Next Article in Journal
Casein Micelles as an Emerging Delivery System for Bioactive Food Components
Next Article in Special Issue
Breba Fruits Characterization from Four Varieties (Ficus carica L.) with Important Commercial Interest in Spain
Previous Article in Journal
Metabolic Engineering of Pediococcus acidilactici BD16 for Heterologous Expression of Synthetic alaD Gene Cassette and L-Alanine Production in the Recombinant Strain Using Fed-Batch Fermentation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Activities for Morus alba L., Focusing on the Immunostimulatory Property from the Fruit Aqueous Extract

1
Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Jeonbuk, Iksan 54538, Korea
2
ForBioKorea Co., Ltd., Geumcheon-gu, Seoul 08592, Korea
*
Author to whom correspondence should be addressed.
Foods 2021, 10(8), 1966; https://doi.org/10.3390/foods10081966
Submission received: 14 July 2021 / Revised: 11 August 2021 / Accepted: 20 August 2021 / Published: 23 August 2021
(This article belongs to the Special Issue Recent Advances in the Biological Functions of Plant-Based Foods)

Abstract

:
Depending on the extraction method, numerous compounds that have specific pharmacological effects can be obtained from M. alba L. There is a growing scientific interest in health problems related to aging. Efforts to develop safe immune-enhancing pharmaceuticals are increasing. This review aims to summarize and critically discuss the immunity enhancement effects and pharmaceutical efficacy of M. alba L. extracts. The scientific database search was conducted using Google Scholar, Web of Science, and PubMed until May 2021. Additional articles were identified and obtained from references in the retrieved articles. Ethanol or methanol extraction of various parts of M. alba L. identified a large amount of phenols and flavonoids, which are effective for immunosuppression, antioxidants, and cardiovascular diseases, and are antibacterial, and anticancer. Water extraction of M. alba L. enhanced the innate immune response based on immune cell activation. A polysaccharide and an alkaloid related to increased macrophage activity were isolated from M. alba L. fruit extracts. M. alba L. fruit water extracts primarily induced the production of pro-inflammatory substances, in model organisms, via TLR4 in immune cells. Water extracts have been shown to be effective in pathogen defense and tumor suppression by enhancing macrophage activity. Based on our literature review on the bioactivity of M. alba L. fruit extracts, particularly in relation to their immunity enhancement activity, we anticipate that M. alba-derived pharmaceuticals will have excellent potential in future medical research.

1. Introduction

The individual fruit of the mulberry is small and round, with several fruits grouped together to form a single long oval-shaped panicle. In Korea, the mulberry fruit is sometimes referred to as “Oddi”. A single tree can be very high yielding, containing more than 10 kg of fruit. Mulberry berries are edible and are consumed worldwide, particularly in Asia. Mulberries change color from white to black as they mature and are commonly eaten fresh or processed into foods, such as wine, fruit juices, and jams [1,2,3,4]. Mulberry trees are deciduous plants belonging to the genus Morus (family Moraceae), which contains more than 17 species growing in tropical climates. These include Morus alba L., Morus rubra, Morus nigra, Morus australis, Morus atropurpurea, Morus cathayana, Morus notabilis, and Morus mesozygia [5,6,7]. The most common species are Morus alba (white mulberry) and Morus nigra (blackberry). There has been much research work that has been done since several reviews on the species have been published.
Chen et al. [8] reported that the total phenolic content of mulberry (M. alba L. fruits), reaching 502.43 ± 5.10 mg equivalents (QE)/100 g fresh weight (FW), is higher than blackberry, blueberry, raspberry, and strawberry, suggesting that mulberry can be used as good sources of phenolic compounds. Therefore, mulberry fruits are rich in diverse phenolic compounds, including polyphenols, anthocyanins, and flavonoids [9]. As shown in Table 1, M. alba (white mulberry) has a higher total dry weight, ash, protein, total carbohydrates, sugar, riboflavin, niacin, total phenols, and alkaloid than M. nigra (black mulberry) [10,11] except for pectin [10]. Y. jiang [12] reported that linoleic acid and α-linolenic acid showed higher contents in M. alba fruits, about 2.5- and 4-fold, respectively, compared to M. nigra fruits. Considering these results, M. alba L. contains active ingredients showing high nutritional importance and pharmaceutical effects among the genus Morus. Donguibogam and Bencao Gangmu, Morus alba L. have long been used as traditional medicine for diabetes, arthritis, rheumatism, and other disorders for thousands of years in East Asia [13,14,15].
Various parts of M. alba, including the roots, stems, leaves, and fruits, have long been used in China, Japan, and Korea as herbal medicines due to their pharmacological effects [18,19,20,21]. Figure 1 illustrates the morphology of M. alba. The root bark is called “Sang Bai Pi” and it is used as a diuretic and to treat lung heat and asthma. The dried branch (“Sang Zhi”), collected in spring and summer, is used alone or in combination with other medications to treat rheumatic or rheumatoid arthritis and muscular contractions. The leaves (Sang Ye) have a variety of culinary, medicinal, and industrial applications. They are very palatable and commonly used to make tinctures and herbal teas (a common health beverage in Asian countries). Leaf extracts are thought to cool the blood and stop bleeding and so are used to treat hemoptysis, epistaxis, and hematemesis. The fruits (“Sang Shen”) are thought to enhance liver and kidney function, nourish yin and the blood, and promote the secretion of saliva and moisten dryness [22].
The efficacy of M. alba extracts and their active ingredients have been scientifically tested in various cell and animal studies. These studies focused on numerous physiological activities, including the antioxidant, neuroprotective, antiarteriosclerosis, immunomodulation, antitumor, and antihyperlipidemia effects of extracts and active ingredients [2]. Among them, the immunomodulatory effects of M. alba extracts are attracting much attention due to an increasing societal interest in delaying ageing and increasing general well-being (e.g., the Lifestyles of Health and Sustainability (LOHAS) demographic) as well as the increasing age profile of certain populations [23,24]. This review summarizes and discusses recent research on the pharmaceutical efficacy of M. alba extracts in relation to various functions in the body, in particular immune activity. In this way, we provide information and guidance for future studies on the uses of M. alba in treating numerous medical conditions.

2. M. alba L. Plant Parts: Pharmacological Potential and Bioactive Phytochemical Composition

Root bark, stem bark, leaf, and fruit extracts taken from a single plant species (for medicinal purposes) generally have similar physiological effects on the human body. These may include anti-inflammatory, antioxidant, antidiabetic, antibacterial, anticancer, hepatoprotective, and cardiovascular system protective effects. However, some studies have shown that the physiological effect may differ depending on which plant part the extract was taken from. Table 2 summarizes the known pharmacological effects of M. alba plant parts (root, stem, leaves, and fruit).

2.1. Root

Methanol extracts of root bark (TRB) of M. alba have traditionally been used to treat blood pressure, stabilize blood sugar, and reduce fever [58,103]. Immune-enhancing effects of polysaccharides isolated from the water extract of Morus alba L. root [31] but immune-inhibiting effects of Kowanon G [104] and Cudraplavone B [42] isolated from methanol extracts were confirmed. Several studies have identified various phytochemical compounds in the root bark. In 1999, the compound moran 20 K, a glycoprotein with antioxidant and antidiabetic effects, was isolated from methanol extracts [70]. Zheng et al. [104] isolated the compounds moracinoside C, moracin O, and moracin P from water extracts of M. alba root bark. Khan et al. [51] investigated the antioxidant activity and phenolic content (total phenols, flavonoids, flavanols and proanthrocyanidins) of methanolic extracts taken from M. alba root bark (TRB). TSB showed the highest antioxidant activity then other parts’ extract [51]. These results indicate that there is a high correlation and regression between the phenolic contents and antioxidant potential of these extracts [10]. Ethanol extract from the dried root of M. australis (MRE) was also found to decrease CCl4-induced hepatic inflammation and necrosis in mice [92].

2.2. Stem

Stems extracts have been shown to have anti-inflammatory [37,105,106,107,108] and antiosteoarthritis [109] effects. Ethanol of M. alba stem extracts (MSEs) has been been found to suppress IL-6 and IL-8 expression in Porphyromonas gingivalis LPS-stimulated hPDL fibroblasts, indicating a possible anti-inflammatory effect. Chen et al. isolated Oxyresveratrol from the methanol of M. alba branch extracts [40]. Riviere et al. isolated one new coumarin glycoside, isoscopoletin 6-(6-O-β-apiofuranosyl-β-glucopyranoside), with seven known polyphenols from acetone of M. alba stem extracts using centrifugal partition chromatography (CPC) [110].

2.3. Leaf

The leaves of M. alba are used as food for silkworms. They are also one of the most important herbs used in the treatment of hyperglycemia. M. alba leaves are also used to treat diabetes mellitus [19,65,66,68,111,112]. According to the investigation by Hunyadi A. [65], an antidiabetic effect of Morus alba L. leaves was demonstrated by several iminosugars [113]. Flavonoids and related constituents [114], polysaccharides [70], volatile oil-like fraction of a hot water extract of M. alba leaves [62], and ecdysteroids [115] (20-hydroxyecdysone and inokosterone) are thought to play an important pharmcological role [111]. Ecdysteroids are also used in aquaculture of crab and shrimp in addition to their pharmacological effects [116]. Antimicrobial activity [73,74,76] and hepatoprotective effects [49,90] of the ethanol extract of M. alba leaves were also observed. The ethanol of M. alba leaf and fruit extract (MLFE) supplementation have been found to stimulate cutaneous NLRP3 inflammasomes in HFD-induced obese mice [117]. Chen et al. [28] isolated gamma aminobutyric acid from water extract of M. alba leaves by using biochemical methods. The neuroprotective effect of Morus alba L. extract is well documented in many studies. Among them, studies on ethanol or methanol extracts from leaves were studied by Chen et al. [96], Yadav et al. [97], and Kang et al. [98]. In terms of the leaf extract, many studies have evaluated the efficacy of the crude extract only, without separating the components.

2.4. Fruit

In the history of M. alba’s usage in medicine, the fruit has been the last part of the plant to be considered for medicinal purposes. However, fruit phytochemical compounds are now more studied than compounds from other M. alba plant parts. Quercetin, pyrrole alkaloids, cyanide, epigallocatechin, epigallocatechin gallate, gallocatechin, gallocatechin gallate, isorhamnetin glucuronide, isorhamnetin hexoside, isorhamnetin hexosylhexoside, kaempferol, glucuronide, kaempferol hexoside, kaempferol hexosylhexoside, kaempferol rhamnosylhexoside, morin, odisolane, and naringin, and phenolic, flavonoid, quinic acid, and anthocyanin compounds have been found in M. alba fruits [46,77,78,93,101]. The extract has been used to treat diabetes, arteriosclerosis, hypertension, blood circulation problems, coughing, and asthma in humans, and has recently been scientifically proven to be effective in animal experiments [2,104,117,118]. M. alba water extracts (MWEs) have been found to help reduce body weight, serum, and liver lipids in high-fat diet (HFD)-induced obesity [119]. Arfan et al. demonstrated that acetone or methanol extract of M. alba have high antioxidant potential, as determined by ABTS, DPPH, and reducing power assays [120]. The fruits of M. alba, unlike other parts of the plant, have been more frequently studied in relation to their immunological effects on the human body.

3. M. alba L. Extraction Solvents and Their Pharmacological Potential

The solvents most commonly used for phytochemical extraction from plant tissue are water, ethanol, methanol, acetone, and ether or a mixture of these [121,122,123]. Table 2 summarizes the pharmacological efficacy of phytochemicals detected in various M. alba extract types. It illustrates how efficacy changes depending on solvent type.
Water is used to extract high polar components, such as carbohydrates, amino acids, and glycosides. Ether and acetone are used to separate low polar or aromatic compounds [121,124]. Water extraction is the safest, least expensive, and most environmentally friendly method. Polysaccharides, proteins, polyphenols, and glycosides, which are soluble in water, are separated during water extraction. According to research by Wang et al., ethanol extracts of M. alba contained a higher number of phenols and flavonoids than water extracts [26,107]. However, according to Milena et al., M. alba water extraction yields more phenolic acids and flavonoids than hydromethanolic extraction methods [125]. Although ethanol or methanol may be selected as a suitable solvent for the separation of active ingredients, such as phenol, plant tissues contain numerous biologically active compounds that require alternate extraction solvents depending on the plant species. In addition, extraction yield is the most important factor in selecting a solvent and is affected by extraction time, temperature, and sample composition, among other factors. Hot water is also used to extract phenolic compounds and polysaccharides at higher concentrations than other solvents, such as ethanol [126]. Research by Peng et al. showed that M. alba water extracts (MWEs) contain polyphenols, including gallic acid, chlorogenic acid, rutin, and anthocyanins [119].
Certain endotoxins (LPS) are well-known immunomodulators and often become contaminants in phytochemical extracts. Therefore, several approaches, such as using ultrapure water and filters, are recommended to prevent possible LPS contamination [127,128].
Most plant or fungi extracts that were found to induce immunological activity in the human body were extracted using hot water. In one study on immune activity, the phytochemicals in dried and crushed M. alba tissues were extracted using hot water, producing a yield of approximately 25–30% [25,129]. In 2009, Yang’s study was the first to report the immunological activity of phytochemicals extracted using water. This study demonstrated increased phagocytosis, co-stimulatory molecules, cytokine secretion, and MAPK-inase activity in macrophage [130]. Hot water extracts of M. alba fruits play an important role in immunity-stimulating activities in the human body.

4. Immune Stimulation by Maturity of M. alba Fruits

The fruits of M. alba have been found to induce the highest immune-stimulating effect compared to other plant parts. According to research by Zhang [9], the physical properties of M. alba fruits change dramatically during maturation. M. alba fruits are classified into four stages: (1) immature, (2) semi-mature, (3) mature, and (4) fully mature. In studies by Liu, mature fruits were found to contain higher total polyphenols and anthocyanins, and also induce higher total antioxidant activity in the model organisms, than immature fruits [131]. Immature fruits have a low pH (which increases storage length). In terms of nutrition, immature fruits have high protein, crude fat, crude fiber, and mineral contents, as well as high concentrations of GABA, amino acids, tocopherols, phenolic acid, and flavonols (which play a functional role in fruit development) (Figure 2).
However, no systematic studies have been conducted on how immunity stimulation activity effects on model organisms may change in relation to the extract source in terms of fruit maturity. We conducted an immunological study comparing the effects of water extracts derived from immature versus fully mature fruits. Phytochemicals in fully mature fruits have been extracted using hot water in previous studies [25] and their effects on immune activity in model organisms compared according to fruit maturity (Figure 3). The results for immature fruits were more than three times higher than that of fully mature fruits at the same concentration (100 μg/mL) (without cytotoxicity). Additionally, ginseng, a popular immune stimulator sold in Korea and throughout the world, is produced from hot water crude extracts [132,133] and is sold in the form of beverages, tablets, and capsules [134]. Figure 3 shows our present results, which we measured using the same experimental method as the previous study [25] for comparison with the macrophage activity of Korean 6-year root red ginseng extracts and M. alba fruits in the present study. Korean 6-year root red ginseng extracts were purchased from a manufacturer (Junggwanjang KCG, Daejeon, Korea).
The results were equivalent to those reported in the P. ginseng experiment and were effective at the same concentrations as those in products that are marketed for immune-enhancing abilities. These results suggest that M. alba may be a suitable substitute for ginseng, which currently monopolizes the immunostimulant market.

5. Immune Stimulation by M. alba Bioactive Phytochemicals

In M. alba, polysaccharides and alkaloids are thought to be the immune-stimulating components and they are more prevalent in the fruits than in any other parts of the plant. Numerous studies have shown that polysaccharides play important roles in various physiological and pathological activities (Figure 4) [135,136].

5.1. Polysaccharides

The vast majority of studies that focused on the isolation of M. alba polysaccharides and their bioactivities have mainly concentrated on its leaves and fruits, which are medicinal and edible. Among the various parts of M. alba, the fruit contains the most polysaccharides. According to a review by Yuan et al., various polysaccharides (FMAP, MFP, MFP-1, MP, etc.) have been isolated and identified in the fruits of M. alba through various extraction and purification processes [2]. The composition of polysaccharides varies according to how they are classified. For example, the polysaccharides isolated in a study by Lee et al. [137] were 1639 kDa in mass and were reported as follows: mannose (1.60 mol%), rhamnose (18.40 mol%), glucose (3.10 mol%), galactose (37.60 mol%), xylose (1.70 mol%), fucose (1.30 mol%), and arabinose (36.30 mol%). However, in a study by Chen, the polysaccharides were measured at 13.6 kDa and were reported as follows: rhamnose (25.98 mol%), glucose (13.06 mol%), galactose (23.10 mol%), galacturonic acid (16.35 mol%), and arabinose (21.51 mol%). Therefore, polysaccharides that have different structures may be separated differently and, thus, vary in their efficacy [43,137,138]. For example, Lee et al. [137] observed that a water-soluble polysaccharide (JS-MP-1) induced immunological activity in Raw264.7 cells [139,140]. Similarly, the macrophage immunomodulatory activity of M. alba-derived polysaccharides appears to correlate positively with the average molecular weight of these polysaccharides, with the higher molecular weight fractions being the most active [26,139]. Chen et al. reported mulberry leaf polysaccharide (MLP) as a potential mucosal vaccine adjuvant candidate against ND in chickens [30].
Immune stimulation in the defense against diseases in humans is currently receiving much attention. In particular, there is a growing interest in mushroom polysaccharides as well as various fruit polysaccharides (FPs), and studies are underway to identify these polysaccharides and their biological activities in model organisms. Shin et al. [7] reported that an M. alba polysaccharide can be used as an adjuvant in dendritic cell-based cancer immunotherapy, as it induces phenotypic maturation of dendritic cells [7].

5.2. Alkaloids

Kim et al. [27] separated several alkaloids from M. alba using spectroscopic data interpretation. Among them, morrole A (Figure 5), first isolated from M. alba fruit, has been reported to cause an increase in nitric oxide, TNF-α, and IL-12 production, as well as phagocytosis, through increased macrophage activity in RAW264.7 cells. However, with plant extracts, it is difficult to separate a single component in order to understand its efficacy in model organisms because various components have shown complex efficacies.

6. The Mechanism of Action of M. alba Fruit Extract on the Immune System

6.1. The Immune Stimulation

M. alba fruit water extracts primarily induce the production of inflammatory substances, in model organisms, via TLR4 in immune cells [25,129]. Toll-like receptors (TLRs) are a group of PRRs found in immune cells (macrophage cells, dendritic cells (DCs), natural killer (NK) cells, T cells, B cells, epithelial cells, and endothelial cells). Muscle cells and adipocytes also play important roles in pathogen recognition and inducing an immune response. TLR stimulation mainly activates the innate immune response [141,142]. Unregulated TLR activity increases the risk of developing chronic inflammatory and autoimmune diseases. Currently, various natural compounds and their derivatives have been found to act as agonists or antagonists to TLR family members and their downstream signal transduction molecules [143]. Research on the relationship between plant phytochemical extracts and TLR4 is increasing. The activation of TLR4 by M. alba activates MAPK-inase, which in turn increases NF-κB-induced expression of pro-inflammatory factors, such as TNF-α, IL-1β, and IL-6. According to a recent study by Chang et al. [25], the effects of M. alba extracts on macrophage NO and TNF-α production were also inhibited by anti-TLR4 antibodies. Moreover, M. alba extracts failed to induce production of NO and TNF-α in peritoneal macrophages obtained from C3H/HeJ mice, which have a point mutation in the TLR4 gene. This suggests that the TLR4 molecule is involved in NO and TNF-α production-mediated macrophage activation.

6.2. Host Defense

Water extracts of M. alba fruits have not been found to be cytotoxic to colon, bladder, breast, or liver cancer cells (in vitro). Chang et al. explored the synergistic antitumor effects of MFE and a drug called 5-fluorouracil in a CT26 cell xenograft model. Leukocyte counts, spleen weight, NK cells, and CTL activity in the tumor xenograft mice significantly increased in the MFE/drug group [25]. In addition, MFE was not found to have any antibacterial effects on S. typhimurium or E. coli. However, an in vivo study showed that oral administration of MFE for five days significantly increased survival in salmonella-infected mice [129]. The antitumor and antibacterial activity of MFE is thought to be a result of immune-stimulatory effects. Approximately 15% of all circulating lymphocytes are NK cells that can lyse cancer cells in vitro without prior immunity sensitization. Their main function is early host defense against both allogenic and autologous cells following infection by a virus, bacteria, or parasites or the growth of tumor cells [144,145].

6.3. Inflammatory Response

The macrophage is one of the most important innate immune cells in the body. It is present in various forms in all tissues. In addition, the action of these macrophages is known to be an important component of major immune responses, such as adaptive immunity, wound healing, and the inflammatory response [146,147,148]. The morphologies and functions of macrophages vary widely. They circulate through the blood and reside in tissues. They are known as microglial cells in the brain or Kupffer cells in the liver [149]. The activities of both types of macrophages are regulated by interaction with adjacent cells. In turn, activated macrophages affect the adjacent environment [150]. Therefore, macrophages perform important functions as part of homeostatic regulation of the body. It is suspected that abnormalities in the function of these macrophages may cause various diseases. Therefore, studies are currently being conducted in order to better understand macrophage activity. It is also important to identify drugs that can regulate macrophages. Various crude extracts of M. alba have been observed to exhibit favorable pharmacological effects due to their ability to modulate macrophage function. Kwon et al. found that a water extract of Mori folium (WEMF) significantly stimulated the production of NO and PGE2 as immune response parameters at non-cytotoxic concentrations. This was associated with increased expression of inducible NO synthase and COX-2. The release and expression of cytokines, such as TNF-α, interleukin (IL)-1β, IL-6, and IL-10, also significantly increased in response to treatment with WEMF [151]. Yang [130] and Chang et al. [25] observed that crude extracts of M. alba fruits induced potent macrophage immunomodulatory activity, as demonstrated by an induction of effector molecules, such as NO and cytokines. In addition, treatment with crude extracts of M. alba before PMA resulted in a significantly enhanced response, which is indicative of a priming effect. Many immunomodulatory compounds, including LPS, can prime phagocytes for enhanced ROS production, and it is generally thought that priming plays a key role in the host-defense process and may be essential for host survival against microbial pathogens [125,139,152].

7. Safety of M. alba Extracts

Toxicity of herbal products is often reported in consumers as a result of incorrect methods of administration, particularly overdoses [153]. Therefore, traditionally used herbal products must be consumed in non-toxic doses that are scientifically verified.
M. alba has long been consumed fresh (i.e., fruits) and in traditional medicine products. Its biological activities have been well studied. The intra-gastric administration of ethanolic extracts of M. alba fruits to rats at a maximum dose of 1000 mg/kg was not found to cause changes in their behavior, such as respiratory changes, weight loss, or death, within one week of continuous administration. This indicates that this extract type at the stated dose does not have any acute toxicity effects [118]. Juhas et al. orally administered Sprague-Dawley (SD) rats with MFE (M. alba fruit water extract) at the highest concentration of 2000 mg/kg, but no toxicity effects were observed [146,154]. In a study by Marx, no toxicity effects on male and female SD rats were observed following administration of an oral dose for 28 days at concentrations of 4000 mg/kg [155]. Peng et al. studied the toxicity of MWE (M. alba fruit water extract) on hamsters by examining the effects of oral administration (0.5–2%, w/w) of the extracts for 12 weeks [119]. Chang et al. investigated the sub-chronic oral toxicity and genotoxicity of MFE. SD rats were treated with MFE (M. alba fruit water extract) daily using oral doses at 40, 200, and 1000 mg/kg concentrations for 90 days [156]. The effect level at which no adverse effect was observed in this subacute toxicity study was considered to be 1000 mg/kg, the highest dose tested. In another study, no Salmonella typhimurium strains (TA98, TA102, and TA1535) treated with MFE exhibited genotoxicity. The effects of ingesting M. alba fruits were examined in health risk tests on adult humans and the results showed that there were no significant carcinogenic risks associated with consumption of M. alba fruits. Therefore, the consumption of M. alba fruits is considered to be safe.

8. Conclusions

M. alba L. products are rich in nutrients and bioactive compounds, and have a variety of pharmacological properties, which may help prevent or treat chronic diseases. M. alba L. contains more phenolic compounds than other berries, and more riboflavin, niacin, total phenols, and alkaloid than other mulberry species. In order to enhance the immunological activity of M. alba L., it is effective to extract immature fruits with water. Polysaccharides and pyrrol alkaloids are known for their macrophage-activating effects in M. alba L. In the crude extract extracted with water, it is considered that unknown components act in combination on immune enhancement. Therefore, there is a need for further research on unidentified compounds in M. alba fruits and their biological effects.It is also important to develop consistent phytochemical profiles for consumption and clinical trials. To date, there is little information on the metabolomics of phytochemicals, such as alkaloids and polysaccharides. Therefore, it is important to investigate the metabolites formed in the body and how they exert their immunopotentiating effects.

Author Contributions

B.-Y.C. and B.-S.K. carried out the literature search and manuscript preparation. B.-Y.C. and S.-Y.K. drafted the manuscript and editing. 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

The data generated during this study are included in this article and are available on request from the corresponding author.

Acknowledgments

This work was supported by Wonkwang University in 2020.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Farshid, M.A.; Fazeli, M.; Shomali, T.; Nazifi, S.; Namazi, F. Protective effect of black mulberry (Morus nigra L.) fruit hydroalcoholic extract against testosterone-induced benign prostatic hyperplasia in rats. Rev. Int. Androl. 2021, 19, 53–61. [Google Scholar] [CrossRef]
  2. 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]
  3. Dhanyalakshmi, K.H.; Nataraja, K.N. Mulberry (Morus spp.) has the features to treat as a potential perennial model system. Plant Signal. Behav. 2018, 13, e1491267. [Google Scholar] [CrossRef]
  4. Mahboubi, M. Morus alba (mulberry), a natural potent compound in management of obesity. Pharmacol. Res. 2019, 146, 104341. [Google Scholar] [CrossRef] [PubMed]
  5. Na, H.-N.; Park, S.; Jeon, H.-J.; Kim, H.-B.; Nam, J.-H. Reduction of adenovirus 36-induced obesity and inflammation by mulberry extract. Microbiol. Immunol. 2014, 58, 303–306. [Google Scholar] [CrossRef] [PubMed]
  6. Song, W.; Wang, H.J.; Bucheli, P.; Zhang, P.F.; Wei, D.Z.; Lu, Y.H. Phytochemical profiles of different mulberry (Morus sp.) species from China. J. Agric. Food Chem. 2009, 57, 9133–9140. [Google Scholar] [CrossRef] [PubMed]
  7. Shin, B.R.; Kim, H.S.; Yun, M.J.; Lee, H.K.; Kim, Y.J.; Kim, S.Y.; Lee, M.K.; Hong, J.T.; Kim, Y.; Han, S.B. Promoting effect of polysaccharide isolated from Mori fructus on dendritic cell maturation. Food Chem. Toxicol. 2013, 51, 411–418. [Google Scholar] [CrossRef]
  8. Chen, W.; Li, Y.; Bao, T.; Gowd, V. Mulberry Fruit Extract Affords Protection against Ethyl Carbamate-Induced Cyto-toxicity and Oxidative Stress. Oxid. Med. Cell. Longev. 2017, 2017, 1594963. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, H.; Ma, Z.F.; Luo, X.; Li, X. Effects of Mulberry Fruit (Morus alba L.) Consumption on Health Outcomes: A Mini-Review. Antioxidants 2018, 7, 69. [Google Scholar] [CrossRef] [Green Version]
  10. Imran, M.; Khan, H.; Shah, M.; Khan, R.; Khan, F. Chemical composition and antioxidant activity of certain Morus species. J. Zhejiang Univ. Sci. B 2010, 11, 973–980. [Google Scholar] [CrossRef] [PubMed]
  11. Mahmood, T.; Anwar, F.; Abbas, M.; Saari, N. Effect of Maturity on Phenolics (Phenolic Acids and Flavonoids) Profile of Strawberry Cultivars and Mulberry Species from Pakistan. Int. J. Mol. Sci. 2012, 13, 4591–4607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Jiang, Y.; Nie, W.-J. Chemical properties in fruits of mulberry species from the Xinjiang province of China. Food Chem. 2014, 174, 460–466. [Google Scholar] [CrossRef] [PubMed]
  13. Jun, H. Principles and Practice of Eastern Medicine; United Nations Educational, Scientific and Cultural Organization: Paris, France, 2009. [Google Scholar]
  14. Yoon, S.; Kim, H. Donguibogam; Donguibogam Publishing Company: Seoul, Korea, 2006; pp. 297–2189. [Google Scholar]
  15. Kwon, O.-M.; Cha, W.-S.; Park, S.-Y.; Oh, J.-H.; Ahn, S.-W. The appropriation of donguibogam and bencao gangmu and the shaping of distinctive Korean medicine in the late joseon dynasty. Korean J. Orient. Med. 2011, 17, 17–24. [Google Scholar]
  16. Jan, B.; Parveen, R.; Zahiruddin, S.; Khan, M.U.; Mohapatra, S.; Ahmad, S. Nutritional constituents of mulberry and their potential applications in food and pharmaceuticals: A review. Saudi J. Biol. Sci. 2021, 28, 3909–3921. [Google Scholar] [CrossRef] [PubMed]
  17. Natić, M.M.; Dabić, D.Č.; Papetti, A.; Fotirić Akšić, M.M.; Ognjanov, V.; Ljubojević, M.; Tešić, Ž.L. Analysis and characterisation of phytochemicals in mulberry (Morus alba L.) fruits grown in Vojvodina, North Serbia. Food Chem. 2015, 171, 128–136. [Google Scholar] [CrossRef] [PubMed]
  18. Huang, H.-P.; Ou, T.-T.; Wang, C.-J. Mulberry (Sang Shèn Zǐ) and Its Bioactive Compounds, the Chemoprevention Effects and Molecular Mechanisms In Vitro and In Vivo. J. Tradit. Complement. Med. 2013, 3, 7–15. [Google Scholar] [CrossRef] [Green Version]
  19. Gryn-Rynko, A.; Bazylak, G.; Olszewska-Słonina, D. New potential phytotherapeutics obtained from white mulberry (Morus alba L.) leaves. Biomed. Pharmacother. 2016, 84, 628–636. [Google Scholar] [CrossRef]
  20. Li, Y.; Bao, T.; Chen, W. Comparison of the protective effect of black and white mulberry against ethyl carbamate-induced cytotoxicity and oxidative damage. Food Chem. 2018, 243, 65–73. [Google Scholar] [CrossRef]
  21. Wu, J.N. Chinese Materia Medica; Oxford University Press: Oxford, UK, 2005. [Google Scholar]
  22. Rodrigues, E.L.; Marcelino, G.; Silva, G.T.; Figueiredo, P.S.; Garcez, W.S.; Corsino, J.; Guimarães, R.D.C.A.; Freitas, K.D.C. Nutraceutical and Medicinal Potential of the Morus Species in Metabolic Dysfunctions. Int. J. Mol. Sci. 2019, 20, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hu, S.; Li, S.-W.; Yan, Q.; Hu, X.-P.; Li, L.-Y.; Zhou, H.; Pan, L.-X.; Li, J.; Shen, C.-P.; Xu, T. Natural products, extracts and formulations comprehensive therapy for the improvement of motor function in alcoholic liver disease. Pharmacol. Res. 2019, 150, 104501. [Google Scholar] [CrossRef]
  24. Abakushina, E.V.; Gelm, Y.V.; Pasova, I.A.; Bazhin, A.V. Immunotherapeutic Approaches for the Treatment of Colorectal Cancer. Biochemistry 2019, 84, 720–728. [Google Scholar] [CrossRef]
  25. Chang, B.Y.; Kim, S.B.; Lee, M.K.; Park, H.; Kim, S.Y. Improved Chemotherapeutic Activity by Morus alba Fruits through Immune Response of Toll-Like Receptor 4. Int. J. Mol. Sci. 2015, 16, 24139–24158. [Google Scholar] [CrossRef] [Green Version]
  26. Wang, D.; Li, H.; Li, B.; Ma, R.; Zhang, N.; Zhang, X.; Jiao, L.; Wu, W. Systematic fractionation and immunoenhancement of water-soluble polysaccharides isolated from fruit of Morus alba L. Int. J. Biol. Macromol. 2018, 116, 1056–1063. [Google Scholar] [CrossRef]
  27. Kim, S.B.; Chang, B.Y.; Jo, Y.H.; Lee, S.H.; Han, S.-B.; Hwang, B.Y.; Kim, S.Y.; Lee, M.K. Macrophage activating activity of pyrrole alkaloids from Morus alba fruits. J. Ethnopharmacol. 2012, 145, 393–396. [Google Scholar] [CrossRef]
  28. Chen, X.; Sheng, Z.; Qiu, S.; Yang, H.; Jia, J.; Wang, J.; Jiang, C. Purification, characterization and in vitro and in vivo immune enhancement of polysaccharides from mulberry leaves. PLoS ONE 2019, 14, e0208611. [Google Scholar] [CrossRef]
  29. Kwon, D.H.; Cheon, J.M.; Choi, E.-O.; Jeong, J.W.; Lee, K.W.; Kim, K.Y.; Kim, S.G.; Kim, S.; Hong, S.H.; Park, C.; et al. The Immunomodulatory Activity of Mori folium, the Leaf of Morus alba L. in RAW 264.7 Macrophages In Vitro. J. Cancer Prev. 2016, 21, 144–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Chen, X.; Yang, H.; Jia, J.; Chen, Y.; Wang, J.; Chen, H.; Jiang, C. Mulberry leaf polysaccharide supplementation contributes to enhancing the respiratory mucosal barrier immune response in Newcastle disease virus—Vaccinated chicks. Poult. Sci. 2020, 100, 592–602. [Google Scholar] [CrossRef] [PubMed]
  31. Kim, H.M.; Han, S.B.; Lee, K.H.; Lee, C.W.; Kim, C.Y.; Lee, E.J.; Huh, H. Immunomodulating activity of a polysaccharide isolated from mori cortex radicis. Arch. Pharmacal Res. 2000, 23, 240–242. [Google Scholar] [CrossRef]
  32. 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] [PubMed]
  33. Oliveira, A.M.; Nascimento, M.F.; Ferreira, M.R.; Moura, D.F.; Souza, T.G.; Silva, G.C.; Ramos, E.H.; Paiva, P.M.; Medeiros, P.L.; Silva, T.G.; et al. Evaluation of acute toxicity, genotoxicity and inhibitory effect on acute inflammation of an ethanol extract of Morus alba L. (Moraceae) in mice. J. Ethnopharmacol. 2016, 194, 162–168. [Google Scholar] [CrossRef] [PubMed]
  34. Leyva-Jiménez, F.J.; Ruiz-Malagón, A.J.; Molina-Tijeras, J.A.; Diez-Echave, P.; Vezza, T.; Hidalgo-García, L.; Lozano-Sánchez, J.; Arráez-Román, D.; Cenis, J.L.; Lozano-Pérez, A.A.; et al. Comparative Study of the Antioxidant and Anti-Inflammatory Effects of Leaf Extracts from Four Different Morus alba Genotypes in High Fat Diet-Induced Obesity in Mice. Antioxidants 2020, 9, 733. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, M.R.; Kim, J.E.; Park, J.J.; Choi, J.Y.; Song, B.R.; Choi, Y.W.; Kim, D.S.; Kim, K.M.; Song, H.K.; Hwang, D.Y. Protective role of fermented mulberry leave extract in LPS-induced inflammation and autophagy of RAW264.7 macrophage cells. Mol. Med. Rep. 2020, 22, 4685–4695. [Google Scholar] [CrossRef] [PubMed]
  36. Bharani, S.E.R.; Asad, M.; Dhamanigi, S.S.; Chandrakala, G.K. Immunomodulatory activity of methanolic extract of Morus alba Linn. (mulberry) leaves. Pak. J. Pharm. Sci. 2010, 23, 63–68. [Google Scholar]
  37. Soonthornsit, N.; Pitaksutheepong, C.; Hemstapat, W.; Utaisincharoen, P.; Pitaksuteepong, T. In Vitro Anti-Inflammatory Activity of Morus alba L. Stem Extract in LPS-Stimulated RAW 264.7 Cells. Evid. Based Complement. Altern. Med. 2017, 2017, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Chauhan, S.; Devi, U.; Kumar, V.R.; Kumar, V.; Anwar, F.; Kaithwas, G. Dual inhibition of arachidonic acid pathway by mulberry leaf extract. Inflammopharmacology 2015, 23, 65–70. [Google Scholar] [CrossRef]
  39. Yiemwattana, I.; Kaomongkolgit, R.; Wirojchanasak, S.; Chaisomboon, N. Morus alba Stem Extract Suppresses Matrix Metalloproteinases (MMP)-1, MMP-9, and Tissue Inhibitors of Metalloproteinase (TIMP)-1 Expression via Inhibition of Ikappa Balpha Degradation Induced by Porphyromonas gingivalis LPS Signal in THP-1 Cells. Eur. J. Dent. 2019, 13, 229–234. [Google Scholar] [CrossRef] [Green Version]
  40. Chen, Y.C.; Tien, Y.J.; Chen, C.H.; Beltran, F.N.; Amor, E.C.; Wang, R.J.; Wu, D.J.; Mettling, C.; Lin, Y.L.; Yang, W.C. Morus alba and active compound oxyresveratrol exert anti-inflammatory activity via inhibition of leukocyte migration involving MEK/ERK signaling. BMC Complement. Altern. Med. 2013, 13, 45. [Google Scholar] [CrossRef] [Green Version]
  41. Wongwat, T.; Srihaphon, K.; Pitaksutheepong, C.; Boonyo, W.; Pitaksuteepong, T. Suppression of inflammatory mediators and matrix metalloproteinase (MMP)-13 by Morus alba stem extract and oxyresveratrol in RAW 264.7 cells and C28/I2 human chondrocytes. J.Tradit. Complementary Med. 2020, 10, 132–140. [Google Scholar] [CrossRef]
  42. Kavitha, Y.; Geetha, A. Anti-inflammatory and preventive activity of white mulberry root bark extract in an experimental model of pancreatitis. J. Tradit. Complement. Med. 2018, 8, 497–505. [Google Scholar] [CrossRef]
  43. Chen, C.; Zhang, B.; Fu, X.; Liu, R.H. A novel polysaccharide isolated from mulberry fruits (Murus alba L.) and its selenide derivative: Structural characterization and biological activities. Food Funct. 2016, 7, 2886–2897. [Google Scholar] [CrossRef]
  44. Zhang, J.; Chen, C.; Fu, X. Fructus mori L. polysaccharide-iron chelates formed by self-embedding with iron(iii) as the core exhibit good antioxidant activity. Food Funct. 2019, 10, 3150–3160. [Google Scholar] [CrossRef]
  45. Gui, Z.; Raman, S.T.; Ganeshan, A.K.P.G.; Chen, C.; Jin, C.; Li, S.-H.; Chen, H.-J. In vitro and In vivo antioxidant activity of flavonoid extracted from mulberry fruit (Morus alba L.). Pharmacogn. Mag. 2016, 12, 128–133. [Google Scholar] [CrossRef] [PubMed]
  46. Li, M.; Li, T.; Hu, X.; Ren, G.; Zhang, H.; Wang, Z.; Teng, Z.; Wu, R.; Wu, J. Structural, rheological properties and antioxidant activities of polysaccharides from mulberry fruits (Murus alba L.) based on different extraction techniques with superfine grinding pretreatment. Int. J. Biol. Macromol. 2021, 183, 1774–1783. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, Y.; Xiang, L.; Wang, C.; Tang, C.; He, X. Antidiabetic and Antioxidant Effects and Phytochemicals of Mulberry Fruit (Morus alba L.) Polyphenol Enhanced Extract. PLoS ONE 2013, 8, e71144. [Google Scholar] [CrossRef]
  48. Liao, B.-Y.; Zhu, D.-Y.; Thakur, K.; Li, L.; Zhang, J.-G.; Wei, Z.-J. Thermal and Antioxidant Properties of Polysaccharides Sequentially Extracted from Mulberry Leaves (Morus alba L.). Molecules 2017, 22, 2271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ann, J.-Y.; Eo, H.; Lim, Y. Mulberry leaves (Morus alba L.) ameliorate obesity-induced hepatic lipogenesis, fibrosis, and oxidative stress in high-fat diet-fed mice. Genes Nutr. 2015, 10, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Cui, H.; Lu, T.; Wang, M.; Zou, X.; Zhang, Y.; Yang, X.; Dong, Y.; Zhou, H. Flavonoids from Morus alba L. Leaves: Opti-mization of Extraction by Response Surface Methodology and Comprehensive Evaluation of Their Antioxidant, Antimi-crobial, and Inhibition of alpha-Amylase Activities through Analytical Hierarchy Process. Molecules 2019, 24, 2398. [Google Scholar] [CrossRef] [Green Version]
  51. Khan, M.A.; Rahman, A.A.; Islam, S.; Khandokhar, P.; Parvin, S.; Islam, B.; Hossain, M.; Rashid, M.; Sadik, G.; Nasrin, S.; et al. A comparative study on the antioxidant activity of methanolic extracts from different parts of Morus alba L. (Moraceae). BMC Res. Notes 2013, 6, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kim, D.-S.; Ji, H.D.; Rhee, M.H.; Sung, Y.-Y.; Yang, W.-K.; Kim, S.H.; Kim, H.-K. Antiplatelet Activity ofMorus albaLeaves Extract, Mediated via Inhibiting Granule Secretion and Blocking the Phosphorylation of Extracellular-Signal-Regulated Kinase and Akt. Evid. Based Complement. Altern. Med. 2014, 2014, 1–11. [Google Scholar] [CrossRef]
  53. Park, S.W.; Shin, K.C.; Yoou, S.-K.; Park, H.J.; Eun, S.H.; Bae, Y.M.; Lee, H.M.; Chae, H.-J.; Chae, S.-W.; Choi, B.H. Effects of an ethanolic extract of mulberry fruit on blood pressure and vascular remodeling in spontaneous hypertensive rats. Clin. Exp. Hypertens. 2018, 41, 280–286. [Google Scholar] [CrossRef] [PubMed]
  54. Xia, M.; Qian, L.; Zhou, X.; Gao, Q.; Bruce, I.; Xia, Q. Endothelium-independent relaxation and contraction of rat aorta induced by ethyl acetate extract from leaves of Morus alba (L.). J. Ethnopharmacol. 2008, 120, 442–446. [Google Scholar] [CrossRef]
  55. Arraki, K.; Totoson, P.; Attia, R.; Zedet, A.; Pudlo, M.; Messaoud, C.; Demougeot, C.; Girard, C. Arginase inhibitory properties of flavonoid compounds from the leaves of Mulberry (Morus alba, Moraceae). J. Pharm. Pharmacol. 2020, 72, 1269–1277. [Google Scholar] [CrossRef]
  56. Zheng, X.-K.; Cao, Y.-G.; Ke, Y.-Y.; Zhang, Y.-L.; Li, F.; Gong, J.-H.; Zhao, X.; Kuang, H.-X.; Feng, W.-S. Phenolic constituents from the root bark of Morus alba L. and their cardioprotective activity in vitro. Phytochemistry 2017, 135, 128–134. [Google Scholar] [CrossRef] [PubMed]
  57. Panth, N.; Paudel, K.R. Vascular Protection by Ethanol Extract of Morus alba Root Bark: Endothelium-Dependent Relaxation of Rat Aorta and Decrease of Smooth Muscle Cell Migration and Proliferation. Evid. Based Complement. Altern. Med. 2018, 2018, 7905763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Yamatake, Y.; Shibata, M.; Nagai, M. Pharmacological studies on root bark of mulberry tree (Morus alba L.). Jpn. J. Pharmacol. 1976, 26, 461–469. [Google Scholar] [CrossRef] [Green Version]
  59. Lee, J.-J.; Yang, H.; Yoo, Y.-M.; Hong, S.S.; Lee, N.; Lee, H.-J.; Lee, H.-J.; Myung, C.-S.; Choi, K.-C.; Jeung, E.-B. Morusinol extracted from Morus alba inhibits arterial thrombosis and modulates platelet activation for the treatment of cardiovascular disease. J. Atheroscler. Thromb. 2012, 19, 516–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Sarikaphuti, A.; Nararatwanchai, T.; Hashiguchi, T.; Ito, T.; Thaworanunta, S.; Kikuchi, K.; Oyama, Y.; Maruyama, I.; Tancharoen, S. Preventive effects of Morus alba L. anthocyanins on diabetes in Zucker diabetic fatty rats. Exp. Ther. Med. 2013, 6, 689–695. [Google Scholar] [CrossRef] [Green Version]
  61. Choi, K.H.; Lee, H.A.; Park, M.H.; Han, J.-S. Mulberry (Morus alba L.) Fruit Extract Containing Anthocyanins Improves Glycemic Control and Insulin Sensitivity via Activation of AMP-Activated Protein Kinase in Diabetic C57BL/Ksj-db/db Mice. J. Med. Food 2016, 19, 737–745. [Google Scholar] [CrossRef]
  62. Hunyadi, A.; Veres, K.; Danko, B.; Kele, Z.; Wéber, E.; Hetenyi, A.; Zupko, I.; Hsieh, T.-J. In Vitro Anti-diabetic Activity and Chemical Characterization of an Apolar Fraction of Morus alba Leaf Water Extract. Phytother. Res. 2012, 27, 847–851. [Google Scholar] [CrossRef]
  63. Assiri, A.M.; Elbeeh, M.; Amin, A.H.; Ramadan, M.F. Ameliorative impact of Morus alba leaves’ aqueous extract against embryonic ophthalmic tissue malformation in streptozotocin-induced diabetic rats. Biomed. Pharmacother. 2017, 95, 1072–1081. [Google Scholar] [CrossRef] [PubMed]
  64. Park, J.M.; Bong, H.Y.; Jeong, H.I.; Kim, Y.K.; Kim, J.Y.; Kwon, O. Postprandial hypoglycemic effect of mulberry leaf in Goto-Kakizaki rats and counterpart control Wistar rats. Nutr. Res. Pr. 2009, 3, 272–278. [Google Scholar] [CrossRef]
  65. Hunyadi, A.; Martins, A.; Hsieh, T.J.; Seres, A.; Zupkó, I. Chlorogenic acid and rutin play a major role in the in vivo an-ti-diabetic activity of Morus alba leaf extract on type II diabetic rats. PLoS ONE 2012, 7, e50619. [Google Scholar] [CrossRef] [Green Version]
  66. Mahmoud, A.M.; El-Twab, S.M.A.; Abdel-Reheim, E.S. Consumption of polyphenol-rich Morus alba leaves extract attenuates early diabetic retinopathy: The underlying mechanism. Eur. J. Nutr. 2016, 56, 1671–1684. [Google Scholar] [CrossRef]
  67. Król, E.; Jeszka-Skowron, M.; Krejpcio, Z.; Flaczyk, E.; Wójciak, R.W. The Effects of Supplementary Mulberry Leaf (Morus alba) Extracts on the Trace Element Status (Fe, Zn and Cu) in Relation to Diabetes Management and Antioxidant Indices in Diabetic Rats. Biol. Trace Element Res. 2016, 174, 158–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Hunyadi, A.; Liktor-Busa, E.; Márki, A.; Martins, A.; Jedlinszki, N.; Hsieh, T.-J.; Báthori, M.; Hohmann, J.; Zupkó, I. Metabolic Effects of Mulberry Leaves: Exploring Potential Benefits in Type 2 Diabetes and Hyperuricemia. Evid. Based Complement. Altern. Med. 2013, 2013, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Singab, A.N.B.; El-Beshbishy, H.A.; Yonekawa, M.; Nomura, T.; Fukai, T. Hypoglycemic effect of Egyptian Morus alba root bark extract: Effect on diabetes and lipid peroxidation of streptozotocin-induced diabetic rats. J. Ethnopharmacol. 2005, 100, 333–338. [Google Scholar] [CrossRef]
  70. Kim, E.-S.; Park, S.-J.; Lee, E.-J.; Kim, B.-K.; Huh, H.; Lee, B.-J. Purification and characterization of Moran 20K from Morus alba. Arch. Pharm. Res. 1999, 22, 9–12. [Google Scholar] [CrossRef] [PubMed]
  71. Zhao, Y.; Kongstad, K.T.; Jager, A.K.; Nielsen, J.; Staerk, D. Quadruple high-resolution al-pha-glucosidase/alpha-amylase/PTP1B/radical scavenging profiling combined with high-performance liquid chromatography-high-resolution mass spectrometry-solid-phase extraction-nuclear magnetic resonance spectroscopy for identification of antidiabetic constituents in crude root bark of Morus alba L. J. Chromatogr. A 2018, 1556, 55–63. [Google Scholar]
  72. Salimi, F.; Moradi, M.; Tajik, H.; Molaei, R. Optimization and characterization of eco-friendly antimicrobial nanocellulose sheet prepared using carbon dots of white mulberry (Morus alba L.). J. Sci. Food Agric. 2020, 101, 3439–3447. [Google Scholar] [CrossRef]
  73. de Oliveira, A.M.; Mesquita Mda, S.; da Silva, G.C.; de Oliveira Lima, E.; de Medeiros, P.L.; Paiva, P.M.; de Souza, I.A.; Napoleao, T.H. Evaluation of Toxicity and Antimicrobial Activity of an Ethanolic Extract from Leaves of Morus alba L. (Moraceae). Evidence-based complementary and alternative medicine. eCAM 2015, 2015, 513978. [Google Scholar]
  74. Bauomy, A.A. The Potential Role of Morus alba Leaves Extract on the Brain of Mice Infected with Schistosoma Mansoni. CNS Neurol. Disord. Drug Targets 2014, 13, 1513–1519. [Google Scholar] [CrossRef]
  75. Sheikhlar, A.; Alimon, A.R.; Daud, H.; Saad, C.R.; Webster, C.D.; Meng, G.Y.; Ebrahimi, M. White Mulberry (Morus alba) Foliage Methanolic Extract Can Alleviate Aeromonas hydrophila Infection in African Catfish (Clarias gariepinus). Sci. World J. 2014, 2014, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Islam, B.; Khan, S.N.; Haque, I.; Alam, M.; Mushfiq, M.; Khan, A.U. Novel anti-adherence activity of mulberry leaves: Inhibition of Streptococcus mutans biofilm by 1-deoxynojirimycin isolated from Morus alba. J. Antimicrob. Chemother. 2008, 62, 751–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lee, D.; Yu, J.S.; Lee, S.R.; Hwang, G.S.; Kang, K.S.; Park, J.G.; Kim, H.Y.; Kim, K.H.; Yamabe, N. Beneficial Effects of Bioactive Compounds in Mulberry Fruits against Cisplatin-Induced Nephrotoxicity. Int. J. Mol. Sci. 2018, 19, 1117. [Google Scholar] [CrossRef] [Green Version]
  78. Lee, S.R.; Park, J.Y.; Hyun, K.K.; Lee, S.O.; Ryu, J.-Y.; Choi, S.-Z.; Kang, K.S.; Yamabe, N.; Kim, K.H. Odisolane, a Novel Oxolane Derivative, and Antiangiogenic Constituents from the Fruits of Mulberry (Morus alba L.). J. Agric. Food Chem. 2016, 64, 3804–3809. [Google Scholar] [CrossRef] [PubMed]
  79. Dabili, S.; Fallah, S.; Aein, M.; Vatannejad, A.; Panahi, G.; Fadaei, R.; Moradi, N.; Shojaii, A. Survey of the effect of doxorubicin and flavonoid extract of white Morus alba leaf on apoptosis induction in a-172 GBM cell line. Arch. Physiol. Biochem. 2019, 125, 136–141. [Google Scholar] [CrossRef]
  80. 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]
  81. Sithara, T.; Arun, K.B.; Syama, H.P.; Reshmitha, T.R.; Nisha, P. Morin Inhibits Proliferation of SW480 Colorectal Cancer Cells by Inducing Apoptosis Mediated by Reactive Oxygen Species Formation and Uncoupling of Warburg Effect. Front. Pharmacol. 2017, 8, 640. [Google Scholar] [CrossRef] [Green Version]
  82. Min, T.R.; Park, H.J.; Park, M.N.; Kim, B. The Root Bark of Morus alba L. Suppressed the Migration of Human Non-Small-Cell Lung Cancer Cells through Inhibition of Epithelial-Mesenchymal Transition Mediated by STAT3 and Src. Int. J. Mol. Sci. 2019, 20, 2244. [Google Scholar] [CrossRef] [Green Version]
  83. 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, 200. [Google Scholar] [CrossRef] [Green Version]
  84. Kikuchi, T.; Nihei, M.; Nagai, H.; Fukushi, H.; Tabata, K.; Suzuki, T.; Akihisa, T. Albanol A from the Root Bark of Morus alba L. Induces Apoptotic Cell Death in HL60 Human Leukemia Cell Line. Chem. Pharm. Bull. 2010, 58, 568–571. [Google Scholar] [CrossRef] [Green Version]
  85. Lim, S.-L.; Park, S.-Y.; Kang, S.; Park, D.; Kim, S.-H.; Um, J.-Y.; Jang, H.-J.; Lee, J.-H.; Jeong, C.-H.; Jang, J.-H.; et al. Morusin induces cell death through inactivating STAT3 signaling in prostate cancer cells. Am. J. Cancer Res. 2014, 5, 289–299. [Google Scholar]
  86. Hu, D.; Bao, T.; Lu, Y.; Su, H.; Ke, H.; Chen, W. Polysaccharide from Mulberry Fruit (Morus alba L.) Protects against Palmitic-Acid-Induced Hepatocyte Lipotoxicity by Activating the Nrf2/ARE Signaling Pathway. J. Agric. Food Chem. 2019, 68, 13016–13024. [Google Scholar] [CrossRef] [PubMed]
  87. Pel, P.; Chae, H.-S.; Nhoek, P.; Kim, Y.-M.; Chin, Y.-W. Chemical Constituents with Proprotein Convertase Subtilisin/Kexin Type 9 mRNA Expression Inhibitory Activity from Dried Immature Morus alba Fruits. J. Agric. Food Chem. 2017, 65, 5316–5321. [Google Scholar] [CrossRef] [PubMed]
  88. Kujawska, M.; Ewertowska, M.; Adamska, T.; Ignatowicz, E.; Flaczyk, E.; Przeor, M.; Kurpik, M.; Liebert, J.J. Protective Effect of Morus alba Leaf Extract on N-Nitrosodiethylamine-induced Hepatocarcinogenesis in Rats. In Vivo 2016, 30, 807–812. [Google Scholar] [CrossRef] [Green Version]
  89. Liang, H.-W.; Yang, T.-Y.; Teng, C.-S.; Lee, Y.-J.; Yu, M.-H.; Lee, H.-J.; Hsu, L.-S.; Wang, C.-J. Mulberry leaves extract ameliorates alcohol-induced liver damages through reduction of acetaldehyde toxicity and inhibition of apoptosis caused by oxidative stress signals. Int. J. Med. Sci. 2021, 18, 53–64. [Google Scholar] [CrossRef]
  90. Lee, M.R.; Kim, J.E.; Park, J.W.; Kang, M.J.; Choi, H.J.; Bae, S.J.; Choi, Y.W.; Kim, K.M.; Hong, J.T.; Hwang, D.Y. Fermented mulberry (Morus alba) leaves suppress high fat diet-induced hepatic steatosis through amelioration of the inflammatory response and autophagy pathway. BMC Complement. Med. Ther. 2020, 20, 283. [Google Scholar] [CrossRef] [PubMed]
  91. Hu, Y.; Xu, J.; Chen, Q.; Liu, M.; Wang, S.; Yu, H.; Zhang, Y.; Wang, T. Regulation effects of total flavonoids in Morus alba L. on hepatic cholesterol disorders in orotic acid induced NAFLD rats. BMC Complement. Med. Ther. 2020, 20, 1–12. [Google Scholar] [CrossRef] [PubMed]
  92. Tseng, T.H.; Lin, W.L.; Chang, C.K.; Lee, K.C.; Tung, S.Y.; Kuo, H.C. Protective Effects of Morus Root Extract (MRE) Against Lipopolysaccharide-Activated RAW264.7 Cells and CCl4-Induced Mouse Hepatic Damage. Cell. Physiol. Biochem. 2018, 51, 1376–1388. [Google Scholar] [CrossRef]
  93. Seo, K.H.; Lee, D.Y.; Jeong, R.H.; Lee, D.S.; Kim, Y.E.; Hong, E.K.; Kim, Y.C.; Baek, N.I. Neuroprotective effect of prenylated arylbenzofuran and flavonoids from Morus alba fruits on glutamate-induced oxidative injury in HT22 hippo-campal cells. J. Med. Food 2015, 18, 403–408. [Google Scholar] [CrossRef]
  94. Liu, D.; Du, D. Mulberry Fruit Extract Alleviates Cognitive Impairment by Promoting the Clearance of Amyloid-β and Inhibiting Neuroinflammation in Alzheimer’s Disease Mice. Neurochem. Res. 2020, 45, 2009–2019. [Google Scholar] [CrossRef] [PubMed]
  95. Shin, S.K.; Yoo, J.-M.; Li, F.Y.; Baek, S.Y.; Kim, M.R. Mulberry fruit improves memory in scopolamine-treated mice: Role of cholinergic function, antioxidant system, and TrkB/Akt signaling. Nutr. Neurosci. 2019, 2, 1–11. [Google Scholar] [CrossRef]
  96. Chen, X.-Y.; Zhang, T.; Wang, X.; Hamann, M.T.; Kang, J.; Yu, D.-Q.; Chen, R.-Y. A Chemical Investigation of the Leaves of Morus alba L. Molecules 2018, 23, 1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Yadav, A.V.; Nade, V.S. Anti-dopaminergic effect of the methanolic extract of Morus alba L. leaves. Indian J. Pharmacol. 2008, 40, 221–226. [Google Scholar] [PubMed] [Green Version]
  98. Kang, T.H.; Oh, H.R.; Jung, S.M.; Ryu, J.H.; Park, M.W.; Park, Y.K.; Kim, S.Y. Enhancement of Neuroprotection of Mulberry Leaves (Morus alba L.) Prepared by the Anaerobic Treatment against Ischemic Damage. Biol. Pharm. Bull. 2006, 29, 270–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Rebai, O.; Belkhir, M.; Boujelben, A.; Fattouch, S.; Amri, M. Morus alba leaf extract mediates neuroprotection against glyphosate-induced toxicity and biochemical alterations in the brain. Environ. Sci. Pollut. Res. 2017, 24, 9605–9613. [Google Scholar] [CrossRef] [Green Version]
  100. Kuk, E.B.; Jo, A.R.; Oh, S.I.; Sohn, H.S.; Seong, S.H.; Roy, A.; Choi, J.S.; Jung, H.A. Anti-Alzheimer’s disease activity of compounds from the root bark of Morus alba L. Arch. Pharmacal Res. 2017, 40, 338–349. [Google Scholar] [CrossRef]
  101. Kim, S.B.; Chang, B.Y.; Hwang, B.Y.; Kim, S.Y.; Lee, M.K. Pyrrole alkaloids from the fruits of Morus alba. Bioorg. Med. Chem. Lett. 2014, 24, 5656–5659. [Google Scholar] [CrossRef]
  102. Peng, C.-H.; Lin, H.-T.; Chung, D.-J.; Huang, C.-N.; Wang, C.-J. Mulberry Leaf Extracts prevent obesity-induced NAFLD with regulating adipocytokines, inflammation and oxidative stress. J. Food Drug Anal. 2018, 26, 778–787. [Google Scholar] [CrossRef] [PubMed]
  103. Park, K.; You, J.; Lee, H.; Baek, N.; Hwang, J. Kuwanon G: An antibacterial agent from the root bark of Morus alba against oral pathogens. J. Ethnopharmacol. 2003, 84, 181–185. [Google Scholar] [CrossRef]
  104. Jung, H.W.; Kang, S.Y.; Kang, J.S.; Kim, A.R.; Woo, E.-R.; Park, Y.-K. Effect of Kuwanon G Isolated from the Root Bark of Morus alba on Ovalbumin-induced Allergic Response in a Mouse Model of Asthma. Phytother. Res. 2014, 28, 1713–1719. [Google Scholar] [CrossRef]
  105. Yiemwattana, I.; Chaisomboon, N.; Jamdee, K. Antibacterial and Anti-inflammatory Potential of Morus alba Stem Extract. Open Dent. J. 2018, 12, 265–274. [Google Scholar] [CrossRef]
  106. Kuete, V.; Fozing, D.; Kapche, W.; Mbaveng, A.; Kuiate, J.; Ngadjui, B.; Abegaz, B. Antimicrobial activity of the methanolic extract and compounds from Morus mesozygia stem bark. J. Ethnopharmacol. 2009, 124, 551–555. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, W.; Zu, Y.; Fu, Y.; Efferth, T. In Vitro Antioxidant and Antimicrobial Activity of Extracts from Morus alba L. Leaves, Stems and Fruits. Am. J. Chin. Med. 2012, 40, 349–356. [Google Scholar] [CrossRef] [PubMed]
  108. Ali, A.; Ali, M. New triterpenoids from Morus alba L. stem bark. Nat. Prod. Res. 2013, 27, 524–531. [Google Scholar] [CrossRef] [PubMed]
  109. Khunakornvichaya, A.; Lekmeechai, S.; Pham, P.P.; Himakoun, W.; Pitaksuteepong, T.; Morales, N.P.; Hemstapat, W. Morus alba L. Stem Extract Attenuates Pain and Articular Cartilage Damage in the Anterior Cruciate Ligament Transection-Induced Rat Model of Osteoarthritis. Pharmacology 2016, 98, 209–216. [Google Scholar] [CrossRef]
  110. Riviere, C.; Krisa, S.; Pechamat, L.; Nassra, M.; Delaunay, J.C.; Marchal, A.; Badoc, A.; Waffo-Teguo, P.; Merillon, J.M. Polyphenols from the stems of Morus alba and their inhibitory activity against nitric oxide production by lipopolysaccha-ride-activated microglia. Fitoterapia 2014, 97, 253–260. [Google Scholar] [CrossRef] [PubMed]
  111. Hunyadi, A.; Herke, I.; Veres, K.; Erdei, A.; Simon, A.; Tóth, G. Volatile Glycosides from the Leaves of Morus alba with a Potential Contribution to the Complex Anti-diabetic Activity. Nat. Prod. Commun. 2014, 9. [Google Scholar] [CrossRef] [Green Version]
  112. Varghese, S.M.; Thomas, J. Polyphenolic constituents in mulberry leaf extract (M. latifolia L. cv. BC259) and its antidiabetic effect in streptozotocin induced diabetic rats. Pak. J. Pharm. Sci. 2019, 32, 69–74. [Google Scholar]
  113. Nakagawa, K.; Ogawa, K.; Higuchi, O.; Kimura, T.; Miyazawa, T.; Hori, M. Determination of iminosugars in mulberry leaves and silkworms using hydrophilic interaction chromatography–tandem mass spectrometry. Anal. Biochem. 2010, 404, 217–222. [Google Scholar] [CrossRef]
  114. Basnet, P.; Kadota, S.; Terashima, S.; Shimizu, M.; Namba, T. Two New 2-Arylbenzofuran Derivatives from Hypoglycemic Activity-Bearing Fractions of Morus insignis. Chem. Pharm. Bull. 1993, 41, 1238–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Lafont, R.; Dinan, L. Practical uses for ecdysteroids in mammals including humans: An update. J. Insect Sci. 2003, 3, 7. [Google Scholar] [CrossRef] [PubMed]
  116. Techa, S.; Chung, J.S. Ecdysteroids Regulate the Levels of Molt-Inhibiting Hormone (MIH) Expression in the Blue Crab, Callinectes sapidus. PLoS ONE 2015, 10, e0117278. [Google Scholar] [CrossRef]
  117. Eo, H.; Lim, Y. Combined Mulberry Leaf and Fruit Extract Improved Early Stage of Cutaneous Wound Healing in High-Fat Diet-Induced Obese Mice. J. Med. Food 2016, 19, 161–169. [Google Scholar] [CrossRef]
  118. Jiao, Y.; Wang, X.; Jiang, X.; Kong, F.; Wang, S.; Yan, C. Antidiabetic effects of Morus alba fruit polysaccharides on high-fat diet- and streptozotocin-induced type 2 diabetes in rats. J. Ethnopharmacol. 2017, 199, 119–127. [Google Scholar] [CrossRef] [PubMed]
  119. Peng, C.H.; Liu, L.K.; Chuang, C.M.; Chyau, C.C.; Huang, C.N.; Wang, C.J. Mulberry water extracts possess an anti-obesity effect and ability to inhibit hepatic lipogenesis and promote lipolysis. J. Agric. Food Chem. 2011, 59, 2663–2671. [Google Scholar] [CrossRef]
  120. 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]
  121. Cheigh, C.-I.; Yoo, S.-Y.; Ko, M.-J.; Chang, P.-S.; Chung, M.-S. Extraction characteristics of subcritical water depending on the number of hydroxyl group in flavonols. Food Chem. 2015, 168, 21–26. [Google Scholar] [CrossRef] [PubMed]
  122. Somekawa, L.; Maegawa, H.; Tsukada, S.; Nakamura, T. Establishment of Application Guidance for OTC non-Kampo Crude Drug Extract Products in Japan. J. Intercult. Ethnopharmacol. 2017, 6, 333–338. [Google Scholar] [CrossRef]
  123. Amakura, Y. Characterization of Phenolic Constituents from Ephedra Herb Extract. Yakugaku zasshi. J. Pharm. Soc. Jpn 2017, 137, 167–171. [Google Scholar] [CrossRef] [Green Version]
  124. Kumarasamy, B.; Manipal, S.; Duraisamy, P.; Ahmed, A.; Mohanaganesh, S.; Jeevika, C. Role of Aqueous Extract of Morinda Citrifolia (Indian Noni) Ripe Fruits in Inhibiting Dental Caries-Causing Streptococcus Mutans and Streptococcus Mitis. J. Dent. (Tehran, Iran) 2014, 11, 703–710. [Google Scholar]
  125. Polumackanycz, M.; Sledzinski, T.; Goyke, E.; Wesolowski, M.; Viapiana, A. A Comparative Study on the Phenolic Composition and Biological Activities of Morus alba L. Commercial Samples. Molecules 2019, 24, 3082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Xu, G.; Chen, J.; Liu, D.; Zhang, Y.; Jiang, P.; Ye, X. Minerals, Phenolic Compounds, and Antioxidant Capacity of Citrus Peel Extract by Hot Water. J. Food Sci. 2007, 73, C11–C18. [Google Scholar] [CrossRef]
  127. Fung, F.M.; Su, M.; Feng, H.-T.; Li, S.F.Y. Extraction, separation and characterization of endotoxins in water samples using solid phase extraction and capillary electrophoresis-laser induced fluorescence. Sci. Rep. 2017, 7, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Seidel, V. Initial and Bulk Extraction of Natural Products Isolation. Nat. Prod. Isol. 2012, 864, 27–41. [Google Scholar] [CrossRef]
  129. Chang, B.; Koo, B.; Lee, H.; Oh, J.S.; Kim, S. Activation of macrophage mediated host defense against Salmonella typhi-murium by Morus alba L. Food Nutr. Res. 2018, 62, 1289. [Google Scholar] [CrossRef] [Green Version]
  130. Yang, X.-Y.; Park, G.-S.; Lee, M.H.; Chang, I.A.; Kim, Y.C.; Kim, S.Y.; Lee, J.Y.; Yun, Y.G.; Park, H. Toll-like receptor 4-mediated immunoregulation by the aqueous extract of Mori Fructus. Phytother. Res. 2009, 23, 1713–1720. [Google Scholar] [CrossRef]
  131. Liu, X.; Xiao, G.; Chen, W.; Xu, Y.; Wu, J. Quantification and Purification of Mulberry Anthocyanins with Macroporous Resins. J. Biomed. Biotechnol. 2004, 2004, 326–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, K.; Zhang, H.; Han, Q.; Lan, J.; Chen, G.; Cao, G.; Yang, C. Effects of astragalus and ginseng polysaccharides on growth performance, immune function and intestinal barrier in weaned piglets challenged with lipopolysaccharide. J. Anim. Physiol. Anim. Nutr. 2019, 104, 1096–1105. [Google Scholar] [CrossRef] [PubMed]
  133. Hong, M.; Cai, Z.; Song, L.; Liu, Y.; Wang, Q.; Feng, X. Gynostemma pentaphyllum Attenuates the Progression of Nonalcoholic Fatty Liver Disease in Mice: A Biomedical Investigation Integrated with In Silico Assay. Evid. Based Complement. Altern. Med. 2018, 2018, 1–13. [Google Scholar] [CrossRef] [Green Version]
  134. Kim, M.H.; Lee, J.; Jung, S.; Kim, J.W.; Shin, J.-H.; Lee, H.-J. The involvement of ginseng berry extract in blood flow via regulation of blood coagulation in rats fed a high-fat diet. J. Ginseng Res. 2016, 41, 120–126. [Google Scholar] [CrossRef]
  135. Friedman, M. Mushroom Polysaccharides: Chemistry and Antiobesity, Antidiabetes, Anticancer, and Antibiotic Properties in Cells, Rodents, and Humans. Foods 2016, 5, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Pramudya, M.; Wahyuningsih, S.P.A. Immunomodulatory potential of polysaccharides from Coriolus versicolor against intracellular bacteria Neisseria gonorrhoeae. Veter. World 2019, 12, 735–739. [Google Scholar] [CrossRef] [PubMed]
  137. Lee, J.S.; Synytsya, A.; Kim, H.B.; Choi, D.J.; Lee, S.; Kim, W.J.; Jang, S.; Park, Y.I. Purification, characterization and immunomodulating activity of a pectic polysaccharide isolated from Korean mulberry fruit Oddi (Morus alba L.). Int. Immunopharmacol. 2013, 17, 858–866. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, C.; You, L.; Abbasi, A.M.; Fu, X.; Liu, R.H.; Li, C. Characterization of polysaccharide fractions in mulberry fruit and assessment of their antioxidant and hypoglycemic activities in vitro. Food Funct. 2015, 7, 530–539. [Google Scholar] [CrossRef] [PubMed]
  139. He, X.; Fang, J.; Ruan, Y.; Wang, X.; Sun, Y.; Wu, N.; Zhao, Z.; Chang, Y.; Ning, N.; Guo, H.; et al. Structures, bioac-tivities and future prospective of polysaccharides from Morus alba (white mulberry): A review. Food Chem. 2018, 245, 899–910. [Google Scholar] [CrossRef] [PubMed]
  140. Choi, J.W.; Synytsya, A.; Capek, P.; Bleha, R.; Pohl, R.; Park, Y.I. Structural analysis and anti-obesity effect of a pectic polysaccharide isolated from Korean mulberry fruit Oddi (Morus alba L.). Carbohydr. Polym. 2016, 146, 187–196. [Google Scholar] [CrossRef]
  141. Seydoux, E.; Liang, H.; Cauwelaert, N.D.; Archer, M.; Rintala, N.D.; Kramer, R.; Carter, D.; Fox, C.B.; Orr, M.T. Effective Combination Adjuvants Engage Both TLR and Inflammasome Pathways to Promote Potent Adaptive Immune Responses. J. Immunol. 2018, 201, 98–112. [Google Scholar] [CrossRef]
  142. Kuzmich, N.N.; Sivak, K.V.; Chubarev, V.N.; Porozov, Y.B.; Savateeva-Lyubimova, T.N.; Peri, F. TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines 2017, 5, 34. [Google Scholar] [CrossRef] [Green Version]
  143. Molteni, M.; Bosi, A.; Rossetti, C. Natural Products with Toll-Like Receptor 4 Antagonist Activity. Int. J. Inflamm. 2018, 2018, 1–9. [Google Scholar] [CrossRef] [Green Version]
  144. Muñoz-Carrillo, J.L.; Contreras-Cordero, J.F.; Gutiérrez-Coronado, O.; Villalobos-Gutiérrez, P.T.; Ramos-Gracia, L.G.; Hernández-Reyes, V.E. Cytokine Profiling Plays a Crucial Role in Activating Immune System to Clear Infectious Pathogens. In Immune Response Activation and Immunomodulation; Tyagi, R., Bisen, P.S., Eds.; IntechOpen: London, UK, 2019; pp. 1–30. [Google Scholar] [CrossRef] [Green Version]
  145. Paulina Pathria, T.L.L.; Varner, J.A. Targeting Tumor—Associated Macrophages in Cancer. Trends Immunol. 2019, 40, 310–327. [Google Scholar] [CrossRef]
  146. Juhas, U.; Ryba-Stanislawowska, M.; Szargiej, P.; Mysliwska, J. Different pathways of macrophage activation and polarization. Postepy higieny i medycyny doswiadczalnej. Postep. Hig. Med. Dosw. 2015, 69, 496–502. [Google Scholar] [CrossRef] [PubMed]
  147. Hamidzadeh, K.; Mosser, D.M. Purinergic Signaling to Terminate TLR Responses in Macrophages. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [Green Version]
  148. Lennartz, M.; Drake, J. Molecular mechanisms of macrophage Toll-like receptor—Fc receptor synergy. F1000 Res. 2018, 7, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Rőszer, T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediat. Inflamm. 2015, 2015, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Stunault, M.I.; Bories, G.; Guinamard, R.R.; Ivanov, S. Metabolism Plays a Key Role during Macrophage Activation. Mediat. Inflamm. 2018, 2018, 1–10. [Google Scholar] [CrossRef]
  151. Rana, A.K.; Li, Y.; Dang, Q.; Yang, F. Monocytes in rheumatoid arthritis: Circulating precursors of macrophages and osteoclasts and, their heterogeneity and plasticity role in RA pathogenesis. Int. Immunopharmacol. 2018, 65, 348–359. [Google Scholar] [CrossRef] [PubMed]
  152. Schepetkin, I.A.; Kirpotina, L.N.; Khlebnikov, A.I.; Balasubramanian, N.; Quinn, M.T. Neutrophil Immunomodulatory Activity of Natural Organosulfur Compounds. Molecules 2019, 24, 1809. [Google Scholar] [CrossRef] [Green Version]
  153. Jordan, S.A.; Cunningham, D.G.; Marles, R. Assessment of herbal medicinal products: Challenges, and opportunities to increase the knowledge base for safety assessment. Toxicol. Appl. Pharmacol. 2010, 243, 198–216. [Google Scholar] [CrossRef]
  154. Tall, A.R.; Yvan-Charvet, L. Cholesterol, inflammation and innate immunity. Nature reviews. Immunology 2015, 15, 104–116. [Google Scholar]
  155. Marx, T.K.; Glávits, R.; Endres, J.R.; Palmer, P.A.; Clewell, A.E.; Murbach, T.S.; Hirka, G.; Pasics, I. A 28-Day Repeated Dose Toxicological Study of an Aqueous Extract of Morus alba L. Int. J. Toxicol. 2016, 35, 683–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Chang, B.Y.; Kim, S.B.; Lee, M.K.; Park, H.; Kim, S.Y. Nonclinical Safety Assessment of Morus alba L. Fruits: Study of 90-D Toxicity in Sprague Dawley Rats and Genotoxicity in Salmonella. J. Food Sci. 2016, 81, T1328–T1335. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Morphology of Morus alba L.
Figure 1. Morphology of Morus alba L.
Foods 10 01966 g001
Figure 2. Phytochemicals of immature Morus alba L. fruits. Structure of (A) phenolic acid, (B) flanovols, and (C) tocopherols.
Figure 2. Phytochemicals of immature Morus alba L. fruits. Structure of (A) phenolic acid, (B) flanovols, and (C) tocopherols.
Foods 10 01966 g002
Figure 3. Comparison of murine peritoneal macrophage (RAW 264.7, ATCC® TIB-71™) activity in immature and mature M. alba L. fruits and Korean 6-year root red ginseng (KCG, daejeon, Korea). Macrophages were treated with extracts from immature and mature M. alba fruits or Korean 6-year root red ginseng extract (100 μg/mL) for 24 h. (A) Nitric oxide and (B) TNF-α production in supernatants were measured using ELISA. Data are expressed as mean ±SD values. All the data were confirmed by technical replicate 3 times. Significant differences were compared using repeated measures ANOVA followed by the Newman–Keuls multiple range test. Statistical significance was defined as * p < 0.05, *** p < 0.001. All statistical analyses were performed using GraphPad Software. Inc. (San Diego, CA, USA).
Figure 3. Comparison of murine peritoneal macrophage (RAW 264.7, ATCC® TIB-71™) activity in immature and mature M. alba L. fruits and Korean 6-year root red ginseng (KCG, daejeon, Korea). Macrophages were treated with extracts from immature and mature M. alba fruits or Korean 6-year root red ginseng extract (100 μg/mL) for 24 h. (A) Nitric oxide and (B) TNF-α production in supernatants were measured using ELISA. Data are expressed as mean ±SD values. All the data were confirmed by technical replicate 3 times. Significant differences were compared using repeated measures ANOVA followed by the Newman–Keuls multiple range test. Statistical significance was defined as * p < 0.05, *** p < 0.001. All statistical analyses were performed using GraphPad Software. Inc. (San Diego, CA, USA).
Foods 10 01966 g003
Figure 4. Summary of methods to improve immunological activity of Morus alba L.
Figure 4. Summary of methods to improve immunological activity of Morus alba L.
Foods 10 01966 g004
Figure 5. Structure of morrole A from Morus alba L. fruits.
Figure 5. Structure of morrole A from Morus alba L. fruits.
Foods 10 01966 g005
Table 1. Proximal composition, sugar, pectin, total phenol, and alkaloid of two species of mulberry.
Table 1. Proximal composition, sugar, pectin, total phenol, and alkaloid of two species of mulberry.
CompositionM. alba (White Mulberry)M. nigra (Black Mulberry)Reference
Ash (mg/100 g DW)0.57 ± 0.110.50 ± 0.08[10,16]
Protein(mg/100 g DW)1.55 ± 0.300.96 ± 0.16[10]
Total sugar (g/100 g FW)7.55 ± 1.016.64 ± 1.12[10]
Reducing sugar (g/100 g FW)5.90 ± 0.924.94 ± 0.73[10]
Pectin (g/100 g FW)Not detected0.76 ± 0.03[10]
Riboflavin (g/100 g FW)0.88 ± 0.000.04 ± 0.00[10]
Niacin (mg/100 g FW)3.10 ± 0.601.60 ± 0.10[10]
Total phenols (mg/100 g FW)1650 ± 12.25880 ± 7.20[10,11]
Alkaloid (mg/100 g FW)660 ± 5.25630 ± 5.93[10]
Total anthocyanins content(C3G µg/g frozen weight)911.8719[17]
(DW: dry weight; FW: fresh weight).
Table 2. Studies that investigated the pharmacological effects of phytochemicals present in M. alba L. extracts (grouped according to different parts or extract solvent types).
Table 2. Studies that investigated the pharmacological effects of phytochemicals present in M. alba L. extracts (grouped according to different parts or extract solvent types).
Pharmacological EffectPartSolventsExtract or PhytochemicalReferenceType (Species)
Immune enhancingFruitWaterCrude extract[25]in vitro and in vivo (mouse)
Polysaccharide[7,26]in vitro
MethanolPyrrol alkaloid Morrole A[27]in vitro
LeafWaterPolysaccharide[28,29,30]in vitro and in vivo (mouse, chicken)
RootWaterPolysaccharide[31]in vitro
Immune inhibitingFruitEthanolCrude extract[32]in vitro
LeafEthanolCrude extract[32,33,34,35]in vivo
MethanolCrude extract[36]in vivo (mouse)
StemEthanolCrude extract[37,38,39]in vitro
Oxyresveratrol[40]in vitro
RootMethanolKuwanon G[41]in vivo (mouse)
Cudraflavone B[42]in vivo (mouse)
AntioxidantFruitWaterPolysaccharide[43,44]in vitro
EthanolCrude extract[45]in vitro
MethanolHydroxycinnamic acid esters, Flavonol glycosides, andAnthocyanins[46]in vitro
Ethyl acetateCrude extract[47]in vitro
LeafWaterPolysaccharide[48]in vitro
EthanolCrude extract[29,34,49,50]in vitro
RootMethanolPhenolic contents[51]in vitro
Cardiovascular system protectionLeafEthanolCrude extract[52,53]in vitro, in vivo (Rat)
Ethyl acetateCrude extract[54]Ex vivo (Rat)
petroleum ether, dichloromethane, ethanolCrude extract[55]in vitro
RootWaterMoracinoside C, Moracin O, Moracin P[56]in vitro
EthanolCrude extract[57]in vitro, in vivo (Rat)
MethanolCrude extract[58]in vitro
Morusinol[59]in vitro, in vivo (Rat)
AntidiabeticFruitWaterCrude extract[43]in vitro
EthanolCrude extract[47,60]in vitroin vivo (Rat)
MethanolCrude extractantocyanins[61]in vitro, in vivo (Mouse)
LeafWaterCrude extract[62,63,64]in vitro, in vivo (Mouse)
EthanolCrude extract[34,65,66,67]in vitro, in vivo (Rat, Mouse)
Methanolchlorogenic acid, rutin, isoquercitrin loliolide,1-deoxynojirimycin, fagomine 2-O-alpha-D-galactopyranosyl-1-deoxynojirimycin[68]in vitro, in vivo (Rat, Mouse)
RootEthanolCrude extract[69]in vitro, in vivo (Rat)
MethanolMoran K[70]in vitro
Ethyl acetateCrude extract[71]in vitro
AntibacterialFruitWaterCrude extract[72]in vitro
LeafEthanolCrude extract[73,74,75]in vitro, in vivo (Rat)
1-deoxynojirimycin[76]in vitro
StemEthanolCrude extract[38]in vitro
AnticancerFruitEthanolButyl pyroglutamate quercetin, 3-O-β-D-glucoside kaempferol, 3-O-β-D-rutinoside, rutin, and 2-phenylethyl d-rutinoside[77]in vitro
Odisolane[78]in vitro
LeafEthanolFlavonoid[79,80]in vitro
MethanolMorin[81]in vitro
RootMethylene chlorideCrude extract[82]in vitro
MethanolCrude extract[83]in vitro
Albanol A[84]in vitro
Morusin[85]in vitro
Hepato-protectionFruitsWaterPolysaccharide[86]in vitro
ChloroformBenzofuran, Isomoracin, N-(N-benzoyl-l-phenylalanyl)-l-phenylalanol[87]in vitro
LeafWaterCrude extract[88,89]in vivo (Rat)
EthanolCrude extract[49,90]in vivo (mouse)
MethanolCrude extract[91]in vivo (mouse)
RootEthanolCrude extract[92]in vitro and in vivo (mouse)
NeuroprotectionFruitEthanolFlavonoid[93,94,95]in vitro and in vivo (mouse)
LeafEthanolsesquiterpenoid glucoside, aromatic glucoside, farnesylacetone derivative, flavan, and (9R)-hydroxyl-(10E, 12Z,15Z)-octadecatrienoic acid[96]in vitro
MethanolCrude extract[97,98]in vivo (mouse)
AcetoneCrude extract[99]in vivo (Rat)
RootMethanolmulberrofuran G, albanol B, kuwanon G[100]in vitro
AntiobesityFruitWaterPyrrol alkaloid[101]in vitro
LeafWaterCrude extract[102]in vivo (mouse)
EthanolCrude extract[49]in vivo (mouse)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chang, B.-Y.; Koo, B.-S.; Kim, S.-Y. Pharmacological Activities for Morus alba L., Focusing on the Immunostimulatory Property from the Fruit Aqueous Extract. Foods 2021, 10, 1966. https://doi.org/10.3390/foods10081966

AMA Style

Chang B-Y, Koo B-S, Kim S-Y. Pharmacological Activities for Morus alba L., Focusing on the Immunostimulatory Property from the Fruit Aqueous Extract. Foods. 2021; 10(8):1966. https://doi.org/10.3390/foods10081966

Chicago/Turabian Style

Chang, Bo-Yoon, Bong-Seong Koo, and Sung-Yeon Kim. 2021. "Pharmacological Activities for Morus alba L., Focusing on the Immunostimulatory Property from the Fruit Aqueous Extract" Foods 10, no. 8: 1966. https://doi.org/10.3390/foods10081966

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

Article Metrics

Back to TopTop