. Manzoor, M.F.; Hussain, A.; Tazeddinova, D.; Abylgazinova, A.; Xu, B. Assessing the Nutritional-Value-Based Therapeutic Potentials and Non-Destructive Approaches for Mulberry Fruit Assessment: An Overview. Comput. Intell. Neurosci. 2022, 2022. [Google Scholar] [CrossRef]
  48. Thaipitakwong, T.; Numhom, S.; Aramwit, P. Mulberry Leaves and Their Potential Effects against Cardiometabolic Risks: A Review of Chemical Compositions, Biological Properties and Clinical Efficacy. Pharm. Biol. 2018, 56, 109–118. [Google Scholar] [CrossRef]
  49. Arfan, M.; Khan, R.; Rybarczyk, A.; Amarowicz, R. Antioxidant Activity of Mulberry Fruit Extracts. Int. J. Mol. Sci. 2012, 13, 2472–2480. [Google Scholar] [CrossRef]
  50. Chan, E.W.C.; Lye, P.Y.; Wong, S.K. Phytochemistry, Pharmacology, and Clinical Trials of Morus alba. Chin. J. Nat. Med. 2016, 14, 17–30. [Google Scholar] [CrossRef]
  51. Liu, Y.; Zhao, Y.B.; Wang, S.W.; Zhou, Y.; Tang, Z.S.; Li, F. Mulberry Granules Protect against Diabetic Cardiomyopathy through the AMPK/Nrf2 Pathway. Int. J. Mol. Med. 2017, 40, 913–921. [Google Scholar] [CrossRef] [Green Version]
  52. Yu, J.S.; Lim, S.H.; Lee, S.R.; Choi, C.I.; Kim, K.H. Antioxidant and Anti-Inflammatory Effects of White Mulberry (Morus alba L.) Fruits on Lipopolysaccharide-Stimulated RAW 264.7 Macrophages. Molecules 2021, 26, 920. [Google Scholar] [CrossRef]
  53. Wu, T.-Y.; Liang, J.; Ai, J.-Y.; Cui, J.-L.; Huang, W.-D.; You, Y.-L.; Zhan, J.-C. Mulberry Ethanol Extract and Rutin Protect Alcohol-Damaged GES-1 Cells by Inhibiting the MAPK Pathway. Molecules 2022, 27, 4266. [Google Scholar] [CrossRef] [PubMed]
  54. Park, E.; Lee, S.M.; Lee, J.; Kim, J.H. Anti-Inflammatory Activity of Mulberry Leaf Extract through Inhibition of NF-ΚB. J. Funct. Foods 2013, 5, 178–186. [Google Scholar] [CrossRef]
  55. Tilak, R.; Rahmathulla, V.K.; Rajan, R.K. Influence of Moisture Content of Mulberry Leaf on Growth and Silk Production in Bombyx mori L. Casp. J. Environ. Sci. 2006, 4, 25–30. [Google Scholar]
  56. Thangapandiyan, S.; Dharanipriya, R. Comparative Study of Nutritional and Economical Parameters of Silkworm (Bombyx mori) Treated with Silver Nanoparticles and Spirulina. J. Basic Appl. Zool. 2019, 80, 1–12. [Google Scholar] [CrossRef] [Green Version]
  57. Abdelkarim, S.; Mohamed, G.S. Effect of Pollen a Bee-Hive Product on Some Physiological, Biochemical and Economical Characteristics of Silkworm, Bombyx mori L. SVU Int. J. Agric. Sci. 2022, 4, 79–89. [Google Scholar] [CrossRef]
  58. Bhatti, M.F.; Azizullah; Shahzadi, N.; Tahir, H.M.; Ali, S.; Zahid, M.T.; Khurshid, R. Effect of Honey (Apis Dorsata [Hymenoptera: Apidae]) on Larval Growth and Silk Cocoon Yield of Bombyx mori (Lepidoptera: Bombycidae). J. Insect Sci. 2019, 19, 11. [Google Scholar] [CrossRef]
  59. He, Z.; Fang, Y.; Li, D.C.; Chen, D.S.; Wu, F. Effect of Lactic Acid Supplementation on the Growth and Reproduction of Bombyx mori (Lepidopteria: Bombycidae). J. Insect Sci. 2021, 21, 7. [Google Scholar] [CrossRef]
  60. Lineke, N.; Tulung, M.; Pelealu, J.; Tulung, B. The Effect of Palm Sugar Supplementation on Feed Consumption and Percentage of Silkworms (Bombyx mori L.) in the End of Instar V. Sci. Pap. Ser. Manag. Econ. Eng. Agric. Rural Dev. 2019, 19, 377–381. [Google Scholar]
  61. Marin, G.; Arivoli, S.; Tennyson, S. Effect of Micronutrient Supplemented Mulberry Leaves on Larvae of Silk Worm Bombyx mori. Indian J. Entomol. 2022, 1–7. [Google Scholar] [CrossRef]
  62. Rani, G.; Padmalatha, C.; Raj, S.R.; Singh, A.J.A.R. Impact of Supplementation of Amway Protein on the Economic Characters and Energy Budget of Silkworm Bombyx mori L. Asian J. Anim. Sci. 2011, 5, 190–195. [Google Scholar] [CrossRef] [Green Version]
  63. Nazim, N.; Buhroo, Z.I.; Mushtaq, N.; Javid, K.; Rasool, S.; Mir, G.M. Medicinal Values of Products and by Products of Sericulture. J. Pharmacogn. Phytochem. 2017, 6, 1388–1392. [Google Scholar]
  64. Qi, Y.; Wang, H.; Wei, K.; Yang, Y.; Zheng, R.Y.; Kim, I.S.; Zhang, K.Q. A Review of Structure Construction of Silk Fibroin Biomaterials from Single Structures to Multi-Level Structures. Int. J. Mol. Sci. 2017, 18, 237. [Google Scholar] [CrossRef] [PubMed]
  65. Jastrzebska, K.; Kucharczyk, K.; Florczak, A.; Dondajewska, E.; Mackiewicz, A.; Dams-Kozlowska, H. Silk as an Innovative Biomaterial for Cancer Therapy. Rep. Pract. Oncol. Radiother. 2015, 20, 87–98. [Google Scholar] [CrossRef] [PubMed]
  66. Asakura, T. Structure of Silk I (Bombyx mori Silk Fibroin before Spinning) -Type II β-Turn, not α-Helix. Molecules 2021, 26, 3706. [Google Scholar] [CrossRef]
  67. Vepari, C.; Kaplan, D.L. Silk as a Biomaterial. Prog. Polym. Sci. 2007, 32, 991–1007. [Google Scholar] [CrossRef]
  68. Bhattacharjee, P.; Gupta, P.; Joseph Christakiran, M.; Nandi, S.K.; Mandal, B.B. Silk-Based Matrices for Bone Tissue Engineering Applications. In Nanostructures for the Engineering of Cells, Tissues and Organs: From Design to Applications; Elsevier: Amsterdam, The Netherlands, 2018; pp. 439–472. [Google Scholar] [CrossRef]
  69. Seib, F.P.; Pritchard, E.M.; Kaplan, D.L. Self-Assembling Doxorubicin Silk Hydrogels for the Focal Treatment of Primary Breast Cancer. Adv. Funct. Mater. 2013, 23, 58–65. [Google Scholar] [CrossRef]
  70. Blake, S.; Kim, N.Y.; Kong, N.; Ouyang, J.; Tao, W. Silk’s Cancer Applications as a Biodegradable Material. Mater. Today Sustain. 2021, 13, 100069. [Google Scholar] [CrossRef]
  71. Talley, N.J. Functional Gastrointestinal Disorders as a Public Health Problem. Neurogastroenterol. Motil. 2008, 20, 121–129. [Google Scholar] [CrossRef]
  72. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, 49–66. [Google Scholar] [CrossRef] [Green Version]
  73. Soccol, C.R.; Porto De Souza Vandenberghe, L.; Spier, M.R.; Bianchi, A.; Medeiros, P.; Yamaguishi, C.T.; De, J.; Lindner, D.; Pandey, A.; Thomaz-Soccol, V. The Potential of Probiotics: A Review. Food Technol. Biotechnol. 2010, 48, 413–434. [Google Scholar]
  74. Zhu, Y.; Wang, Z.; Bai, L.; Deng, J.; Zhou, Q. Biomaterial-Based Encapsulated Probiotics for Biomedical Applications: Current Status and Future Perspectives. Mater. Des. 2021, 210, 110018. [Google Scholar] [CrossRef]
  75. Muhardina, V.; Ermaya, D.; Aisyah, Y.; Haryani, S. Quality Evaluation of Probiotic Capsule Prepared from Alginate, Carrageenan and Tofu Waste Flour Based on Bacterial Activity and Organoleptic Test. In IOP Conference Series: Earth and Environmental Science; Institute of Physics Publishing: Bristol, UK, 2018; Volume 122. [Google Scholar] [CrossRef]
  76. Diep, E.; Schiffman, J.D. Encapsulating Bacteria in Alginate-Based Electrospun Nanofibers. Biomater. Sci. 2021, 9, 4364–4373. [Google Scholar] [CrossRef] [PubMed]
  77. Hu, Y.; Lee, A.; Chang, S.L.; Shien-Fong, L. Biomaterial-Induced Conversion of Quiescent Cardiomyocytes into Pacemaker Cells in Rats. Nat. Biomed. Eng. 2022, 6, 421–434. [Google Scholar] [CrossRef] [PubMed]
  78. Phamornnak, C.; Han, B.; Spencer, B.F.; Ashton, M.D.; Blanford, C.F.; Hardy, J.G.; Blaker, J.J.; Cartmell, S.H. Instructive Electroactive Electrospun Silk Fibroin-Based Biomaterials for Peripheral Nerve Tissue Engineering. Biomater. Adv. 2022, 141, 213094. [Google Scholar] [CrossRef]
  79. Liu, S.; Gao, X.; Wang, Y.; Wang, J.; Qi, X.; Dong, K.; Shi, D.; Wu, X.; Guo, C. Baicalein-Loaded Silk Fibroin Peptide Nanofibers Protect against Cisplatin-Induced Acute Kidney Injury: Fabrication, Characterization and Mechanism. Int. J. Pharm. 2022, 626, 122161. [Google Scholar] [CrossRef]
  80. Lee, Y.J.; Lee, J.S.; Ajiteru, O.; Lee, O.J.; Lee, J.S.; Lee, H.; Kim, S.W.; Park, J.W.; Kim, K.Y.; Choi, K.Y.; et al. Biocompatible Fluorescent Silk Fibroin Bioink for Digital Light Processing 3D Printing. Int. J. Biol. Macromol. 2022, 213, 317–327. [Google Scholar] [CrossRef]
  81. Lau, K.; Waterhouse, A.; Akhavan, B.; Gao, L.; Kim, H.N.; 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]
  82. Gharehnazifam, Z.; Dolatabadi, R.; Baniassadi, M.; Shahsavari, H.; Kajbafzadeh, A.M.; Abrinia, K.; Gharehnazifam, K.; Baghani, M. Multiphysics Modeling and Experiments on Ultrasound-Triggered Drug Delivery from Silk Fibroin Hydrogel for Wilms Tumor. Int. J. Pharm. 2022, 621, 121787. [Google Scholar] [CrossRef]
  83. Phan, V.H.G.; Murugesan, M.; Nguyen, P.P.T.; Luu, C.H.; Le, N.H.H.; Nguyen, H.T.; Manivasagan, P.; Jang, E.S.; Li, Y.; Thambi, T. Biomimetic Injectable Hydrogel Based on Silk Fibroin/Hyaluronic Acid Embedded with Methylprednisolone for Cartilage Regeneration. Colloids Surf. B Biointerfaces 2022, 219, 112859. [Google Scholar] [CrossRef]
  84. Lu, X.; Sun, Y.; Han, M.; Chen, D.; Wang, A.; Sun, K. Silk Fibroin Double-Layer Microneedles for the Encapsulation and Controlled Release of Triptorelin. Int. J. Pharm. 2022, 613, 121433. [Google Scholar] [CrossRef]
  85. Crakes, K.R.; Herrera, C.; Morgan, J.L.; Olstad, K.; Hessell, A.J.; Ziprin, P.; Liwang, P.J.; Dandekar, S. Efficacy of Silk Fibroin Biomaterial Vehicle for in Vivo Mucosal Delivery of Griffithsin and Protection against HIV and SHIV Infection Ex Vivo. J. Int. AIDS Soc. 2020, 23, e25628. [Google Scholar] [CrossRef]
  86. Križman, K.; Novak, S.; Kristl, J.; Majdič, G.; Drnovšek, N. Long-Acting Silk Fibroin Xerogel Delivery Systems for Controlled Release of Estradiol. J. Drug Deliv. Sci. Technol. 2021, 65, 102701. [Google Scholar] [CrossRef]
  87. Li, Z.; Meng, Z.; Zhao, Z. Silk Fibroin Nanofibrous Scaffolds Incorporated with MicroRNA-222 Loaded Chitosan Nanoparticles for Enhanced Neuronal Differentiation of Neural Stem Cells. Carbohydr. Polym. 2022, 277, 118791. [Google Scholar] [CrossRef] [PubMed]
  88. Liang, Y.; Mitriashkin, A.; Lim, T.T.; Goh, J.C.H. Conductive Polypyrrole-Encapsulated Silk Fibroin Fibers for Cardiac Tissue Engineering. Biomaterials 2021, 276, 121008. [Google Scholar] [CrossRef] [PubMed]
  89. Liu, X.; Sun, Y.; Chen, B.; Li, Y.; Zhu, P.; Wang, P.; Yan, S.; Li, Y.; Yang, F.; Gu, N. Novel Magnetic Silk Fibroin Scaffolds with Delayed Degradation for Potential Long-Distance Vascular Repair. Bioact. Mater. 2022, 7, 126–143. [Google Scholar] [CrossRef]
  90. Liu, J.; Chen, H.; Wang, Y.; Li, G.; Zheng, Z.; Kaplan, D.L.; Wang, X.; Wang, X. Flexible Water-Absorbing Silk-Fibroin Biomaterial Sponges with Unique Pore Structure for Tissue Engineering. ACS Biomater. Sci. Eng. 2020, 6, 1641–1649. [Google Scholar] [CrossRef]
  91. Baptista, M.; Joukhdar, H.; Alcala-Orozco, C.R.; Lau, K.; Jiang, S.; Cui, X.; He, S.; Tang, F.; Heu, C.; Woodfield, T.B.F.; et al. Silk Fibroin Photo-Lyogels Containing Microchannels as a Biomaterial Platform for: In Situ Tissue Engineering. Biomater. Sci. 2020, 8, 7093–7105. [Google Scholar] [CrossRef]
  92. Qi, Z.; Cao, J.; Tao, X.; Wu, X.; Kundu, S.C.; Lu, S. Silk Fibroin Microneedle Patches for the Treatment of Insomnia. Pharmaceutics 2021, 13, 2198. [Google Scholar] [CrossRef]
  93. Chou, K.C.; Chen, C.T.; Cherng, J.H.; Li, M.C.; Wen, C.C.; Hu, S.I.; Wang, Y.W. Cutaneous Regeneration Mechanism of β-Sheet Silk Fibroin in a Rat Burn Wound Healing Model. Polymers 2021, 13, 3537. [Google Scholar] [CrossRef]
  94. Sun, M.; Li, Q.; Yu, H.; Cheng, J.; Wu, N.; Shi, W.; Zhao, F.; Shao, Z.; Meng, Q.; Chen, H.; et al. Cryo-Self-Assembled Silk Fibroin Sponge as a Biodegradable Platform for Enzyme-Responsive Delivery of Exosomes. Bioact. Mater. 2022, 8, 505–514. [Google Scholar] [CrossRef]
  95. Kiritani, S.; Kaneko, J.; Ito, D.; Morito, M.; Ishizawa, T.; Akamatsu, N.; Tanaka, M.; Iida, T.; Tanaka, T.; Tanaka, R.; et al. Silk Fibroin Vascular Graft: A Promising Tissue-Engineered Scaffold Material for Abdominal Venous System Replacement. Sci. Rep. 2020, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
  96. Lovati, A.B.; Lopa, S.; Bottagisio, M.; Talò, G.; Canciani, E.; Dellavia, C.; Alessandrino, A.; Biagiotti, M.; Freddi, G.; Segatti, F.; et al. Peptide-Enriched Silk Fibroin Sponge and Trabecular Titanium Composites to Enhance Bone Ingrowth of Prosthetic Implants in an Ovine Model of Bone Gaps. Front. Bioeng. Biotechnol. 2020, 8, 563203. [Google Scholar] [CrossRef] [PubMed]
  97. Shimada, K.; Higuchi, A.; Kubo, R.; Murakami, T.; Nakazawa, Y.; Tanaka, R. The Effect of a Silk Fibroin/Polyurethane Blend Patch on Rat Vessels. Organogenesis 2017, 13, 115–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Petrovska, B.B. Historical Review of Medicinal Plants’ Usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [Green Version]
  99. Jamshidi-Kia, F.; Lorigooini, Z.; Amini-khoei, H. Medicinal Plants: Past History and Future Perspective. J. Herbmed Pharmacol. 2018, 7, 1–7. [Google Scholar] [CrossRef]
  100. Sofowora, A.; Ogunbodede, E.; Onayade, A.; Dentistry, C. The Role and Place of Medicinal Plants In The Strategies For Disease Prevention. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 210–229. [Google Scholar] [CrossRef]
  101. Londhe, V.P.; Gavasane, A.T.; Nipate, S.S.; Bandawane, D.D.; Chaudhari, P.D. Role of Garlic (Allium Sativum) in Various Diseases: An Overview. J. Pharm. Res. Opin. 2011, 1, 13. [Google Scholar]
  102. Klimek-Szczykutowicz, M.; Szopa, A.; Ekiert, H. Citrus Limon (Lemon) Phenomenon—A Review of the Chemistry, Pharmacological Properties, Applications in the Modern Pharmaceutical, Food, and Cosmetics Industries, and Biotechnological Studies. Plants 2020, 9, 119. [Google Scholar] [CrossRef] [Green Version]
  103. Pervin, M.; Hasnat, M.A.; Lim, B.O. Antibacterial and Antioxidant Activities of Vaccinium Corymbosum L. Leaf Extract. Asian Pac. J. Trop. Dis. 2013, 3, 444–453. [Google Scholar] [CrossRef]
  104. Hao, J.; Gao, Y.; Xue, J.; Yang, Y.; Yin, J.; Wu, T. Phytochemicals, Pharmacological Effects and Molecular Mechanisms of Mulberry. Foods 2022, 11, 1170. [Google Scholar] [CrossRef]
  105. Ramesh, H.L.; Sivaram, V.; Murthy, V.N.Y. Antioxidant and Medicinal Properties of Mulberry (Morus Sp.): A Review. World J. Pharm. Res. 2014, 3, 320–343. [Google Scholar]
  106. Memete, A.R.; Timar, A.V.; Vuscan, A.N.; Groza, F.M.; Venter, A.C.; Vicas, S.I. Phytochemical Composition of Different Botanical Parts of Morus Species, Health Benefits and Application In food industry. Plants 2022, 11, 152. [Google Scholar] [CrossRef] [PubMed]
  107. Yuan, Q.; Zhao, L. The Mulberry (Morus alba L.) Fruit—A Review of Characteristic Components and Health Benefits. J. Agric. Food Chem. 2017, 65, 10383–10394. [Google Scholar] [CrossRef] [PubMed]
  108. Rani, R.; Arora, S.; Kaur, J.; Manhas, R.K. Phenolic Compounds as Antioxidants and Chemopreventive Drugs from Streptomyces Cellulosae Strain TES17 Isolated from Rhizosphere of Camellia Sinensis. BMC Complement. Altern. Med. 2018, 18, 1–15. [Google Scholar] [CrossRef] [PubMed]
  109. Lin, D.; Xiao, M.; Zhao, J.; Li, Z.; Xing, B. An Overview of Plant Phenolic Compounds and Their Importance in Human Nutrition and Management of Type 2 Diabetes. Molecules 2016, 21, 1374. [Google Scholar] [CrossRef]
  110. Cheynier, V. Phenolic Compounds: From Plants to Foods. Phytochem. Rev. 2012, 11, 153–177. [Google Scholar] [CrossRef]
  111. Zhu, Z.; Jun-jie, J.; Jie, Y.U.; Xiang-bing, M.A.O.; Bing, Y.U.; Dai-wen, C. Effect of Dietary Supplementation with Mulberry (Morus alba L.) Leaves on the Growth Performance, Meat Quality and Antioxidative Capacity of Finishing Pigs. J. Integr. Agric. 2019, 18, 143–151. [Google Scholar] [CrossRef]
  112. Hu, L.; Chen, D.; Zhou, W.; Chen, X.; Zhang, Q. Effects of Growth Period and Storage Methods on Primary Metabolite Contents and Antioxidant Activities of Morus alba L. Leaf. Molecules 2023, 28, 148. [Google Scholar] [CrossRef]
  113. Horng, C.; Liu, Z.; Huang, Y.; Lee, H. Extract from Mulberry (Morus Australis) Leaf Decelerate Acetaminophen Induced Hepatic Inflammation Involving Downregulation of Myeloid Differentiation Factor 88 (MyD88) Signals. J. Food Drug Anal. 2016, 25, 862–871. [Google Scholar] [CrossRef] [Green Version]
  114. Gu, P.S.; Moon, M.; Choi, J.G.; Oh, M.S. Mulberry Fruit Ameliorates Parkinson’s-Disease-Related Pathology by Reducing α -Synuclein and Ubiquitin Levels in a. J. Nutr. Biochem. 2017, 39, 15–21. [Google Scholar] [CrossRef]
  115. Hu, L.; Wang, C.; Guo, X.; Chen, D.; Zhou, W.; Chen, X. Flavonoid Levels and Antioxidant Capacity of Mulberry Leaves: Effects of Growth Period and Drying Methods. Front. Plant Sci. 2021, 12, 1–11. [Google Scholar] [CrossRef] [PubMed]
  116. Jaruchotikamol, A.; Pannangpetch, P. Cytoprotective Activity of Mulberry Leaf Extract against Oxidative Stress-Induced Cellular Injury in Rats. Pak. J. Pharm. Sci. 2013, 26, 163–168. [Google Scholar]
  117. Xv, Z.C.; He, G.L.; Wang, X.L.; Shun, H.; Chen, Y.J.; Lin, S.M. Mulberry Leaf Powder Ameliorate High Starch-Induced Hepatic Oxidative Stress and Inflammation in Fish Model. Anim. Feed Sci. Technol. 2021, 278, 114834. [Google Scholar] [CrossRef]
  118. Li, R.; Wang, C.; Chen, Y.; Li, N.; Wang, Q.; Zhang, M.; He, C.; Chen, H. A Combined Network Pharmacology and Molecular Biology Approach to Investigate the Active Ingredients and Potential Mechanisms of Mulberry (Morus alba L.) Leaf on Obesity. Phytomedicine 2021, 92, 153714. [Google Scholar] [CrossRef]
  119. Tu, J.; Shi, D.; Wen, L.; Jiang, Y.; Zhao, Y.; Yang, J.; Liu, H.; Liu, G.; Yang, B. Identification of Moracin N in Mulberry Leaf and Evaluation of Antioxidant Activity. Food Chem. Toxicol. 2019, 132, 110730. [Google Scholar] [CrossRef] [PubMed]
  120. Sun, C.; Tang, X.; Shao, X.; Han, D.; Zhang, H.; Shan, Y.; Gooneratne, R.; Shi, L.; Wu, X.; Hosseininezhad, M. Mulberry (Morus Atropurpurea Roxb.) Leaf Protein Hydrolysates Ameliorate Dextran Sodium Sulfate-Induced Colitis via Integrated Modulation of Gut Microbiota and Immunity. J. Funct. Foods 2021, 84, 104575. [Google Scholar] [CrossRef]
  121. Fan, L.; Peng, Y.; Wu, D.; Hu, J.; Shi, X.; Yang, G.; Li, X. Dietary Supplementation of Morus Nigra L. Leaves Decrease Fat Mass Partially through Elevating Leptin-Stimulated Lipolysis in Pig Model. J. Ethnopharmacol. 2020, 249, 112416. [Google Scholar] [CrossRef] [PubMed]
  122. Cao, Y.; Jiang, W.; Bai, H.; Li, J.; Zhu, H.; Xu, L.; Li, Y.; Li, K.; Tang, H.; Duan, W.; et al. Study on Active Components of Mulberry Leaf for the Prevention and Treatment of Cardiovascular Complications of Diabetes. J. Funct. Foods 2021, 83, 104549. [Google Scholar] [CrossRef]
  123. Wen, L.; Zhou, T.; Jiang, Y.; Gong, L.; Yang, B. Identification of Prenylated Phenolics in Mulberry Leaf and Their Neuroprotective Activity. Phytomedicine 2021, 90, 153641. [Google Scholar] [CrossRef]
  124. Li, R.; Xue, Z.; Jia, Y.; Wang, Y.; Li, S.; Zhou, J.; Liu, J.; Zhang, M.; He, C.; Chen, H. Polysaccharides from Mulberry (Morus alba L.) Leaf Prevents Obesity by Inhibiting Pancreatic Lipase in High-Fat Diet Induced Mice. Int. J. Biol. Macromol. 2021, 192, 452–460. [Google Scholar] [CrossRef]
  125. Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural Product-Based Nanoformulations for Cancer Therapy: Opportunities and Challenges. Semin. Cancer Biol. 2021, 69, 5–23. [Google Scholar] [CrossRef] [PubMed]
  126. Wathoni, N.; Nguyen, A.N.; Rusdin, A.; Umar, A.K.; Mohammed, A.F.A.; Motoyama, K.; Joni, I.M.; Muchtaridi, M. Enteric-Coated Strategies in Colorectal Cancer Nanoparticle Drug Delivery System. Drug Des. Dev. Ther. 2020, 14, 4387–4405. [Google Scholar] [CrossRef] [PubMed]
  127. Mottaghitalab, F.; Kiani, M.; Farokhi, M.; Kundu, S.C.; Reis, R.L.; Gholami, M.; Bardania, H.; Dinarvand, R.; Geramifar, P.; Beiki, D.; et al. Targeted Delivery System Based on Gemcitabine-Loaded Silk Fibroin Nanoparticles for Lung Cancer Therapy. ACS Appl. Mater. Interfaces 2017, 9, 31600–31611. [Google Scholar] [CrossRef]
  128. Zare-Zardini, H.; Vojdani Nejad Yazdi, S.; Zandian, A.; Zare, F.; Miresmaeili, S.M.; Dehghan-Manshadi, M.; Fesahat, F. Synthesis, Characterization, and Biological Evaluation of Doxorubicin Containing Silk Fibroin Micro- and Nanoparticles. J. Indian Chem. Soc. 2021, 98, 100161. [Google Scholar] [CrossRef]
  129. Wu, P.; Liu, Q.; Wang, Q.; Qian, H.; Yu, L.; Liu, B.; Li, R. Novel Silk Fibroin Nanoparticles Incorporated Silk Fibroin Hydrogel for Inhibition of Cancer Stem Cells and Tumor Growth. Int. J. Nanomed. 2018, 13, 5405–5418. [Google Scholar] [CrossRef]
  130. Zhou, J.; Sun, M.; Jin, S.; Fan, L.; Zhu, W.; Sui, X.; Cao, L.; Yang, C.; Han, C. Combined Using of Paclitaxel and Salinomycin Active Targeting Nanostructured Lipid Carriers against Non-Small Cell Lung Cancer and Cancer Stem Cells. Drug Deliv. 2019, 26, 281–289. [Google Scholar] [CrossRef]
  131. Lee, H.G.; Shin, S.J.; Chung, H.W.; Kwon, S.H.; do Cha, S.; Lee, J.E.; Cho, C.H. Salinomycin Reduces Stemness and Induces Apoptosis on Human Ovarian Cancer Stem Cell. J. Gynecol. Oncol. 2017, 28, e14. [Google Scholar] [CrossRef] [Green Version]
  132. Suyamud, C.; Phetdee, C.; Jaimalai, T.; Prangkio, P. Silk Fibroin-Coated Liposomes as Biomimetic Nanocarrier for Long-Term Release Delivery System in Cancer Therapy. Molecules 2021, 26, 4936. [Google Scholar] [CrossRef]
  133. 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] [Green Version]
  134. Nasrine, A.; Gulzar Ahmed, M.; Narayana, S. Silk Fibroin-Anastrozole Loaded Prolonged-Release Biodegradable Nanomedicine: A Promising Drug Delivery System for Breast Cancer Therapy. Mater. Today Proc. 2022, 68, 56–65. [Google Scholar] [CrossRef]
  135. Tan, M.; Liu, W.; Liu, F.; Zhang, W.; Gao, H.; Cheng, J.; Chen, Y.; Wang, Z.; Cao, Y.; Ran, H. Silk Fibroin-Coated Nanoagents for Acidic Lysosome Targeting by a Functional Preservation Strategy in Cancer Chemotherapy. Theranostics 2019, 9, 961–973. [Google Scholar] [CrossRef] [PubMed]
  136. Adali, T.; Tulay, P.; Altinoglu, G. Micro-Encapsulation of Anticancer Drug Carboplatin in Silk Fibroin and Chitosan Nano/Micro Particles as Breast Cancer Treatment. J. Biotechnol. 2019, 305, S65. [Google Scholar] [CrossRef]
  137. Qu, J.; Liu, Y.; Yu, Y.; Li, J.; Luo, J.; Li, M. Silk Fibroin Nanoparticles Prepared by Electrospray as Controlled Release Carriers of Cisplatin. Mater. Sci. Eng. C 2014, 44, 166–174. [Google Scholar] [CrossRef] [PubMed]
  138. Lerdchai, K.; Kitsongsermthon, J.; Ratanavaraporn, J.; Kanokpanont, S.; Damrongsakkul, S. Thai Silk Fibroin/Gelatin Sponges for the Dual Controlled Release of Curcumin and Docosahexaenoic Acid for Anticancer Treatment. J. Pharm. Sci. 2016, 105, 221–230. [Google Scholar] [CrossRef]
  139. Norouzi, P.; Motasadizadeh, H.; Atyabi, F.; Dinarvand, R.; Gholami, M.; Farokhi, M.; Shokrgozar, M.A.; Mottaghitalab, F. Combination Therapy of Breast Cancer by Codelivery of Doxorubicin and Survivin SiRNA Using Polyethylenimine Modified Silk Fibroin Nanoparticles. ACS Biomater. Sci. Eng. 2021, 7, 1074–1087. [Google Scholar] [CrossRef]
  140. Fuster, M.G.; Carissimi, G.; Montalbán, M.G.; Víllora, G. Antitumor Activity of Rosmarinic Acid-Loaded Silk Fibroin Nanoparticles on Hela and MCF-7 Cells. Polymers 2021, 13, 3169. [Google Scholar] [CrossRef]
  141. Tian, Y.; Jiang, X.; Chen, X.; Shao, Z.; Yang, W. Doxorubicin-Loaded Magnetic Silk Fibroin Nanoparticles for Targeted Therapy of Multidrug-Resistant Cancer. Adv. Mater. 2014, 26, 7393–7398. [Google Scholar] [CrossRef]
  142. Peng, Z.; Li, M.; Wang, Y.; Yang, H.; Wei, W.; Liang, M.; Shi, J.; Liu, R.; Li, R.; Zhang, Y.; et al. Self-Assembling Imageable Silk Hydrogels for the Focal Treatment of Osteosarcoma. Front. Cell Dev. Biol. 2022, 10, 698282. [Google Scholar] [CrossRef]
  143. Pham, D.T.; Saelim, N.; Tiyaboonchai, W. Paclitaxel Loaded EDC-Crosslinked Fibroin Nanoparticles: A Potential Approach for Colon Cancer Treatment. Drug Deliv. Transl. Res. 2020, 10, 413–424. [Google Scholar] [CrossRef]
  144. Yavuz, B.; Zeki, J.; Coburn, J.M.; Ikegaki, N.; Levitin, D.; Kaplan, D.L.; Chiu, B. In Vitro and in Vivo Evaluation of Etoposide—Silk Wafers for Neuroblastoma Treatment. J. Control. Release 2018, 285, 162–171. [Google Scholar] [CrossRef]
  145. Ornell, K.J.; Chiu, B.; Coburn, J.M. Development of a Dinutuximab Delivery System Using Silk Foams for GD2 Targeted Neuroblastoma Cell Death. J. Biomed. Mater. Res. A 2021, 109, 1393–1405. [Google Scholar] [CrossRef] [PubMed]
  146. Ding, B.; Wahid, M.A.; Wang, Z.; Xie, C.; Thakkar, A.; Prabhu, S.; Wang, J. Triptolide and Celastrol Loaded Silk Fibroin Nanoparticles Show Synergistic Effect against Human Pancreatic Cancer Cells. Nanoscale 2017, 9, 11739–11753. [Google Scholar] [CrossRef] [PubMed]
  147. Rahmani, H.; Fattahi, A.; Sadrjavadi, K.; Khaledian, S.; Shokoohinia, Y. Preparation and Characterization of Silk Fibroin Nanoparticles as a Potential Drug Delivery System for 5-Fluorouracil. Adv. Pharm. Bull. 2019, 9, 601–608. [Google Scholar] [CrossRef] [PubMed]
  148. Niu, L.; Chen, G.; Feng, Y.; Liu, X.; Pan, P.; Huang, L.; Guo, Y.; Li, M. Polyethylenimine-Modified Bombyx mori Silk Fibroin as a Delivery Carrier of the ING4-IL-24 Coexpression Plasmid. Polymers 2021, 13, 3592. [Google Scholar] [CrossRef] [PubMed]
  149. al Saqr, A.; Wani, S.U.D.; Gangadharappa, H.v.; Aldawsari, M.F.; Khafagy, E.S.; Abu Lila, A.S. Enhanced Cytotoxic Activity of Docetaxel-Loaded Silk Fibroin Nanoparticles against Breast Cancer Cells. Polymers 2021, 13, 1416. [Google Scholar] [CrossRef]
  150. Ma, P.; Gou, S.; Wang, M.; Chen, J.; Hu, W.; Xiao, B. Knitted Silk Fibroin-Reinforced Bead-on-String Electrospun Fibers for Sustained Drug Delivery Against Colon Cancer. Macromol. Mater. Eng. 2018, 303, 1700666. [Google Scholar] [CrossRef]
  151. Moin, A.; Wani, S.U.D.; Osmani, R.A.; Abu Lila, A.S.; Khafagy, E.S.; Arab, H.H.; Gangadharappa, H.v.; Allam, A.N. Formulation, Characterization, and Cellular Toxicity Assessment of Tamoxifen-Loaded Silk Fibroin Nanoparticles in Breast Cancer. Drug Deliv. 2021, 28, 1626–1636. [Google Scholar] [CrossRef] [PubMed]
  152. Zou, X.; Jiang, Z.; Li, L.; Huang, Z. Selenium Nanoparticles Coated with PH Responsive Silk Fibroin Complex for Fingolimod Release and Enhanced Targeting in Thyroid Cancer. Artif. Cells Nanomed. Biotechnol. 2021, 49, 83–95. [Google Scholar] [CrossRef]
  153. Balachander, G.M.; Kotcherlakota, R.; Nayak, B.; Kedaria, D.; Rangarajan, A.; Chatterjee, K. 3D Tumor Models for Breast Cancer: Whither We Are and What We Need. ACS Biomater. Sci. Eng. 2021, 7, 3470–3486. [Google Scholar] [CrossRef]
  154. Braccini, S.; Tacchini, C.; Chiellini, F.; Puppi, D. Polymeric Hydrogels for In Vitro 3D Ovarian Cancer Modeling. Int. J. Mol. Sci. 2022, 23, 3265. [Google Scholar] [CrossRef]
  155. Carvalho, M.R.; Lima, D.; Reis, R.L.; Correlo, V.M.; Oliveira, J.M. Evaluating Biomaterial- and Microfluidic-Based 3D Tumor Models. Trends Biotechnol. 2015, 33, 667–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Bahcecioglu, G.; Basara, G.; Ellis, B.W.; Ren, X.; Zorlutuna, P. Breast Cancer Models: Engineering the Tumor Microenvironment. Acta Biomater. 2020, 106, 1–21. [Google Scholar] [CrossRef] [PubMed]
  157. Buhome, O.; Wongwattanakul, M.; Daduang, J.; Limpaiboon, T. 3D Silk Fibroin-Gelatin/Hyaluronic Acid/Heparan Sulfate Scaffold Enhances Expression of Stemness and EMT Markers in Cholangiocarcinoma. In Vivo 2022, 36, 1155–1167. [Google Scholar] [CrossRef]
  158. Ribeiro, V.P.; Silva-Correia, J.; Gonçalves, C.; Pina, S.; Radhouani, H.; Montonen, T.; Hyttinen, J.; Roy, A.; Oliveira, A.L.; Reis, R.L.; et al. Rapidly Responsive Silk Fibroin Hydrogels as an Artificial Matrix for the Programmed Tumor Cells Death. PLoS ONE 2018, 13, e0194441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Dondajewska, E.; Juzwa, W.; Mackiewicz, A.; Dams-Kozlowska, H. Heterotypic Breast Cancer Model Based on a Silk Fibroin Scaffold to Study the Tumor Microenvironment. Oncotarget 2017, 9, 4935. [Google Scholar] [CrossRef] [PubMed]
  160. Niu, H.; Xiao, J.; Lou, X.; Guo, L.; Zhang, Y.; Yang, R.; Yang, H.; Wang, S.; Niu, F. Three-Dimensional Silk Fibroin/Chitosan Based Microscaffold for Anticancer Drug Screening. Front. Bioeng. Biotechnol. 2022, 10, 800830. [Google Scholar] [CrossRef]
  161. Li, J.; Zhou, Y.; Chen, W.; Yuan, Z.; You, B.; Liu, Y.; Yang, S.; Li, F.; Qu, C.; Zhang, X. A Novel 3D in Vitro Tumor Model Based on Silk Fibroin/Chitosan Scaffolds to Mimic the Tumor Microenvironment. ACS Appl. Mater. Interfaces 2018, 10, 36641–36651. [Google Scholar] [CrossRef]
  162. Jeong, H.-L.; Kang, E.-B.; Yun, S.-G.; Park, D.-b.; Lim, J.-O.; Suh, J.-S. Effect of a Silk Sericin and Methylsulfonylmethane (MSM) Blends on Inflammatory Response and Wound Healing. Appl. Sci. 2023, 13, 288. [Google Scholar] [CrossRef]
  163. Ashraaf, S.; Muhammad Tahir, H.; Yar Khan, S.; Tahir, H.M.; Ashraaf, S. Synergistic Effect of Silk Sericin and Curcumin to Treat an Inflammatory Condition. J. Burn Care Res. 2023, 44, 106–113. [Google Scholar] [CrossRef]
  164. Suzuki, S.; Sakiragaoglu, O.; Chirila, T.v. Study of the Antioxidative Effects of Bombyx mori Silk Sericin in Cultures of Murine Retinal Photoreceptor Cells. Molecules 2022, 27, 4635. [Google Scholar] [CrossRef]
  165. 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] [Green Version]
  166. Radu, I.C.; Zaharia, C.; Hudiță, A.; Tanasă, E.; Ginghină, O.; Marin, M.; Gălățeanu, B.; Costache, M. In Vitro Interaction of Doxorubicin-Loaded Silk Sericin Nanocarriers with Mcf-7 Breast Cancer Cells Leads to Dna Damage. Polymers 2021, 13, 2047. [Google Scholar] [CrossRef] [PubMed]
  167. Guo, W.; Deng, L.; Yu, J.; Chen, Z.; Woo, Y.; Liu, H.; Li, T.; Lin, T.; Chen, H.; Zhao, M.; et al. Sericin Nanomicelles with Enhanced Cellular Uptake and Ph-Triggered Release of Doxorubicin Reverse Cancer Drug Resistance. Drug Deliv. 2018, 25, 1103–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Gao, Y.E.; Hou, S.; Cheng, J.; Li, X.; Wu, Y.; Tang, Y.; Li, Y.; Xue, P.; Kang, Y.; Xu, Z.; et al. Silk Sericin-Based Nanoparticle as the Photosensitizer Chlorin E6 Carrier for Enhanced Cancer Photodynamic Therapy. ACS Sustain. Chem. Eng. 2021, 9, 3213–3222. [Google Scholar] [CrossRef]
  169. Liu, J.; Deng, Y.; Fu, D.; Yuan, Y.; Li, Q.; Shi, L.; Wang, G.; Wang, Z.; Wang, L. Sericin Microparticles Enveloped with Metal-Organic Networks as a Pulmonary Targeting Delivery System for Intra-Tracheally Treating Metastatic Lung Cancer. Bioact. Mater. 2021, 6, 273–284. [Google Scholar] [CrossRef]
  170. Liu, J.; Qi, C.; Tao, K.; Zhang, J.; Zhang, J.; Xu, L.; Jiang, X.; Zhang, Y.; Huang, L.; Li, Q.; et al. Sericin/Dextran Injectable Hydrogel as an Optically Trackable Drug Delivery System for Malignant Melanoma Treatment. ACS Appl. Mater. Interfaces 2016, 8, 6411–6422. [Google Scholar] [CrossRef]
  171. Guo, W.; Deng, L.; Chen, Z.; Chen, Z.; Yu, J.; Liu, H.; Li, T.; Lin, T.; Chen, H.; Zhao, M.; et al. Vitamin B12-Conjugated Sericin Micelles for Targeting CD320-Overexpressed Gastric Cancer and Reversing Drug Resistance. Nanomedicine 2019, 14, 353–370. [Google Scholar] [CrossRef]
  172. Akbal, Ö. Sericin-Montmorillonite Composite Nanoparticles as Drug Delivery System in Human Liver Cancer: Development, Drug Release, Cellular Uptake and Cytotoxicity. Süleyman Demirel Üniversitesi Fen Bilimleri Enstitüsü Dergisi 2020, 24, 169–177. [Google Scholar] [CrossRef]
  173. Gagliardi, A.; Ambrosio, N.; Voci, S.; Salvatici, M.C.; Fresta, M.; Cosco, D. Easy Preparation, Characterization and Cytotoxic Investigation of 5-Fluorouracil-Loaded Zein/Sericin Nanoblends. J. Mol. Liq. 2022, 366, 120344. [Google Scholar] [CrossRef]
  174. Kanoujia, J.; Faizan, M.; Parashar, P.; Singh, N.; Saraf, S.A. Curcumin Loaded Sericin Nanoparticles: Assessment for Biomedical Application. Food Hydrocoll. Health 2021, 1, 100029. [Google Scholar] [CrossRef]
  175. Suktham, K.; Koobkokkruad, T.; Wutikhun, T.; Surassmo, S. Efficiency of Resveratrol-Loaded Sericin Nanoparticles: Promising Bionanocarriers for Drug Delivery. Int. J. Pharm. 2018, 537, 48–56. [Google Scholar] [CrossRef]
  176. Yalcin, E.; Kara, G.; Celik, E.; Pinarli, F.A.; Saylam, G.; Sucularli, C.; Ozturk, S.; Yilmaz, E.; Bayir, O.; Korkmaz, M.H.; et al. Preparation and Characterization of Novel Albumin-Sericin Nanoparticles as SiRNA Delivery Vehicle for Laryngeal Cancer Treatment. Prep. Biochem. Biotechnol. 2019, 49, 659–670. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, J.; Li, Q.; Zhang, J.; Huang, L.; Qi, C.; Xu, L.; Liu, X.; Wang, G.; Wang, L.; Wang, Z. Safe and Effective Reversal of Cancer Multidrug Resistance Using Sericin-Coated Mesoporous Silica Nanoparticles for Lysosome-Targeting Delivery in Mice. Small 2017, 13, 1602567. [Google Scholar] [CrossRef]
  178. Niu, L.; Yang, S.; Zhao, X.; Liu, X.; Si, L.; Wei, M.; Liu, L.; Cheng, L.; Qiao, Y.; Chen, Z. Sericin Inhibits MDA-MB-468 Cell Proliferation via the PI3K/Akt Pathway in Triple-negative Breast Cancer. Mol. Med. Rep. 2021, 23, 140. [Google Scholar] [CrossRef]
  179. Kumar, J.P.; Mandal, B.B. Silk Sericin Induced Pro-Oxidative Stress Leads to Apoptosis in Human Cancer Cells. Food Chem. Toxicol. 2019, 123, 275–287. [Google Scholar] [CrossRef]
  180. Shu, Y.H.; Yuan, H.H.; Xu, M.T.; Hong, Y.T.; Gao, C.C.; Wu, Z.P.; Han, H.T.; Sun, X.; Gao, R.L.; Yang, S.F.; et al. A Novel Diels–Alder Adduct of Mulberry Leaves Exerts Anticancer Effect through Autophagy-Mediated Cell Death. Acta Pharmacol. Sin. 2021, 42, 780–790. [Google Scholar] [CrossRef] [PubMed]
  181. Han, H.; Chou, C.C.; Li, R.; Liu, J.; Zhang, L.; Zhu, W.; Hu, J.; Yang, B.; Tian, J. Chalcomoracin Is a Potent Anticancer Agent Acting through Triggering Oxidative Stress via a Mitophagy- and Paraptosis-Dependent Mechanism. Sci. Rep. 2018, 8, 1–14. [Google Scholar] [CrossRef] [Green Version]
  182. Dalkiliç, S.; Dalkiliç, L.K.; İnci, Ş.; Korkmaz, İ.; Kırbağ, S. Investigation of Cytotoxic Effect of Black Mulberry (Morus Nigra L.) Fruit. Indian J. Tradit. Knowl. 2021, 20, 1–5. [Google Scholar]
  183. Fallah, S.; Hajihassan, Z.; Zarkar, N.; Rabbani-Chadegani, A.; Mohammadnejad, J.; Hajimirzamohammad, M. Evaluation of Anticancer Activity of Extracted Flavonoids from Morus alba Leaves and Its Interaction with DNA. Braz. Arch. Biol. Technol. 2018, 61, 18160623. [Google Scholar] [CrossRef]
  184. Erden, Y. Sour Black Mulberry (Morus Nigra L.) Causes Cell Death by Decreasing Mutant P53 Expression in HT-29 Human Colon Cancer Cells. Food Biosci. 2021, 42, 101113. [Google Scholar] [CrossRef]
  185. Long, H.L.; Zhang, F.F.; Wang, H.L.; Yang, W.S.; Hou, H.T.; Yu, J.K.; Liu, B. Mulberry Anthocyanins Improves Thyroid Cancer Progression Mainly by Inducing Apoptosis and Autophagy Cell Death. Kaohsiung J. Med. Sci. 2018, 34, 255–262. [Google Scholar] [CrossRef] [Green Version]
  186. Phan, T.N.; Kim, O.; Ha, M.T.; Hwangbo, C.; Min, B.S.; Lee, J.H. Albanol b from Mulberries Exerts Anti-Cancer Effect through Mitochondria Ros Production in Lung Cancer Cells and Suppresses in Vivo Tumor Growth. Int. J. Mol. Sci. 2020, 21, 9502. [Google Scholar] [CrossRef]
  187. Cho, E.; Chung, E.Y.; Jang, H.-Y.; Hong, O.-Y.; Chae, H.S.; Jeong, Y.-J.; Kim, S.-Y.; Kim, B.-S.; Yoo, D.J.; Kim, J.-S.; et al. Anti-Cancer Effect of Cyanidin-3-Glucoside from Mulberry via Caspase-3 Cleavage and DNA Fragmentation in Vitro and in Vivo. Anticancer Agents Med. Chem. 2017, 17, 1519–1525. [Google Scholar] [CrossRef] [PubMed]
  188. Cheng, K.C.; Wang, C.J.; Chang, Y.C.; Hung, T.W.; Lai, C.J.; Kuo, C.W.; Huang, H.P. Mulberry Fruits Extracts Induce Apoptosis and Autophagy of Liver Cancer Cell and Prevent Hepatocarcinogenesis in Vivo. J. Food Drug Anal. 2020, 28, 84–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Yu, J.S.; Lee, D.; Lee, S.R.; Lee, J.W.; Choi, C.I.; Jang, T.S.; Kang, K.S.; Kim, K.H. Chemical Characterization of Cytotoxic Indole Acetic Acid Derivative from Mulberry Fruit (Morus alba L.) against Human Cervical Cancer. Bioorg. Chem. 2018, 76, 28–36. [Google Scholar] [CrossRef] [PubMed]
  190. Turan, I.; Demir, S.; Kilinc, K.; Burnaz, N.A.; Yaman, S.O.; Akbulut, K.; Mentese, A.; Aliyazicioglu, Y.; Deger, O. Antiproliferative and Apoptotic Effect of Morus Nigra Extract on Human Prostate Cancer Cells. Saudi Pharm. J. 2017, 25, 241–248. [Google Scholar] [CrossRef] [PubMed]
  191. Deepa, M.; Sureshkumar, T.; Satheeshkumar, P.K.; Priya, S. Purified Mulberry Leaf Lectin (MLL) Induces Apoptosis and Cell Cycle Arrest in Human Breast Cancer and Colon Cancer Cells. Chem. Biol. Interact. 2012, 200, 38–44. [Google Scholar] [CrossRef]
  192. Feng, F.; Hu, P.; Tao, X. Mulberry Leaf Polysaccharide Extracted by Response Surface Methodolog Suppresses the Proliferation, Invasion and Migration of MCF-7 Breast Cancer Cells. Food Sci. Technol. 2022, 42. [Google Scholar] [CrossRef]
  193. Çakıroğlu, E.; Uysal, T.; Çalıbaşı Koçal, G.; Aygenli, F.; Baran, G.; Baskın, Y. The Role of Morus Nigra Extract and Its Active Compounds as Drug Candidate on Human Colorectal Adenocarcinoma Cell Line HT-29. Int. J. Clin. Oncol. Cancer Res. 2017, 2, 10–14. [Google Scholar] [CrossRef]
Figure 1. B. mori’s applicability in biomedicine (Created with BioRender.com, accessed on 23 November 2022)).
Figure 1. B. mori’s applicability in biomedicine (Created with BioRender.com, accessed on 23 November 2022)).
Molecules 28 00850 g001
Figure 2. Silk fibroin-based biomaterials (Created with BioRender.com, accessed on 23 November 2022)).
Figure 2. Silk fibroin-based biomaterials (Created with BioRender.com, accessed on 23 November 2022)).
Molecules 28 00850 g002
Figure 3. Main bioactive compounds in the mulberry (Created with BioRender.com, accessed on 23 November 2022).
Figure 3. Main bioactive compounds in the mulberry (Created with BioRender.com, accessed on 23 November 2022).
Molecules 28 00850 g003
Table 1. Clinically significant silk-based biomaterials.
Table 1. Clinically significant silk-based biomaterials.
BiomaterialTargetClinical SignificanceExperimental ModelReference
Silk fibroin (SF) hydrogelQuiescent ventricular cardiomyocytesBiological pacemakers cells production intended for human useRats[77]
Electrically conductive SF-based scaffoldsPeripheral nerve tissuePeripheral nerve injury regenerationIn vitro[78]
SF nanofibersKidney tissueRenal protectionMice[79]
Fluorescent SF bioink-hydrogel printingTissuesTo keep track of encapsulated cell and the evaluation of hydrogel degradationMice[80]
Biomimetic SF-based biomaterialsWhole blood and plateletBlood contacting devices production for diagnostic and therapyIn vitro; ex vivo[81]
SF hydrogelHuman Wilms tumorDrug deliveryIn vitro[82]
SF/hyaluronic acid injectable hydrogelsArticular cartilageCartilage defects regenerationMice[83]
SF microneedlesProstate glandDrugs sustained releaseRats[84]
SF discsMucosal tissueViral prevention and drug deliveryIn vivo, ex vivo[85]
SF xerogelsHormonesDrug deliveryIn vitro[86]
Chitin/SF nanoribbonsNeural tissueNeural tissue regenerationIn vitro[87]
SF nanofibersCardiac tissueCardiac tissue regenerationIn vitro[88]
SF scaffoldsVascular tissueLong-distance vascular defect restaurationRabbits[89]
SF spongesSoft tissuesWound injuries regenerationIn vitro[90]
Photo-lyogels SF-basedTissuesTissue engineeringIn vitro[91]
Microneedles patches SF-basedBloodTreatment of InsomniaRat[92]
SF dressingsCutaneous woundsBurn wound healingRat[93]
Cryo-sponges SF-basedBone marrow mesenchymal stem cellsExosome therapyMice[94]
Vascular graft SF basedAbdominal venous systemReplacement of abdominal venous systemRat[95]
SF sponges enriched with peptideCartilage and bone tissueBiological functionality improvement of materials that are used for prosthetic devicesOvine[96]
SF/Polyurethane patch graftsVesselsVascular diseases treatmentRat[97]
Table 2. Bioactivities of mulberry leaf extracts and their clinical significance.
Table 2. Bioactivities of mulberry leaf extracts and their clinical significance.
FormulationBioactivityClinical SignificanceExperimental ModelReference
PowderAntioxidant and anti-inflammatoryAlleviation of hepatic injuriesFish[117]
Flavonoids extractAnti-obesityObesity prevention and therapyIn vitro[118]
Moracin N extractAntioxidantMaintenance of oxidation-antioxidation balanceIn vitro[119]
Protein hydrolysatesAnti-inflammatoryPrevention and treatment of colitisMice[120]
Mulberry leaves dietary supplementationAnti-obesityRegulation of lipid metabolismPig[121]
Mulberry leaves extractHypoglycemic, antioxidative, cardioprotectiveProtection against diabetic myocardiumMice[122]
Moracin NAntioxidative, neuroprotectiveProtection against neurotoxicityIn vitro[123]
Mulberry leaf polysaccharidesAnti-obesityObesity prevention and treatmentIn vitro[124]
Table 3. Fibroin-based carriers for various cancer types.
Table 3. Fibroin-based carriers for various cancer types.
Carrier FormulationLoadingCancer TypeStudy TypePreparation MethodReference
NanoparticlesAnastrozoleBreast cancerIn vitroSolvent change[134]
NanoparticlesDoxorubicinNot specifiedIn vitro, in vivoGas diffusion method[135]
Silk fibroin (SF)/Chitosan nano- and microparticlesCarboplatinBreast cancerIn vivoIonotropic gelation[136]
NanoparticlesCisplatinLung cancerIn vivoElectrospray[137]
SF/Gelatin spongesCurcumin, docosahexaenoic acidCervical cancerIn vitroFreeze-drying; glutaraldehyde cross-linking[138]
Polyethylenimine-modified SF nanoparticlesDoxorubicin, surviving siRNABreast cancerIn vivoDropwise addition of acetone in SF solution (2% w/v)[139]
NanoparticlesRosmarinic acidCervical carcinoma and breast cancerIn vitroDissolution of SF in ionic liquid by using high power ultrasound[140]
Magnetic nanoparticles SFDoxorubicinBreast cancerIn vitroSalting-out precipitation of potassium phosphate and including hydrophilic magnetic iron oxide nanoparticles into the phosphate solution[141]
HydrogelsIodineOsteosarcomaIn vivoMixing Sf solution, polyethylene glycol 400, polyvinylpyrrolidone iodine, and meglumine diatrizoate[142]
NanoparticlesPaclitaxelColon cancer, breast cancerIn vitroDesolvation method[143]
SF wafersEtoposideNeuroblastomaIn vivoUtilization of a bench-top compression press[144]
SF foamDinutuximabNeuroblastomaIn vitroMixing SF, glycerol and dinutuximab[145]
NanoparticlesTriptolide, celastrolPancreatic cancerIn vitroDesolvation method[146]
Nanoparticles5-fluorouracilAdenocarcinomaIn vitroNanoprecipitation[147]
Gene delivery systemInhibitor of growth 4 and interleukin-24 co-expression plasmidLung cancerIn vitroFreeze-drying[148]
NanoparticlesDocetaxelBreast cancerIn vitroNanoprecipitation[149]
SF meshesCamptothecinColon cancerIn vitroElectrospinning process[150]
NanoparticlesTamoxifenBreast cancerIn vitroDesolvation method[151]
SF/Selenium nanoparticlesFingolimodThyroid cancerIn vivoFreeze-drying[152]
Table 4. Silk sericin carriers for anticancer therapy.
Table 4. Silk sericin carriers for anticancer therapy.
Carrier FormulationLoadingCancer TypeStudy TypeReference
NanoparticlesDoxorubicinBreast cancerIn vitro[166]
Sericin/poly(γ-benzyl-l-glutamate nanomicellesDoxorubicinHepatocellular carcinoma; breast cancerIn vitro; in vivo[167]
NanoparticlesChlorin e6Breast cancerIn vitro; in vivo[168]
MicroparticlesMetal-organic networks; DoxorubicinLung cancerIn vitro; in vivo[169]
Sericin/Dextran hydrogelDoxorubicinMelanomaIn vitro; in vivo[170]
Sericin/Synthetic poly(γ-benzyl-l-glutamatePaclitaxelGastric cancerIn vitro; in vivo[171]
Sericin-Montmorillonite nanoparticlesDoxorubicinHepatocarcinomaIn vitro[172]
Zein/Sericin nanoblends5-FluorouracilBreast cancer; colon carcinomaIn vitro[173]
NanoparticlesCurcuminNot specifiedIn vitro; in vivo[174]
NanoparticlesResveratrolColorectal carcinoma In vitro[175]
Albumin-Sericin nanoparticlesSmall interfering RNALaryngeal cancerIn vitro[176]
NanoparticlesDoxorubicinBreast cancerIn vitro; in vivo[177]
Table 5. The antitumoral activity of mulberry compounds.
Table 5. The antitumoral activity of mulberry compounds.
Mulberry SpeciesCompoundTarget CancerMolecular Cell Death BasisReference
M. nigraTotal flavonoids, phenolic compoundsColon cancerEnhanced expression level of Bax/Bcl-2 genes and a decrease expression ratio of p53 gene[184]
M. albaAnthocyaninsThyroid cancerApoptosis and autophagy[185]
M. albaAlbanol BLung cancerMitochondrial reactive oxygen species production[186]
M. albaCyanidin-3-glucosideBreast cancerCaspase-3 cleavage, DNA fragmentation[187]
M. albaPolyphenolsHepatocellular carcinomaApoptosis; autophagy, PI2K/Akt pathway[188]
M. albaIndole acetic acidCervical cancerCaspase-8 and -9 activation[189]
M. nigraPhenolic compoundsProstate adenocarcinomaEnhanced caspase activity; low mitochondrial membrane potential[190]
M. albaLectinBreast cancerCaspase dependent pathway[191]
M. albaPolysaccharidesBreast cancerNot applicable[192]
M. nigraMorniga G, chalcone 4 hydrateColorectal cancerNot applicable[193]
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

Baci, G.-M.; Baciu, E.-D.; Cucu, A.-A.; Muscă, A.-S.; Giurgiu, A.I.; Moise, A.R.; Zăhan, M.; Dezmirean, D.S. Sericultural By-Products: The Potential for Alternative Therapy in Cancer Drug Design. Molecules 2023, 28, 850. https://doi.org/10.3390/molecules28020850

AMA Style

Baci G-M, Baciu E-D, Cucu A-A, Muscă A-S, Giurgiu AI, Moise AR, Zăhan M, Dezmirean DS. Sericultural By-Products: The Potential for Alternative Therapy in Cancer Drug Design. Molecules. 2023; 28(2):850. https://doi.org/10.3390/molecules28020850

Chicago/Turabian Style

Baci, Gabriela-Maria, Ecaterina-Daniela Baciu, Alexandra-Antonia Cucu, Adriana-Sebastiana Muscă, Alexandru Ioan Giurgiu, Adela Ramona Moise, Marius Zăhan, and Daniel Severus Dezmirean. 2023. "Sericultural By-Products: The Potential for Alternative Therapy in Cancer Drug Design" Molecules 28, no. 2: 850. https://doi.org/10.3390/molecules28020850

Article Metrics

Back to TopTop