Next Article in Journal
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities
Next Article in Special Issue
UPLC-ESI-QTOF-MS Profiling of Phenolic Compounds from Eriocephalus africanus: In Vitro Antioxidant, Antidiabetic, and Anti-Inflammatory Potentials
Previous Article in Journal
Bifunctional Peptidomimetic G Protein-Biased Mu-Opioid Receptor Agonist and Neuropeptide FF Receptor Antagonist KGFF09 Shows Efficacy in Visceral Pain without Rewarding Effects after Subcutaneous Administration in Mice
Previous Article in Special Issue
The Chemical Signatures of Water Extract of Zingiber officinale Rosc
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Arbutin: Occurrence in Plants, and Its Potential as an Anticancer Agent

1
Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Šlechtitelů 27, 78371 Olomouc, Czech Republic
2
Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
3
Faculty of Pharmacy, Tripoli University, Tripoli 42300, Libya
4
Department of Biotechnology, Government V. Y. T. PG Autonomous College, Durg 491001, Chhattisgarh, India
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(24), 8786; https://doi.org/10.3390/molecules27248786
Submission received: 5 November 2022 / Revised: 7 December 2022 / Accepted: 8 December 2022 / Published: 11 December 2022

Abstract

:
Arbutin, a hydroquinone glucoside, has been detected in ca. 50 plant families, especially in the plants of the Asteraceae, Ericaceae, Proteaceae and Rosaceae families. It is one of the most widely used natural skin-whitening agents. In addition to its skin whitening property, arbutin possesses other therapeutically relevant biological properties, e.g., antioxidant, antimicrobial and anti-inflammatory, as well as anticancer potential. This review presents, for the first time, a comprehensive overview of the distribution of arbutin in the plant kingdom and critically appraises its therapeutic potential as an anticancer agent based on the literature published until the end of August 2022, accessed via several databases, e.g., Web of Science, Science Direct, Dictionary of Natural Products, PubMed and Google Scholar. The keywords used in the search were arbutin, cancer, anticancer, distribution and hydroquinone. Published outputs suggest that arbutin has potential anticancer properties against bladder, bone, brain, breast, cervix, colon, liver, prostate and skin cancers and a low level of acute or chronic toxicity.

1. Introduction

Arbutin (1, C12H16O7), also known as β-arbutin, is a hydroquinone glucoside (Figure 1). This compound was first reported from the leaves of Arbutus unedo L. (family: Ericaceae) [1]. Arbutin structurally differs from its isomer α-arbutin by the presence of a β-glucose unit instead of an α-glucose one. Since its discovery, arbutin (1) has been detected in ca. 50 other plant families. As this glycoside (1) is capable of inhibiting melanin production by inhibiting tyrosinase, it has long been used as a skin whitening (depigmenting) agent in various commercially available topical cosmetic products [2,3]. It should be mentioned here that tyrosinase is a multi-copper enzyme that plays a pivotal role in melanogenesis and enzymatic browning. The objectives of this review are to extensively explore, for the first time, the distribution of arbutin (1) in the plant kingdom (Table 1) and critically appraise its therapeutic potential as an anticancer agent. In order to achieve these objectives, an extensive literature search was conducted covering the literature published until the end of August 2022, accessed through several databases, e.g., Web of Science, Science Direct, Dictionary of Natural Products, PubMed and Google Scholar, and using the keywords, arbutin, cancer, anticancer, distribution and hydroquinone.

2. Distribution of Arbutin (1) in the Plant Kingdom

Arbutin (1) is widely distributed in the plant kingdom (Table 1) [4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162]. While the plants from the families, Asteraceae, Ericaceae, Proteaceae and Rosaceae are the main sources, to date, at least 45 other plant families have been reported to produce this glycoside (Table 1). In the Asteraceae, the genera Ainsliaea [7], Artemisia [20], Centaurea [46], Mutisia [80], Petasites [92], Podospermum [98] and Serratula [136] are known to produce arbutin (1), while the genera Arbutus [12], Arctostaphylos [15], Arctous [19], Calluna [17], Pyrola [101], Rhododendron [125] and Vaccinium [147] from the family Ericaceae are seven other major sources thereof (Table 1). Plants from at least seven genera within the Proteaceae, e.g., Bellendena [25], Cenarrhenes [25], Grevillea [59], Hakea [60], Heliciopsis [63], Leucadendron [73] and Persoonia [25] biosynthesize arbutin. The family Rosaceae includes the highest number of genera that produce the compound, including Cotoneaster [50], Eriobotrya [53], Fragaria [57], Malus [77], Prunophora [67], Pyrus [103], Rosa [126] and Sorbaria [140] (Table 1).
The highest concentration (ca. 1.7%) of arbutin was found in the leaves of Pyrus communis [163]. Certain plants from families like Fabaceae [5,72,74,86], Lamiaceae [87,128,141] and Plantaginaceae [132,154,160] are also notable sources of this hydroquinone glycoside (Table 1). At least three genera of each of the families Rutaceae [48,94,161] and Saxifragaceae [22,36,131] are known to produce arbutin (Table 1). While leaves are the main source of the compound, it is present in other plant parts, e.g., aerial parts, flowers, fruits, stem and twigs (Table 1). The presence of arbutin in roots was only reported in Picrorhiza scrophulariiflora [96].
Grisdale and Towers [163] demonstrated that arbutin is biosynthesized in the young leaves of Pyrus communis and Grevillea robusta from shikimic acid, as well as from phenylpropanoid compounds (Scheme 1). Evidence has suggested that the hydroquinone skeleton could have been formed by the removal of the propyl side chain of certain phenylpropane derivatives, e.g., cinnamic acid and phenylalanine. However, there are several reports available in the literature that describe various engineered and artificial methods for enhanced biosynthesis of arbutin [164]. For example, Shen et al. [165] demonstrated an artificial pathway in Escherichia coli for increased production of arbutin from simple carbon sources.

3. Anticancer Potential of Arbutin

In addition to its skin whitening property which has been known for at least seven decades, arbutin (1) has been shown to possess various other therapeutically relevant biological properties, e.g., antioxidant, antimicrobial and anti-inflammatory [164,165]; it also has the potential as an anticancer agent [166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181]. Information obtained from the published literature on arbutin shows that this compound possesses cytotoxic properties against several human cancer and tumor cell lines including bladder, bone, brain, breast, cervical, colon, gastric, liver, prostate and skin cancers (Table 2) [166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181]. Most of these activities have been demonstrated in vitro, and in some cases, plausible mechanisms of action, e.g., apoptosis, have been identified (Table 2). A pictorial summary is presented in Figure 2. The activity of arbutin against various cancer cell lines is discussed in the following subsections.

3.1. Bladder Cancer

When malignant cells are formed in bladder tissue or lining, it is known as bladder cancer; this disease affects more than 10,000 people every year in the UK [182]. A study conducted with the TCCSUP (an anaplastic transitional cell carcinoma in the neck of the urinary bladder) human bladder cancer cell line revealed that arbutin did not have any cytotoxicity against this cell line at a concentration of <500 mg/mL, but it considerably decreased proliferation of this cell line in a concentration- and time-dependent manner in the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay [166]. It was also shown that arbutin could time-dependently disrupt the cell cycle and inactivate extracellular signal-regulated kinase (ERK), which is an intrinsic regulator of cell proliferation and a key mediator of p53-dependent cell cycle arrest [183]. The ERK signaling pathway is implicated in the mitogenic signaling of several growth factors [166]. It was postulated that the cell cycle disruption by arbutin could be mediated by an increase in the cyclin-dependent kinase inhibitor p21(WAF1/C1P1)(p21). That study demonstrated that arbutin could inhibit the cell proliferation of bladder cancer cells in vitro via extracellular signal-regulated kinase inactivation and p21 up-regulation [166,183].

3.2. Brain Tumour

In a recent study on the effect of arbutin on brain tumor, it was found that it could kill C6 glioma cells by inducing apoptosis (IC50 = 30 mM) and inhibiting the inflammatory markers and P13/AKT/mTOR cascade [167]. It should be noted that P13/AKT/mTOR is an intracellular signaling pathway that regulates the cell cycle and, thus, is linked to cell proliferation. It is known that reactive oxygen species (ROS) can activate this cascade [184]. It was demonstrated that arbutin-generated excessive ROS could disrupt the mitochondrial membrane, resulting in apoptosis in cells and inhibition of the cell adhesion property of C6 glioma cells. C6 glioma cells are spindle-like cells; they are able to stimulate human glioblastoma multiforme (GBM) when injected into the brain of neonatal rats and have been used to develop a glioma model in Wistar rats. These cells exhibit the same histological features as human GBM [185]. Like bladder cancer, over 11,000 people are diagnosed with primary brain tumors every year in the UK, and a half of those are cancerous [186]. A recent study [167] suggested that arbutin could be a potential anti-brain tumor drug for the treatment of glioma. However, further studies are obviously necessary in this regard. An earlier study also showed significant antiproliferative activity of arbutin against C6 rat brain tumor cells in an enzyme-linked immunosorbent assay (ELISA) [168].

3.3. Breast Cancer

Breast cancer is the most common type of cancer in the UK and is usually treated with chemotherapy and radiotherapy [187]. In the search for natural products as potential cures for breast cancer, the cytotoxicity of an arbutin-containing methanol extract of Turnera diffusa was evaluated using the MTT assay against epithelial-like MDA-MB-231 breast cancer cells; the IC50 value was determined to be 30.67 mg/mL [145]. It was also assessed against the human breast carcinoma T-47D cell line, showing an IC50 value of 54.02 mg/mL. It was demonstrated that the cytotoxic effect of an arbutin-containing extract was mediated via apoptosis. It is worth noting that T-47D are epithelial cells obtained from a pleural effusion from a 54-year-old female patient with an infiltrating ductal carcinoma of the breast [188]. This assessment did not use purified arbutin, but rather, tested a crude methanol extract that contained arbutin as well as the flavone apigenin. More recently, Hazman et al. [169] reported the cytotoxicity of purified arbutin against the MCF-7 human breast cancer cell line; cytotoxicity was shown to be mediated through the induction of apoptosis via estrogen receptors and the alpha signal pathway, as well as through inflammation and genotoxicity. It was observed that the administration of a lethal dose (LD50 = 69.6 mM) of 50% arbutin could induce inflammation in MCF-7 cells linked to pro-inflammatory cytokine levels and increase genotoxicity in the cells. It was noted, however, that while at high doses arbutin could induce apoptosis, at low concentrations, it had the opposite effect, i.e., inhibiting apoptosis and thus, assisting cancer cell growth and survival. Earlier, a similar study was conducted to determine the cytotoxicity of arbutin against adriamycin-resistant MCF-7 and wild-type MCF-7 cell lines using the MTT assay [170]. It was found that arbutin at a high concentration (5–10 mM) was the least cytotoxic (15–42% inhibition of cell growth) among the tested phenolic compounds against both cell lines, while at low concentrations (0.32–1.25 mM), this compound raised cell viability by approximately 20%. The effective concentrations (EC50) of arbutin against the adriamycin-resistant MCF-7 and wild-type MCF-7 cell lines were 5.85 mM and >1000 mM, respectively.

3.4. Cervical Cancer

Cervical cancer is cancer of the cervix, caused predominantly by infection from certain human papillomaviruses [189]. This cancer is most common among young females under 45 years of age. An arbutin-rich methanolic extract of the leaves of Turnera diffusa, i.e., not purified arbutin, was tested for its cytotoxicity against human cervical carcinoma HPV-16 positive (SiHa) and HPV negative (C-33) tumor cell lines. Its cytotoxicity against these cell lines was much less prominent than its effect against the MDA-MB-231 breast cancer cell line [145]. The IC50 values of this methanol extract against the SiHa and C-33 cell lines were 50.14 and 40.1 mg/mL, respectively. A year later, Erenler et al. [168] reported the antiproliferative property of purified arbutin against the HeLa cell line, which was first developed from cervical cancer cells in 1951. A real-time cell analyzer single plate instrument (RTCA) and electronic cell sensory array, the xCELLigence RTCA, were used to analyze this antiproliferative effect at concentrations of 10, 50 and 100 mg/mL against the HeLA cell line; however, no attempt was made to determine the IC50 value. Additionally, none of the above experiments explored the possible mechanism of action of arbutin against the human cervical cell lines.

3.5. Colon Cancer

Arbutin displayed cytotoxicity against HCT-15 cell line, a quasidiploid human cell line derived from the large intestine of a male colorectal cancer patient [171]. In that study, culture cells were incubated with various concentrations of this hydroquinone glycoside for four days in a 5% CO2 incubator before cell numbers were counted. However, since this preliminary cytotoxicity result [171], no follow up data on the cytotoxicity of arbutin against various other colon cancer cell lines have been published in the literature, despite the fact that colon cancer, also known as bowel cancer, is one of the most common types of cancer among people of over 60 years of age in the UK [190].

3.6. Gastric Cancer

Gastric cancer, a form of stomach cancer, is the disease in which cancer cells grow in the lining of the stomach, whereas stomach cancer can be found anywhere in the stomach. This form of cancer is not common in the UK [191]. The inhibitory effect of several derivatives of arbutin, isolated from the leaves of Heliciopsis lobata, against cultured gastric carcinoma MGC-803 cells invasion was reported by Qi et al. [63]. All these derivatives contained various acyl substituents on the glycone moiety of arbutin, e.g., cinnamoyl and butenyl. Most of these compounds displayed a moderate level of activity, with IC50 values between 11 and 45 mg/mL.

3.7. Liver Cancer

While most of the aforementioned potential anticancer activities were assessed in vitro, recently, Zeng et al. [172] reported in vivo anticancer activity of arbutin against diethylnitrosamine-initiated liver carcinogenesis in rats. Liver cancer is one of the leading causes of cancer deaths worldwide and is the sixth most common form of cancer in humans, with almost a million new cases in 2020 [172,192]. The administration (30 mg/kg body weight) of arbutin was found to improve body weight, reduce liver weight, improve the albumin, globulin and total protein contents, reduce liver injury marker enzyme function and increase the c-JNK (c-Jun N-terminal kinase), caspase-8 and p53 contents in rats with diethylnitrosamine-triggered liver carcinogenesis.
This effect was attributed to the anti-inflammatory and antioxidant properties of arbutin, as evident from a series of in vitro bioassays with isolated rat liver tissue involving various inflammatory markers. Furthermore, arbutin was shown to decrease the expression of GRP78 (78-kDa glucose-regulated protein), PDIA4 (protein disulfide isomerase family A member 4), GRP94 (94-kDa glucose-regulated protein), ERDJ4 (endoplasmic reticulum-localized DNA J4), ATF4 (activating transcription factor 4) and GADD34 (growth arrest and DNA damage-inducible protein 34) in liver tissues. Earlier, a similar in vivo experiment, albeit a preliminary one, was conducted with hydroquinone, which is the aglycone of arbutin [173]. It was reported that hydroquinone could inhibit the initiation of DNA-reactive carcinogen acetylaminofluorene induction of rat liver carcinogenesis. However, the authors did not observe any significant body weight gains or changes in liver weight in hydroquinone-treated rats.
In addition to the above in vivo studies, there are a few in vitro studies available in the literature where the effect of arbutin was studied against the HepG2 hepatocellular cancer cell line [145,174,175]. An arbutin-rich methanolic extract of the leaves of Turnera difusa was found to exert cytotoxicity toward the HepG2 cell line with an IC50 value of 43.87 mg/mL [145]. Hazman et al. [174] reported the effects of α-arbutin (but not β-arbutin) on HepG2 cells and cisplatin toxication in this cell line. At low doses, α-arbutin did not show any genotoxicity or cytotoxicity toward HepG2 cells, and no effects on apoptosis, inflammation or proliferation were observed. However, when the same low dose was used with cisplatin, oxidative stress, inflammation and genotoxicity levels increased, resulting from cisplatin toxicity without any change in caspase 3 levels. At high doses, α-arbutin displayed anticarcinogenic effects, mediated through increased oxidative stress, genotoxicity, inflammation and apoptosis and suppression of cell proliferation. A decade before this study, Kang et al. [175] reported on the in vitro cytotoxicity of arbutin in the HepG2 cell line.

3.8. Melanoma or Skin Cancer

Melanoma is a type of skin cancer, the most common sign of which is the appearance of a new mole or a noticeable change in an existing mole [193]. Melanoma is thought to be caused by exposure to ultraviolet (UV) light from the sun or from a sunbed. It is the fifth most common cancer in the UK and there are ca. 16,000 new cases of it reported in the UK every year. Jiang et al. [176] reported the potential anti-melanoma activity of arbutin and showed its effect on melanogenesis, as well as its pro-apoptotic effect, on B16 murine melanoma cells. Arbutin was shown to significantly reduce cell viability, promote cell apoptosis, cause G1 cell cycle arrest (after 24 h of treatment) and induce mitochondrial disruption in B16 murine melanoma cells. It also caused a reduction in the expression of B-cell lymphoma-extra large (Bcl-xL) and Bcl-2 arbutin-treated cells. The inhibition of cell viability by arbutin was found to be time- and dose-dependent, and it could inhibit melanogenesis by ca. 46% at a concentration of 5.4 mM. Its pro-apoptotic effect was detected by flow cytometry using Annexin V-FITC labeling for the detection of phosphatidylserine externalization. Arbutin was found to be able to cause apoptosis in 23.7% of the cells after 24 h of treatment at 5.4 mM. The results from this study indicated that arbutin could be a candidate for anti-melanoma drug development. Earlier, the anti-skin cancer potential of arbutin was reported by studying the molecular spectroscopic behavior of this compound and its action on the carcinogen trans-crotonaldehyde [177].

3.9. Osteosarcoma

Osteosarcoma is a type of bone cancer. It starts in the cells that form bones, especially long bones. Children, teens and young adults are the main sufferers from this cancer [194]. Just over 500 new cases are reported each year in the UK National Health Service (NHS) [195]. Wang et al. [178] demonstrated that arbutin could time- and dose-dependently suppress the progression of osteosarcoma in vitro using the osteosarcoma cell lines MG-63 and SW1353 and applying the Cell Counting Kit-8 assay. It was suggested that arbutin could inhibit osteosarcoma cell proliferation, migration and invasion via miR-338-3pl MTHFD1L (methylenetetrahydrofolate dehydrogenase (NADP+ Dependent) 1 Like) and by inactivating the AKT (protein kinase B)/mTOR (mammalian target of rapamycin) signaling pathway.

3.10. Prostate Cancer

Safari et al. [179] first reported the anti-prostate cancer potential of arbutin and looked into the molecular mechanism of activity against the prostate cancer cell line LNCap (androgen-sensitive human prostate adenocarcinoma cells). It was demonstrated that 1 mM of arbutin could induce apoptosis, reduce the level of reactive oxygen and decrease the expression of pro-inflammatory 1L-1β (interleukin-1 beta) and TNF-α (tumor necrosis factor alpha) genes. A year later, the effect of arbutin on the expression of tumor suppressor p53, BAX/BCL-2 (BCL 2 associated X/B cell lymphoma protein 2) ratio and oxidative stress induced by t-butyl hydroperoxide in fibroblast and LNCap cell lines was studied [180]. It was observed that arbutin could enhance the total antioxidant power and cell viability in the MTT assay, as well as reducing the BAX/BCL-2 ratio, p53 mRNA expression and necrosis in fibroblasts exposed to an oxidative agent. Additionally, it was shown to decrease cell viability, induce apoptosis and increase the BAX/BCL-2 ratio in LNCap cells at certain concentrations (e.g., 1 mM).
Recently, a dichloromethane extract of the leaves of Arbutus pavarii was shown to possess cytotoxicity against the PC3 human prostate cancer cell line. Employing a bioassay-guided isolation protocol, arbutin was isolated as one of the major bioactive compounds [12]. One in eight men in the UK is likely to have prostate cancer, which can develop when cells in the prostate start to grow in an uncontrolled way [196]. Prostate cancer is the most common cancer in men and more than 52,000 men are diagnosed with it every year in the UK. Fatalities from this disease every year in the UK are over 12,000. The in vitro activity of arbutin against prostate cancer cell lines requires further extensive investigation to examine the potential of this compound or its analogues as prostate cancer therapeutics.

3.11. Miscellaneous

In discussing the regulatory impact of miRNA-338-3p on cancer growth and migration, the antitumor effect of arbutin, i.e., suppressing cancer progression by promoting the expression of miRNA-338-3p, was highlighted by Mirzaei et al. [181].

4. Toxicological Aspects

Generally, arbutin is considered safe for external use, particularly at the concentrations at which it is used in various cosmetic products. However, a few studies conducted to date on the toxicity of this compound have reveled certain levels of in vivo and in vitro toxicity at various concentrations [197]. At high doses, the aglycone hydroquinone can exert hepato- and nephron-toxicity and mutagenicity [197]. Kang et al. [175] demonstrated the ability of arbutin to induce immunotoxicity in splenocyte cultures from mice. The genotoxic effect of arbutin on the differential gene expression profiling in A375 human malignant melanoma cells through its effect on tumorigenesis and related side-effects has been reported [198]. It was found that the level of toxicity may be dependent on the route of exposure, as well as on the sex, species and strain in rodents. Meanwhile, the subchronic and chronic toxicity in animal models was limited to nephrotoxicity [199]. However, no developmental and reproductive toxicity or carcinogenicity have been detected with arbutin [200,201]. Information available in various online databases suggests that it may exert a low level of toxicity at high doses when given orally to mice (LD50 = 9804 mg/kg) and rats (LD50 = 8715 mg/kg) [202], as well as dermal toxicity in rat and mouse (LD50 = 928 mg/kg). However, far more published papers have highlighted various protective and health promoting effects of arbutin, e.g., cytoprotective and hepatoprotective effects [103,202,203,204], the benefits of which probably outweigh the minimal toxic effect of this compound.

5. Conclusions

Arbutin is widely distributed in the plant kingdom; plants from the Asteraceae, Ericaceae, Proteaceae and Rosaceae families are the main sources of this compound. However, the compound has been detected in at least 45 other plant families to date. Published data suggest that arbutin possesses potential anticancer properties against bladder, bone, brain, breast, cervix, colon, liver, prostate and skin cancers, and a low level of toxicity. Further in silico studies and in vivo pre-clinical and randomized clinical investigations are essential to establish its true potential as an anticancer drug candidate.

Author Contributions

All authors (L.N., A.A.-G., A.K. and S.D.S.) contributed equally to the data collection and compilation of information. Additionally, L.N. and S.D.S. played the lead role in writing, editing and submission of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The European Regional Development Fund–Project ENOCH (No. CZ.02.1.01/0.0/0.0/16_019/0000868) and the Czech Grant Agency Grant–Project (23-05474S).

Acknowledgments

Lutfun Nahar gratefully acknowledges the financial support of the European Regional Development Fund–Project ENOCH (No. CZ.02.1.01/0.0/0.0/16_019/0000868) and the Czech Grant Agency Grant–Project (23-05474S), and A. Al-Groshi thanks the Libyan Government for a studentship.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thies, H.; Sulc, D. Arbutus unedo L. I. Determination of arbutin in the leaves of the strawberry tree. Pharmazie 1950, 5, 553–555. [Google Scholar] [PubMed]
  2. Liu, Y.F.; Liu, D.Z.; Xie, Z.M. Rapid and specific fluorescence method for the quantification of arbutin in cosmetics. Anal. Lett. 2022, 55, 318–326. [Google Scholar] [CrossRef]
  3. Boo, Y.C. Arbutin as a skin depigmenting agent with antimelanogenic and antioxidant properties. Antioxidants 2021, 10, 1129. [Google Scholar] [CrossRef] [PubMed]
  4. Kubo, I.; Ying, B.P. Phenolic constituents of California buckeye fruit. Phytochemistry 1992, 31, 3793–3794. [Google Scholar] [CrossRef]
  5. Suktap, C.; Lee, H.K.; Amnuaypol, S.; Suttisri, R.; Sukrong, S. Would healing effect of flavonoid glycosides from Afgekia mahidolae B L Burtt & Chermsir. leaves. Rec. Nat. Prod. 2018, 12, 391–396. [Google Scholar]
  6. Ni, J.C.; Shi, J.T.; Tan, Q.W.; Chen, Q.J. Phenylpropionamides, piperidine, and phenolic derivatives from the fruit of Ailanthus Altissima. Molecules 2017, 22, 2107. [Google Scholar] [CrossRef] [Green Version]
  7. Ma, C.Y.; He, N.; Zhao, Y.Y.; Xia, D.D.; Wei, J.F.; Kang, W.Y. Antimicrobial mechanism of hydroquinone. Appl. Biochem. Biotechnol. 2019, 189, 1291–1303. [Google Scholar] [CrossRef]
  8. Ahmed, M.; Ramachandraiah, K.; Jiang, G.H.; Eun, J.B. Effects of ultra-sonication and agitation on bioactive compounds and structure of Amaranth extract. Foods 2020, 9, 1116. [Google Scholar] [CrossRef]
  9. Gins, M.S.; Gins, V.K.; Motyleva, S.M.; Kulikov, I.M.; Medvedev, S.M.; Pivovarov, V.F.; Mertvishcheva, M.E. Metabolites with antioxidant and protective functions from leaves of vegetable Amaranth (Amaranthus tricolor L.). Sel’skokhozyaistvennaya Biol. 2017, 52, 1030–1040. [Google Scholar] [CrossRef]
  10. Suwanprasert, T. The analysis of arbutin in Mao (Antidesma thwaitesianum Muell. Arg.) extracts. Pertanika J. Trop. Agric. Sci. 2018, 41, 621–636. [Google Scholar]
  11. Sakar, M.K.; Berkman, M.Z.; Calis, I.; Ruedi, P. Constituents of Arbutus Andrachne. Fitoterapia 1991, 62, 176–177. [Google Scholar]
  12. Al Groshi, A.; Nahar, L.; Ismail, F.M.D.; Evans, A.R.; Sarker, S.D. Dichloromethane extract of the leaves of Arbutus pavarii Pamp. exhibits cytotoxicity against the prostate cancer cell line PC3: A bioassay-guided isolation and identification of arbutin and betulinic acid methyl ester. J. Nat. Prod. Discovery. 2022, in press. [Google Scholar] [CrossRef]
  13. Karikas, G.A.; Giannitsaros, A. Phenolic glucosides from the leaves of Arbutus Unedo. Plantes Med. Et. Phytother. 1990, 24, 27–30. [Google Scholar]
  14. Pawlowska, A.M.; De Leo, M.; Braca, A. Phenolics of Arbutus unedo. (Ericaceae) fruits: Identification of anthocyanins and gallic acid derivatives. J. Agric. Food Chem. 2006, 54, 10234–10238. [Google Scholar] [CrossRef]
  15. Panusa, A.; Petrucci, R.; Marrosu, G.; Multari, G.; Gallo, F.R. UHPLC-PDA-ESI-TOF/MS metabolic profiling of Arctostaphylos pungens and Arctostaphylos uva-ursi. A comparative study of phenolic compounds from leaf methanolic extracts. Phytochemistry 2015, 115, 79–88. [Google Scholar] [CrossRef]
  16. Hamberg, T. Determination of arbutin in bearberry leaves under various conditions. Sven. Farm. Tidskr. 1952, 56, 401–407. [Google Scholar]
  17. Sticher, O.; Soldati, F.; Lehmann, D. High-performance liquid-chromatographic separation and quantitative-determination of arbutin, methylarbutin, hydroquinone and hydroquinonemonomethylether in Arctostaphylos, Bergenia, Calluna and Vaccinium species. Planta Med. 1979, 35, 253–261. [Google Scholar] [CrossRef]
  18. Kreitmair, H. Arctostaphylos uva-ursi—Bearberry. Pharmazie 1953, 8, 347–349. [Google Scholar]
  19. Fursa, N.S.; Ermolaeva, O.M. Arctous alpina leaves: Arbutin content. Farmatsiya 2013, 13–15. [Google Scholar]
  20. Garai, S.; Garai, S.; Jaisankar, P.; Singh, J.K.; Elango, A. A comprehensive study on crude methanolic extract of Artemisia pallens (Asteraceae) and its active component as effective corrosion inhibitors of mild steel in acid solution. Corros. Sci. 2012, 60, 193–204. [Google Scholar]
  21. Noikotr, K.; Chaveerach, A.; Sudmoon, R.; Tanee, T.; Patarapadungkit, N. Phytochemicals, cytotoxicity and genotoxicity of three Artocarpus species reveal arbutin in A. Lacucha. Scienceasia 2018, 44, 170–178. [Google Scholar] [CrossRef] [Green Version]
  22. Rajbhandari, M.; Lalk, M.; Mentel, R.; Lindequist, U. Antiviral activity and constituents of the Nepalese medicinal plant Astilbe Rivularis. Rec. Nat. Prod. 2011, 5, 138–142. [Google Scholar]
  23. Godevac, D.; Stankovic, J.; Novakovic, M.; Andelkovic, B.; Dajic-Stevanovic, Z.; Petrovic, M. Phenolic compounds from Atriplex littoralis and their radiation mitigating activity. J. Nat. Prod. 2015, 78, 2198–2204. [Google Scholar] [CrossRef] [PubMed]
  24. Gonzalez-Cortazar, M.; Lopez-Gayou, V.; Tortoriello, J.; Dominguez-Mendoza, B.E.; Rios-Cortes, A.M.; Ble-Gonzalez, E.A.; Zamilpa, A. Antimicrobial gastrodin derivatives isolated from Bacopa procumbens. Phytochem. Lett. 2019, 31, 33–38. [Google Scholar] [CrossRef]
  25. Dean, B.J.; Kilah, N.L.; Jordan, G.J.; Bissember, A.C.; Smith, J.A. Arbutin derivatives isolated from ancient Proteaceae: Potential phytochemical markers present in Bellendena, Cenarrhenes and Pers. Genera. J. Nat. Prod. 2018, 81, 1241–1251. [Google Scholar] [CrossRef]
  26. Han, X.N.; Liu, C.Y.; Liu, Y.L.; Xu, Q.M.; Li, X.R.; Yang, S.L. New triterpenes and other constituents from the fruits of Benincasa hispida (Thunb.) Cogn. J. Agric. Food Chem. 2014, 61, 12692–12699. [Google Scholar] [CrossRef]
  27. Fuji, M.; Miyaichi, Y.; Tomimori, T. Studies on Nepalese crude drugs: XXII. On the phenolic constituents of the rhizome of Bergenia ciliata (Haw.) Sternb. Nat. Med. 1996, 50, 404–407. [Google Scholar]
  28. Roselli, M.; Lentini, G.; Habtemariam, S. Phytochemical, antioxidant and anti-alpha-glucosidase activity evaluations of Bergenia Cordifolia. Phytother. Res. 2012, 26, 908–914. [Google Scholar] [CrossRef]
  29. Kuz’min, V.I.; Gontar’E, M.; Pushkarev, G.N. Productivity of Bergenia crassifolia raw material and the content of phenol compounds in it Western Baikal-Amur railway and Khakass autonomous Oblast Russian-SFSR USSR. Rastit. Resur. 1985, 21, 180–183. [Google Scholar]
  30. Pozharitskaya, O.N.; Ivanova, S.A.; Shikov, A.N.; Makarov, V.G.; Galamnosi, B. Separation and evaluation of free radical-scavenging activity of phenol components of green, brown, and black leaves of Bergenia crassifolia by using HPTLC-DPPH center dot method. J. Sep. Sci. 2007, 30, 2447–2451. [Google Scholar] [CrossRef]
  31. Pop, C.; Vlase, L.; Tamas, M. Natural resources containing arbutin. determination of arbutin in the leaves of Bergenia crassifolia (L.) Fritsch acclimated in Romania. Not. Bot. Horti Agrobot. Cluj-Napoca 2009, 37, 129–132. [Google Scholar]
  32. Qu, Y.X.; Zhang, C.N.; Liu, R.H.; Wu, H.; Sun, Y.; Zhang, N.; Nima, C.R.; Danpei, Q.Z.; Zhang, S.F.; Sun, Y.K. Rapid characterization the chemical constituents of Bergenia purpurascens and explore potential mechanism in treating osteoarthritis by ultra-high performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry combined with network pharmacology. J. Sep. Sci. 2020, 43, 3333–3348. [Google Scholar] [PubMed]
  33. Friedrich, H. Studies on tanning principle of Bergenia species and its relation to arbutin. Pharmazie 1954, 9, 138–155. [Google Scholar] [PubMed]
  34. Frohne, D. Occurrence of arbutin in Saxafragaceae—D. Pharmazie 1969, 24, 701–702. [Google Scholar] [PubMed]
  35. Friedrich, H.; Wehnert, H.U. Distribution of arbutin and bergenine in Bergenia plants. Arch. Pharm. 1973, 306, 757–764. [Google Scholar] [CrossRef] [PubMed]
  36. Hou, Y.; Ali, I.; Li, Z.; Sulaiman, A.; Aziz, S.; Chen, L.; Hussain, H.; Cui, L.; Wang, D.J.; Zheng, X. Separation of constituents from Bergenia stracheyi (Hook. F. & Thoms.) Engl. by high-speed countercurrent chromatography with elution mode and its antidiabetic and antioxidant in vitro evaluation. J. Sep. Sci. 2020, 44, 202000999. [Google Scholar]
  37. Koltunov, Y. The effect of the stem rot at composition and content of phenolic compounds in leaves of Birch (Betula pendula Roth.). Khimia Rastit. Syr’ja 2019, 169–176. [Google Scholar] [CrossRef]
  38. Shen, Y.B.; Kojima, Y.; Terazawa, M. Four glucosides of p-hydroxyphenyl derivatives from birch leaves. J. Wood Sci. 1999, 45, 332–336. [Google Scholar] [CrossRef]
  39. Wang, D.-H.; Sook, C.H. Identification of antioxidant and anti-tyrosinase activity of phenolic components isolated from Betula Schmidtii. Korean J. Food Nutr. 2021, 34, 553–559. [Google Scholar]
  40. Morikawa, H.; Kasai, R.; Otsuka, H.; Hirata, E.; Shinzato, T.; Aramoto, M.; Takeda, Y. Terpenic and phenolic glycosides form leaves of Breynia officinalis Hemsl. Chem. Pharm. Bull. 2004, 52, 1086–1090. [Google Scholar] [CrossRef] [Green Version]
  41. Li, C.-H.; Yang, X.-D.; Zhao, J.-F.; Li, L. The chemical constituents of Breynia Rostrata. Yao Xue Xue Bao 2006, 41, 125–127. [Google Scholar] [PubMed]
  42. Leifertova, I.; Lisa, M.; Pechova, Z.; Prokes, J. Evaluation of the composition of Calluna vulgaris L. Hull. Farm. Obz. 1989, 58, 349–354. [Google Scholar]
  43. Fursa, N.S.; Onegin, S.V. Arbutin levels in the ling (Calluna Vulgaris). Farmatsiya 2007, 2007, 12–14. [Google Scholar]
  44. Gupta, P.; Patil, D.; Patil, A. Qualitative HPTLC phytochemical profiling of Careya arborea Roxb. bark, leaves and seeds. 3 Biotech 2019, 9, 311. [Google Scholar] [CrossRef]
  45. Mosaddik, M.A.; Flowers, A.; Karagianis, G.; Waterman, P.G. New phenolic glycosides from the stems and leaves of Casearia Multinervosa. Nat. Prod. Res. 2006, 20, 641–647. [Google Scholar]
  46. Gulcemal, D.; Alankus-Caliskan, O.; Karaalp, C.; Ors, A.U.; Ballar, P.; Bedird, E. Phenolic glycosides with antiproteasomal activity from Centaurea urvillei DC. subsp. urvillei. Carbohydr. Res. 2010, 345, 2529–2533. [Google Scholar] [CrossRef]
  47. Basavand, E.; Charkhabi, N.F.; Khodaygan, P.; Rahimian, H. Agrobacterium pusense, a new plant tumour-inducing pathogen isolated from Lawson cypress. For. Pathol. 2020, 51, e12655. [Google Scholar] [CrossRef]
  48. Wang, R.M.; He, R.P.; Li, Z.H.; Wang, L. LC-Q-Orbitrap-MS/MS characterization, antioxidant activity, and alpha-glucosidase-inhibiting activity with in silico analysis of extract from Clausena Indica (Datz.) Oliv fruit pericarps. Front. Nutr. 2021, 8, 727087. [Google Scholar] [CrossRef]
  49. Anesyan, E.T.; Nersesyan, Z.M.; Parkhomenko, A.Y. Chemical composition of the above-ground part of Coriandrum sativum. Khimiko-Farnatsevticheskii Zhurnal 2007, 41, 30–34. [Google Scholar]
  50. Palme, E.; Bilia, A.R.; Morelli, I. Flavonols and isoflavonols from Cotoneaster simonsii. Phytochemistry 1996, 42, 903–905. [Google Scholar] [CrossRef]
  51. Yahara, S.; Domoto, H.; Sugimura, C.; Nohara, T.; Niho, Y.; Nakajima, Y.; Ito, H. An alkaloid and two lignans from Cuscuta Sinensis. Phytochemistry 1994, 37, 1755–1757. [Google Scholar] [CrossRef]
  52. Feng, W.-S.; Cao, X.-W.; Kuang, H.-X.; Zheng, X.-K. Flavanone O-glycosides from the rhizomes of Dryopteris Sublaeta. Yao Xue Xue Bao 2007, 42, 867–871. [Google Scholar] [PubMed]
  53. Hong, Y.P.; Qiao, Y.C.; Lin, S.Q.; Jiang, Y.M.; Chen, F. Characterization of antioxidant compounds in Eriobotrya fragrans Champ. Leaf. Sci. Hortic. 2008, 118, 288–292. [Google Scholar] [CrossRef]
  54. Cadiz-Gurrea, M.D.; Fernandez-Arroyo, S.; Joven, J.; Segura-Carretero, A. Comprehensive characterization by UHPLC-ESI-Q-TOF-MS from an Eryngium bourgatii extract and their antioxidant and anti-inflammatory activities. Food Res. Int. 2013, 50, 197–204. [Google Scholar] [CrossRef]
  55. Bokesch, H.R.; Wamiru, A.; Le Grice, S.F.J.; Beutler, J.A.; Mckee, T.C.; McMahon, J.B. HIV-1 ribonuclease H inhibitory phenolic glycosides from Eugenia Hyemalis. J. Nat. Prod. 2008, 71, 1634–1636. [Google Scholar] [CrossRef] [Green Version]
  56. Hu, Q.H.; Wang, D.; Yu, J.; Ma, G.X.; Pei, F.; Yang, W.J. Neuroprotective effects of six components from Flammulina velutipes on H2O2-induced oxidative damage in PC12 cells. J. Funct. Foods 2018, 37, 586–593. [Google Scholar] [CrossRef]
  57. Nemec, S. Phenolics in strawberry root. Ann. Bot. 1973, 37, 935–941. [Google Scholar] [CrossRef]
  58. Garcia, J.; Mpondo, E.M.; Kaouadji, M.; Mariotte, A.M. Arbutin derivatives in Gentiana Pyrenaica. J. Nat. Prod. 1989, 52, 858–860. [Google Scholar] [CrossRef]
  59. Wang, H.; Leach, D.; Thomas, M.C.; Blanksby, S.J.; Forster, P.I.; Waterman, P.G. Bisresorcinols and arbutin derivatives from Grevillea banksii R. Br. Nat. Prod. Commun. 2008, 3, 57–64. [Google Scholar] [CrossRef] [Green Version]
  60. Manju, M.; Varma, R.S.; Parthasarathy, M.R. New arbutin derivatives from leaves of Grevillea robusta and Hakea Saligna. Phytochemistry 1977, 16, 793–794. [Google Scholar] [CrossRef]
  61. Ahmed, A.S. Phytochemical and biological study of Grevillea robusta A. Cunn, cultivated in Egypt. Bull. Pharm. Sci. 2006, 29, 272–288. [Google Scholar] [CrossRef] [Green Version]
  62. Perry, N.B.; Benn, M.H.; Foster, L.M.; Routledge, A.; Weavers, R.T. The glycosidic precursor of (Z)-5-ethylidene-1(5H)-furanone in Halocarpus biformisjuvenile foliage. Phytochemistry 1996, 42, 453–459. [Google Scholar] [CrossRef] [PubMed]
  63. Qi, W.Y.; Ou, N.; Wu, X.D.; Xu, H.M. New arbutin derivatives from the leaves of Heliciopsis lobata with cytotoxicity. Chin. J. Nat. Remedies 2016, 14, 789–793. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, B.; Yamg, P.-P.; Wang, P.-L.; Ling, H.-L.; Chen, M. Study on the constituents of Herpetospermum Caudigerum. J. Chin. Med. Mater. 2012, 35, 1080–1082. [Google Scholar]
  65. Rout, D.; Dash, U.C.; Kanhar, S.; Swain, S.K.; Sahoo, A.K. Homalium zeylanicum attenuates streptozotocin-induced hyperglycemia and cellular stress in experimental rats via attenuation of oxidative stress imparts inflammation. J. Ethnopharmacol. 2022, 283, 114649. [Google Scholar] [CrossRef]
  66. Gao, W.Y.; Wang, B.D.; Li, Y.M.; Jiang, S.H.; Zhu, D.Y. A new alkaloid and arbutin from the whole plant Huperzia serrata. Chin. J. Chem. 2000, 18, 614–616. [Google Scholar]
  67. Kim, Y.C. Whitening efficacy of water-soluble extracts from Prunophora salicina’s (Daeseokjosaeng, Purplekin, Formosa) peel. J. Investig. Cosmetol. 2013, 9, 27–32. [Google Scholar]
  68. Miura, H.; Inoue, E.; Kitamura, Y.; Sugii, M. Examination and determination of arbutin in the leaves of Viburnum spp. and Ilex Spp. Shoyakugaku Zasshi 1985, 39, 181–184. [Google Scholar]
  69. Gousiadou, C.; Li, H.Q.; Gotfredsen, C.; Jensen, S.R. Iridoids in Hydrangeaceae. Biochem. Syst. Ecol. 2016, 64, 122–130. [Google Scholar] [CrossRef] [Green Version]
  70. Fuentealba, C.; Hernandez, I.; Saa, S.; Toledo, L.; Burdiles, P.; Chirinos, R.; Campos, D.; Brown, P.; Pedreschi, R. Colour and in vitro quality attributes of walnuts from different growing conditions correlate with key precursors of primary and secondary metabolism. Food Chem. 2017, 232, 664–672. [Google Scholar] [CrossRef]
  71. Niemann, G.J. Phenolics from Larix needles.5. Phenolic glucosides from needles of Larix leptolepis. Phytochemistry 1973, 12, 723–724. [Google Scholar] [CrossRef]
  72. Tsopmo, A.; Muir, A.D. Chemical profiling of lentil (Lens culinaris Medik.) cultivars and isolation of compounds. J. Agric. Food Chem. 2010, 58, 8715–8721. [Google Scholar] [CrossRef] [PubMed]
  73. Glennie, C.W. Flavonoid glycosides of Leucadendron and their chemotaxonomic significance. J. South Afr. Bot. 1980, 46, 147–156. [Google Scholar]
  74. Hernandez-Bolio, G.I.; Kutzner, E.; Eisenreich, W.; Torres-Acosta, J.F.D.; Pena-Rodriguez, L.M. The use of H-1-NMR metabolomics to optimise the extraction and preliminary identification of anthelmintic products from the leaves of Lysiloma Latisiliquum. Phytochem. Anal. 2018, 29, 413–420. [Google Scholar] [CrossRef]
  75. Khan, S.; Kardar, M.N.; Siddiqui, B.S. Arbutin derivatives from the seeds of Madhuca latifolia. Nat. Prod. Commun. 2011, 6, 1661–1664. [Google Scholar] [CrossRef] [Green Version]
  76. Begum, S.; Banerjee, A.; De, B. Antioxidant and enzyme inhibitory properties of Magnifera indica leaf extract. Nat. Prod. J. 2020, 10, 384–394. [Google Scholar]
  77. Nesterova, N.V.; Kuzmenko, A.N.; Kuzmenko, I.A.; Krasnyk, I.I., Jr.; Evgrafov, A.A. Quantitative determination of arbutin in leaves of Malus sylvestris by method of high-efficient liquid chromatography. Mosc. Univ. Chem. Bull. 2019, 74, 42–45. [Google Scholar] [CrossRef]
  78. Nesterova, N.V.; Samylina, I.A. Impact of a preservation method on the content of biologically active substances in apples. Farmatsiya 2017, 66, 24–26. [Google Scholar]
  79. Gug, K. Physiological and whitening effects of Morus alba extracts. J. Chosun Nat. Sci. 2012, 5, 46–52. [Google Scholar] [CrossRef] [Green Version]
  80. Catalano, S.; Cioni, P.L.; Flamini, G.; Defeo, V.; Morelli, I. Chemical investigation of the aerial parts of Mutisia acuminata. Int. J. Pharmacogn. 1995, 33, 73–74. [Google Scholar] [CrossRef]
  81. Daily, A.; Seligmann, O.; Nonnenmacher, G.; Fessler, B.; Wong, S.M.; Wagner, H. New chromone, coumarin and coumestan derivatives from Mutisia Acuminata Var Hirsute. Planta Med. 1988, 54, 270. [Google Scholar] [CrossRef] [PubMed]
  82. Zhong, X.N.; Otsuka, H.; Ide, T.; Hirata, E.; Takushi, A.; Takeda, Y. Hydroquinone glycosides from leaves of Myrsine Seguinii. Phytochemistry 1998, 49, 2149–2153. [Google Scholar] [CrossRef]
  83. Suau, R.; Cuevas, A.; Valpuest, V.; Reid, M.S. Arbutin and sucrose in the leaves of the resurrection plant Myrothamnus flabellifolia. Phytochemistry 1991, 30, 2555–2556. [Google Scholar] [CrossRef]
  84. Engelhardt, C.; Petereit, F.; Anke, J.; Hensel, A. A new arbutin derivative from the herb of Myrothamnus flabellifolia Welw. Pharmazie 2007, 62, 558–559. [Google Scholar] [CrossRef]
  85. Moniava, I.I. Arbutin from Onobrychis kachetica. Chem. Nat. Compd. 1970, 6, 270. [Google Scholar] [CrossRef]
  86. Regos, I.; Urbanella, A.; Treutter, D. Identification and quantification of phenolic compounds from the forage legume Sainfoin (Onobrychis viciifolia). J. Agric. Food Chem. 2009, 57, 5843–5852. [Google Scholar] [CrossRef]
  87. Karioti, A.; Milosevic-Ifantis, T.; Pachopos, N.; Niryiannaki, N.; Hadjipavlou-Latina, D.; Skaltsa, H. Antioxidant, anti-inflammatory potential and chemical constituents of Origanum dubium Boiss., growing in Cyprus. J. Enzym. Inhib. Med. Chem. 2015, 30, 38–43. [Google Scholar] [CrossRef]
  88. Assaf, M.H.; Ali, A.A.; Makboul, M.A.; Beck, J.P.; Anton, R. Preliminary study of phenolic glycosides from Origanum majorana–quantitative estimation of arbutin–cytotoxic activity of hydroquinone. Planta Med. 1987, 53, 343–345. [Google Scholar] [CrossRef]
  89. Moghrovyan, A.; Sahakyan, N.; Babayan, A.; Chichoyan, N.; Petrosyan, M.; Trchounian, A. Essential oil and ethanol extract of Oregano (Origanum vulgare L.) from Armenian flora as a natural source of terpenes, flavonoids and other phytochemicals with antiradical, antioxidant, metal chelating, tyrosinase inhibitory and antibacterial activity. Curr. Pharm. Des. 2019, 25, 1809–1816. [Google Scholar] [CrossRef]
  90. Chen, Z.G.; Zhang, K.; Mo, J.Y.; Pan, A.H.; Zhou, Q. The determination of arbutin in Paederia scandens (Lour) Merr by capillary electrophoresis with amperometric detection. Chin. J. Anal. Chem. 2002, 30, 886. [Google Scholar]
  91. Li, X.-Q.; Zhang, P.-F.; Duan, W.-D.; Zhang, D.-L.; Li, C. Studies on the chemical constituents from flowers of Paulownia Fortunei. J. Chin. Med. Mater. 2009, 32, 1227–1229. [Google Scholar]
  92. Zhang, Y.; Guo, F.; Zeng, P.; Jia, Q.; Li, Y.; Zhu, W.; Chen, K. Phenolic components from Petasites Tricholobus. China J. Chin. Mater. Med. 2012, 37, 1782–1787. [Google Scholar]
  93. Kang, H. Peroxynitrite scavengers from Phellinus linteus. Nat. Prod. Sci. 2008, 14, 1–11. [Google Scholar]
  94. Yan, C.; Wang, Y.; Hao, X. Water-soluble chemical constituents from fruits of Phellodendron chinense var. glabriusculum. China J. Chin. Mater. Med. 2009, 34, 2895–2897. [Google Scholar]
  95. Yoshimura, M.; Ochi, K.; Sekiya, H.; Tamai, E.; Maki, J.; Tada, A.; Sugimoto, N.; Akiyama, H.; Amakura, Y. Identification of characteristic phenolic constituents in Mousouchiku extract used as food additives. Chem. Pharm. Bull. 2017, 65, 878–882. [Google Scholar] [CrossRef] [Green Version]
  96. Yin, L.Z.; Ouyang, P.; Xu, X.; Zhou, L.G.; Wang, D.C.; Deng, X.M. Isolation and identification of a new compounds from the roots of Picrorhiza Scrophulariiflora. Chem. J. Chin. Univ. Chin. 2010, 31, 84–87. [Google Scholar]
  97. Ma, X.T.; Shao, S.; Xiao, F.Q.; Zhang, H.Y.; Zhang, R.R.; Wang, M.; Li, G.Z.; Yan, M.M. Platycodon grandiflorum extract: Chemical composition and whitening, antioxidant, and anti-inflammatory effects. RSC Adv. 2021, 11, 10814–10826. [Google Scholar] [CrossRef]
  98. Acikara, O.B.; Ilhan, M.; Kurtul, E.; Smejkal, K.; Akkol, E.K. Inhibitory activity of Podospermum canum and its active components on collagenase, elastase and hyaluronidase enzymes. Bioorganic Chem. 2019, 93, 103330. [Google Scholar] [CrossRef]
  99. Rao, M.R.K.; Lakshmi, N.V. Preliminary phytochemical and GC-MS analysis of different extracts of Psophocarpus tetragonolobus leaves. Indo Am. J. Pharm. Sci. 2018, 5, 1649–1656. [Google Scholar]
  100. Zhang, D.-K. Quantitative determination of arbutin and gallotannin in Pyrola calliantha. Bull. Chin. Mater. Med. 1987, 12, 45–46. [Google Scholar]
  101. Zhang, D.Y.; Yao, X.H.; Duan, M.H.; Luo, M.; Zhao, C.J.; Zu, Y.G.; Fu, Y.J. An effective homogenate-assisted negative pressure cavitation extraction for the determination of phenolic compounds in Pyrola by LC-MS/MS and the evaluation of its antioxidant activity. Food Funct. 2015, 6, 3323–3333. [Google Scholar] [CrossRef] [PubMed]
  102. Bulduk, I.; Sahin, M.D.; Sanli, S. Arbutin analysis in leaves, fruits and branches of Pyrus anatolica, method optimization. Eurasian J. Anal. Chem. 2016, 11, 233–244. [Google Scholar]
  103. Mir, H.; Komi, D.E.A.; Pouramir, M.; Parsian, H.; Moghadamnia, A.A.; Seyfizadeh, N.; Lakzaei, M. The hepatoprotective effects of Pyrus biossieriana Buhse. leaf extract on tert-butyl hydroperoxide toxicity in HepG2 cell line. BMC Res. Notes 2021, 14, 298. [Google Scholar] [CrossRef] [PubMed]
  104. Dong, X.G.; Zheng, Y.; Cao, Y.F.; Tian, L.M.; Zhang, Y.; Qi, D.; Huo, H.L.; Wang, D.J. Evaluation of phenolic composition and content of pear varieties in leaves from China. Erwerbs Obstbau 2018, 60, 331–340. [Google Scholar] [CrossRef]
  105. Bilia, A.R.; Rubio, M.D.E.; Alvarez, M.L.; Morelli, I.; Gonzalez, J.M. New benzyl alcohol glycosides from Pyrus bourgaeana. Planta Med. 1994, 60, 569–571. [Google Scholar] [CrossRef] [PubMed]
  106. Friedrich, H. Studies on phenolic components of Pyrus communis. II. Arbutin content of pear leaves. Pharmazie 1957, 12, 831–834. [Google Scholar] [PubMed]
  107. Salta, J.; Martins, A.; Santos, R.G.; Neng, N.R.; Nogueira, J.M.P.; Justino, J.; Rauter, A.P. Phenolic composition and antioxidant activity of Rocha pear and other pear cultivars—A comparative study. J. Funct. Foods 2010, 2, 153–157. [Google Scholar] [CrossRef]
  108. Friedrich, H. Studies on phenolic constituents of Pyrus communis. IV. Content of arbutin in germinating pear seeds and distribution in young plants. Pharmazie 1958, 13, 153–155. [Google Scholar]
  109. Rychlinska, I.; Gudej, J. Qualitative and quantitative chromatographic investigation of hydroquinone derivatives in Pyrus communis L. flowers. Acta Pol. Pharm. 2003, 60, 309–312. [Google Scholar]
  110. Liu, J.-K.; Zuo, C.-X. Studies of the chemical constituents of Pyrus Communis. Acta Bot. Sin. 1987, 29, 84–87. [Google Scholar]
  111. Zhou, S.; Feng, Y.X.; Zhao, Z.; Cheng, Y.D.; Guan, J.F. The involvement of phenolic metabolism in superficial scald development in ‘Wujiuxiang’ pear. J. Appl. Bot. Food Qual. 2020, 93, 20–25. [Google Scholar]
  112. Kavac, D.D.; Kececi, S. Extraction of phenolic antioxidants from Pyrus elaeagrifolia Pallas: Process optimization, investigation of the bioactivity and beta-glucuronidase inhibitory potential. J. Food Meas. Charact. 2019, 13, 2894–2902. [Google Scholar] [CrossRef]
  113. Om, P.; Gopinath, M.S.; Kumar, P.M.; Kumar, S.P.M.; Kudachikar, V.B. Ethanolic extract of Pyrus pashia Buch ham ex Don (Kainth): A bioaccessible source of polyphenolics with anti-inflammatory activity in vitro and in vivo. J. Ethnopharmacol. 2022, 2982, 114628. [Google Scholar] [CrossRef]
  114. Fuertes-Lasala, E.; Fernandez, M.; Martinez, L.; Garcia-Mina, M.C.; Vega, F.A. Phenolic compounds in Pyrus Pyraster. An. Inst. Bot. A. J. Cavinilles 1975, 32, 245–267. [Google Scholar]
  115. Usjak, L.J.; Milutinovic, V.M.; Crnogorac, M.J.D.; Stanojkovic, T.P.; Niketic, M.S.; Kukic-Markovic, J.M.; Petrovic, S.D. Barks of three wild Pyrus taxa: Phenolic constituents, antioxidant activity and in vitro and in silico investigations of alpha-amylase and alpha-glucosidase inhibition. Chem. Biodivers. 2021, 18, e2100446. [Google Scholar] [CrossRef]
  116. Jiang, G.H.; Lee, K.C.; Ameer, K.; Eun, J.B. Comparison of freeze-drying and hot air-drying on Asian pear (Pyrus pyrifolia Nakai Niitaka) powder: Changes in bioaccessibility, antioxidant activity, and bioactive and volatile compounds. J. Food Sci. Technol. Mysore 2019, 56, 2836–2844. [Google Scholar] [CrossRef]
  117. Eun, J.-B. Changes in phenolic substances and pectin according to the growth period of the pear. Korean J. Food Sci. Technol. 2007, 39, 7–13. [Google Scholar]
  118. Lee, K.H.; Cho, J.Y.; Lee, H.J.; Park, K.Y.; Ma, Y.K.; Lee, S.H.; Cho, J.A.; Kim, W.S.; Park, K.H.; Moon, J.H. Isolation and identification of phenolic compounds from an Asian pear (Pyrus pyrifolia Nakai) fruit peel. Food Sci. Biotechnol. 2011, 20, 1539–1545. [Google Scholar] [CrossRef]
  119. Sasaki, C.; Ichitani, M.; Kunimoto, K.K.; Asada, C.; Nakamura, Y. Extraction of arbutin and its comparative content in branches, leaves, stems and fruits of Japanese pear Pyrys cv. Kousui. Biosci. Biotechnol. Biochem. 2014, 78, 874–877. [Google Scholar] [CrossRef]
  120. Sugawara, T.; Igarashi, K. Variation in polyphenol components and radical scavenging activity of Japanese pear (Pyrus serotina Rehder var. culta Rehder) during fruit maturation. J. Jpn. Soc. Food Sci. Technol. 2013, 60, 516–520. [Google Scholar] [CrossRef]
  121. Challice, J.S. Phenolic compounds of genus Pyrus. 6. Distribution of phenols among various tissues of Pyrus stem. J. Sci. Food Agric. 1973, 24, 285–293. [Google Scholar] [CrossRef]
  122. Khoruzhaya, T.G.; Krasnov, E.A. Phenol compounds of Rhodiola Coccinea. Khimiya Prir. Soedin. 1972, 677–678. [Google Scholar]
  123. Wang, Y.S.; Zhou, S.S.; Shen, C.Y.; Jiang, J.G. Isolation and identification of four antioxidants from Rhodiola crenulata and evaluation of their UV photoprotection capacity in vitro. J. Funct. Foods 2020, 66, 103825. [Google Scholar] [CrossRef]
  124. Zhou, W.; Chen, K.; Liu, Q.; Luo, Y.; Zhang, C.; Zheng, Y.; Zhuo, Z.; Guo, K.; Wang, J.; Chen, H.; et al. The protective effect of Rhodiola rosea on radiation induced intestinal injury. Chem. Biodivers. 2020, 17, e2000652. [Google Scholar] [CrossRef] [PubMed]
  125. Zhavoronkova, M.E.; Belousov, M.V.; Fursa, N.S. Arbutin levels in the leaves of several species of the genus Rhododendron. I.P. Pavlov. Russ. Med. Biol. Her. 2008, 16, 91–94. [Google Scholar]
  126. Wang, R.M.; He, R.P.; Li, Z.H.; Lin, X.; Wang, L. HPLC-Q-Orbitrap-MS/MS phenolic profiles and biological activities of extracts from roxburgh rose (Rosa roxburghii Tratt.) leaves. Arab. J. Chem. 2021, 14, 103257. [Google Scholar] [CrossRef]
  127. Iqbal, K.; Malik, A.; Mehmood, A.; Mukhtar, N.; Tareen, R.B. Phytochemical studies of Salix acmophylla Boiss. J. Chem. Soc. Pak. 2004, 26, 392–394. [Google Scholar]
  128. De Falco, B.; Grauso, L.; Fiore, A.; Bochicchio, R.; Amato, M.; Lanzotti, V. Metabolomic analysis and antioxidant activity of wild type and mutant chia (Salvia hispanica L.) stem and flower grown under different irrigation regimes. J. Sci. Food Agric. 2021, 101, 6010–6019. [Google Scholar] [CrossRef]
  129. Frontana-Uribe, B.A.; Escarcega-Bobadilla, M.V.; Estrada-Reyes, R.; Morales-Serna, J.A.; Salmon, M.; Cardenas, J. A new languidulane diterpenenoid from Salvia mexicana var. Mexicana. Mol. 2011, 16, 8866–8873. [Google Scholar]
  130. Natic, M.; Pavlovic, A.; Lo Bosco, F.; Stanisavljevic, N.; Zagorac, D.D.; Aksic, M.F.; Papetti, A. Nutraceutical properties and phytochemical characterization of wild Serbian fruits. Eur. Food Res. Technol. 2019, 245, 469–478. [Google Scholar] [CrossRef]
  131. Taneyama, M.; Yoshida, S. Studies on C-glycosides in higher plants. 2. Incorporation of glucose C-14 into bergenin and arbutin in Saxifraga stolonifera. Bot. Mag. Tokyo 1979, 92, 69–73. [Google Scholar] [CrossRef]
  132. Sun, X.-H.; Shen, G.-M.; Tian, X. Chemical components of Scrofella chinensis (I). Xibei Zhiwu Xue Bao 2006, 26, 412–415. [Google Scholar]
  133. Pokotylo, I.V.; Gumenyuk, L.A.; Dykhavov, N.N. Phenol compounds from Sedum Purpureum. Khimiya Prir. Soedin. 1974, 2, 252–253. [Google Scholar]
  134. Krasnov, E.A.; Petrova, L.V. Arbutin in certain plants of Sedum genus. Khimiya Prir. Soedin. 1970, 4, 476. [Google Scholar]
  135. Zheng, X.-K.; Bi, Y.-F.; Feng, W.-S.; Shi, S.-P.; Wang, J.-F.; Niu, J.-Z. Study on the chemical constituents of Selaginella tamariscina (Beauv.) Spring. Yao Xue Xue Bao 2004, 39, 266–268. [Google Scholar] [PubMed]
  136. Myagchilov, A.V.; Mineev, S.A.; Sokolova, L.I.; Gardasova, E.D.; Gorovoi, P.G. Arbutin content in the far-eastern species Serratula Komaroviilljin. Pharm. Chem. J. 2020, 54, 377–379. [Google Scholar] [CrossRef]
  137. Nycz, J.E.; Malecki, G.; Morag, M.; Nowak, G.; Ponikiewski, L.; Kusz, J.; Switlicka, A. Arbutin: Isolation, X-ray structure and computational studies. J. Mol. Struct. 2010, 980, 13–17. [Google Scholar] [CrossRef]
  138. Zatsny, I.L.; Gorovits, M.B.; Abubakir, N.K. Arbutin from Serratula Sogdiana. Chem. Nat. Compd. 1973, 9, 415. [Google Scholar] [CrossRef]
  139. Katsutani, K.; Sugimoto, S.; Yamano, Y.; Otsuka, H.; Matsunami, K.; Mizuta, T. Eudesmane-type sesquiterpene glycosides: Sonneratiosides A-E and eudesmol beta-D-glucopyranoside from the leaves of Sonneratia alba. J. Nat. Med. 2020, 74, 119–126. [Google Scholar] [CrossRef]
  140. Wang, J.; Ma, Y.-M.; Yan, M.-R.; Xu, Q.; Qu, Z.-R.; Miao, Z. Chemical composition of stems and branches of Sorboria arborea. J. Chin. Med. Mater. 2015, 38, 2098–2101. [Google Scholar]
  141. Venditti, A.; Frezza, C.; Lorenzetti, L.M.; Maggi, F.; Serafini, M.; Bianco, A. Reassessment of the polar fraction of Stachys alopecuros (L.) Benth. subsp. divulsa (Ten.) Grande (Lamiaceae) from the Monti Sibillini National Park: A potential source of bioactive compounds. J. Intercult. Ethnopharmacol. 2017, 6, 144–153. [Google Scholar] [CrossRef] [PubMed]
  142. Venditti, A.; Serrilli, A.M.; Di Cecco, M.; Ciaschetti, G.; Andrisano, T.; Bianca, A. Phytochemical composition of polar fraction of Stachys gemanica L. subsp. Salviifolia (Ten.) Gams., a typical plant of Majella National Park. Nat. Prod. Res. 2013, 27, 190–193. [Google Scholar] [CrossRef] [PubMed]
  143. Tundis, R.; Bonesi, M.; Pugliese, A.; Nadjafi, F.; Menchini, F.; Loizzo, M.R. Tyrosinase, acetyl- and butyryl-cholinesterase inhibitory activity of Stachys lavandulifolia Vahl (Lamiaceae) and its major constituents. Rec. Nat. Prod. 2015, 9, 81–93. [Google Scholar]
  144. Frezza, C.; Venditti, A.; Matrone, G.; Serafini, I.; Foddai, S.; Bianco, A.; Serafini, M. Iridoid glycosides and polyphenolic compounds from Teucrium chamaedrys L. Nat. Prod. Res. 2018, 32, 1583–1589. [Google Scholar] [CrossRef] [PubMed]
  145. Avelino-Flores, M.D.; Cruz-Lopez, M.D.; Jimenez-Montejo, F.E.; Reyes-Leyva, J. Cytotoxic activity of the methanolic extract of Turnea diffusa Willd. on breast cancer cells. J. Med. Food. 2015, 18, 299–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Mzhavanadze, V.V.; Targamadze, I.L.; Dranik, L.I. Phenol compounds from the leaves of Vaccinium Arctostaphylos. Khim. Prir. Soedin. 1972, 8, 124. [Google Scholar]
  147. Gao, S.H.; Zhao, T.R.; Liu, Y.P.; Wang, Y.F.; Cheng, G.G.; Cao, J.X. Phenolic constituents, antioxidant activity and neuroprotective effects of ethanol extracts of fruits, leaves and flower buds from Vaccinium dunalianum Wight. Food Chem. 2021, 374, 131752. [Google Scholar] [CrossRef]
  148. Friedrich, H.; Schonert, J. Phytochemical investigation of leaves and fruits of Vaccinium Myrtillus. Planta Med. 1973, 24, 90–100. [Google Scholar] [CrossRef]
  149. Kocik, H.; Wojciechowska, B.; Filec, J. Analysis of flavonoids and phenol compounds of Vaccinium Myrtillus. Prod. Nauk. Uniw. Slaskiego W. Katowicach 1980, 38–48. [Google Scholar]
  150. Askari, A.; Worthen, L.R. Isolation of isopyroside from Vaccinium Vacillans. Phytochemistry 1971, 11, 1509–1510. [Google Scholar] [CrossRef]
  151. Bandzaitene, Z.Y.U.; Butkus, V.F. Biological and biochemical characteristics of cowberry. Part 4. Content of some organic substances in the leaves and berries. Liet. TSR Moksl. Akad. Darb. Ser. C Biol. Moksl. 1975, 13–21. [Google Scholar]
  152. Pyka, A.; Bober, K.; Strolarczyk, A. Densitometric determination of arbutin in cowberry leaves (Vaccinium vitis idaeae). Acta Pol. Pharm. 2007, 64, 395–400. [Google Scholar] [PubMed]
  153. Sun, H.; Wang, X.; Huang, R.; Yuan, C. Determination of arbutin in the herbs of Vaccinium vitis-idaea L. by RP-HPLC. Zhongguo Zhongyao Zazhi 1997, 22, 555–577. [Google Scholar] [PubMed]
  154. Dimitrova, P.A.; Alipieva, K.; Grozdanova, T.; Leseva, M.; Gerginova, D.; Simova, S.; Marchev, A.S.; Bankova, V.; Georgiev, M.I.; Popova, M.P. Veronica austriaca L. extract and arbutin mature double TNF-alpha/IFN-gamma neutrophils in murine bone marrow pool. Molecules 2020, 25, 3410. [Google Scholar] [CrossRef] [PubMed]
  155. Kostadinova, E.P.; Alipieva, K.I.; Kokubun, T.; Taskova, R.M.; Handjieva, N.V. Phenylethanoids, iridoids and a spirostanol saponin from Veronica turrilliana. Phytochemistry 2007, 68, 1321–1326. [Google Scholar] [CrossRef]
  156. Shao, J.H.; Chen, J.; Zhao, C.C.; Shen, J.; Liu, W.Y.; Gu, W.Y.; Li, K.H. Insecticidal and alpha-glucosidase inhibitory activities of chemical constituents from Viburnum fordiae Hance. Nat. Prod. Res. 2019, 33, 2662–2667. [Google Scholar]
  157. Petricic, J.; Stanic, G.; Holic, L. Flavonoids saponins tannins and arbutin as constituents of leaves of Viburnum tinus, Viburnum opulus and Viburnum lanata. Acta Pharm. Jugosl. 1980, 30, 97–102. [Google Scholar]
  158. Takido, M.; Fukuhara, K.; Yamanouchi, S.; Takahashi, S. Phlebotrichin, a phenolic compound from the fresh leaves of Viburnum phlebotrichum. Phytochemistry 1983, 22, 223–225. [Google Scholar] [CrossRef]
  159. Bubenchicov, R.A.; Goncharov, N.F. The study of the composition of the phenolic compounds of Viola Arvensis. Khimiko-Farmatsevticheskii Zhurnal 2005, 39, 31–32. [Google Scholar]
  160. Jensen, S.R.; Gotfredsen, C.H.; Zidorn, C. Iridoids and phenylethanoids in Lagotis integrifolia and Wulfeniopsis amherstiana (Plantaginaceae). Biochem. Syst. Ecol. 2009, 37, 421–425. [Google Scholar] [CrossRef]
  161. Hisatomi, E.; Matsui, M.; Kobayashi, A.; Kubota, K. Antioxidative activity in the pericarp and sees of Japanese pepper (Xanthoxylum piperitum DC). J. Agric. Food Chem. 2000, 48, 4924–4928. [Google Scholar] [CrossRef] [PubMed]
  162. Xiong, Q.B.; Shi, D.W.; Mizuno, M. Flavonol glucosides in pericarps of Zanthoxylum Bungeanum. Phytochem. 1995, 39, 723–725. [Google Scholar] [CrossRef]
  163. Grisdale, S.K.; Towers, G.H.N. Biosynthesis of arbutin from some phenylpropanoid compounds in Pyrus Communis. Nat. 1960, 188, 1130–1131. [Google Scholar] [CrossRef]
  164. Xu, K.-X.; Xue, M.-G.; Li, Z.; Ye, B.-C.; Zhang, B. Recent progress on feasible strategies for arbutin production. Front. Bioeng. Biotechnol. 2022, 10, 914280. [Google Scholar] [CrossRef]
  165. Shen, X.; Wang, J.; Wang, J.; Chen, Z.; Yuan, Q.; Yan, Y. High-level de novo biosynthesis of arbutin in engineered Escherichia coli. Metab. Eng. 2017, 42, 52–58. [Google Scholar] [CrossRef]
  166. Li, H.; Jeong, Y.M.; Kim, S.Y.; Kim, M.K.; Kim, D.S. Arbutin inhibits TCCSUP human bladder cancer cell proliferation via up-regulation of p21. Pharmazie 2011, 66, 306–309. [Google Scholar]
  167. Yang, Z.K.; Shi, H.Y.; Chinnathambi, A.; Salmen, S.H.; Alharbi, S.A.; Veeraraghavan, V.P.; Surapaneni, K.M.; Arulselvan, P. Arbutin exerts anticancer activity against rat C6 glioma cells by inducing apoptosis and inhibiting the inflammatory markers and P13/Akt/mTOR cascade. J. Biochem. Mol. Toxicol. 2021, 35, e22857. [Google Scholar]
  168. Erenler, R.; Sen, O.; Aksit, H.; Demirtas, I.; Yaglioglu, A.S.; Elmastas, M.; Telci, I. Isolation and identification of chemical constituents from origanum majorana and investigation of antiproliferative and antioxidant activities. J. Sci. Food Agric. 2016, 96, 822–836. [Google Scholar] [CrossRef]
  169. Hazman, O.; Sariova, A.; Bozkurt, M.F.; Cigerci, I.H. The anticarcinogen activity of beta-arbutin on MSC-7 cells: Stimulation of apoptosis through estrogen receptors-alpha signal pathway, inflammation and genotoxicity. Mol. Cell. Biochem. 2021, 476, 349–360. [Google Scholar] [CrossRef]
  170. Berdowska, I.; Zielinski, B.; Fecka, I.; Kulbacka, J.; Saczko, J.; Gamian, A. Cytotoxic impact of phenolics from Lamiaceae species on human breast cancer cells. Food Chem. 2013, 141, 1313–1321. [Google Scholar] [CrossRef]
  171. Kamei, H.; Kojima, T.; Hashimoto, Y.; Hasegawa, M. Inhibition of cells growth in culture by quinones. Cancer Biother. Radiopharm. 1998, 13, 185–188. [Google Scholar] [CrossRef] [PubMed]
  172. Zeng, X.T.; Liu, H.P.; Huang, Z.P.; Dong, P.; Chen, X. Anticancer effect of arbutin on diethylnitrosamine-induced liver carcinoma in rats via the GRP and GADD pathway. J. Environ. Pathol. Toxicol. Oncol. 2022, 41, 15–26. [Google Scholar] [CrossRef] [PubMed]
  173. Williams, G.M.; Latropoulos, M.; Jeffrey, A.M.; Duan, J.D. Inhibition by dietary hydroquinone of acetylaminofluorene induction of initiation of rat carcinogenesis. Food Chem. Toxicol. 2007, 45, 1620–1625. [Google Scholar] [CrossRef] [PubMed]
  174. Hazman, O.; Evin, H.; Bozkurt, M.F.; Cigerci, I.H. Two faces of arbutin in hepatocellular carcinoma (HepG2) cells: Anticarcinogenic effect in high concentration and protective effect against cisplatin toxicity through its antioxidant and anti-inflammatory activity in low concentration. Biologia 2022, 77, 225–239. [Google Scholar] [CrossRef]
  175. Kang, M.; Ha, H.W.; Kim, H.G.; Lee, D.H.; Kong, M.; Ahn, Y.T.; Kim, D.H.; Shin, B.S.; Kang, W.; Jeong, H.G. Role of metabolism by intestinal bacteria in arbutin-induced toxicity in vitro. Arch. Pharmacal Res. 2011, 34, 687–693. [Google Scholar] [CrossRef] [PubMed]
  176. Jiang, L.Y.; Wang, D.; Zhang, Y.F.; Li, J.Y.; Wu, Z.P.; Wang, Z.; Wang, D. Investigation of the pro-apoptotic effects of arbutin and its acylated derivatives on murine melanoma cells. Int. J. Mol. Med. 2021, 41, 1048–1054. [Google Scholar]
  177. Su, Y.B.; Sun, X.W.; Wu, R.X.; Zhang, X.; Tu, Y.Z. Molecular spectroscopic behaviors of beta-arbutin in anti-skin cancer. Spectrosc. Lett. 2020, 53, 172–183. [Google Scholar] [CrossRef]
  178. Wang, C.Q.; Wang, X.M.; Li, B.L.; Zhang, Y.M.; Wang, L. Arbutin suppresses osteosarcoma progression via miR-338-3p/MTHFD1L and inactivation of the AKT/mTOR pathway. FEBS Open Bio 2021, 11, 289–299. [Google Scholar] [CrossRef]
  179. Safari, H.; Zabihi, E.; Pouramir, M.; Morakabati, P.; Abedian, Z.; Karkhah, A.; Nouri, H.R. Decrease of intracellular ROS by arbutin is associated with apoptosis induction and downregulation of IL-1 beat and TNF-alpha in LNCaP; prostate cancer. J. Food Biochem. 2020, 44, e13360. [Google Scholar] [CrossRef]
  180. Ebadollahi, S.H.; Pouramir, M.; Zabihi, E.; Golpour, M.; Aghajanpour-Mir, M. The effect of arbutin on the expression of tumor suppressor P53, BAX/BCL-2 ratio and oxidative stress induced by tert-butyl hydroperoxide in fibroblast and LNCap cell lines. Cell J. 2021, 22, 532–541. [Google Scholar]
  181. Mirzaei, S.; Zarrabi, A.; Asnaf, S.E.; Hashemi, F.; Zabolian, A.; Hushmandi, K.; Raej, M.; Goharrizi, M.A.S.B.; Makyandi, P.; Samarghandian, S.; et al. The role of microRNA-338-3p in cancer: Growth, invasion, chemoresistance, and mediators. Life Sci. 2021, 268, 119005. [Google Scholar] [CrossRef] [PubMed]
  182. National Health Service (NHS). Overview—Bladder Cancer. Available online: https://www.nhs.uk/conditions/bladder-cancer/ (accessed on 8 October 2022).
  183. Gartel, A. p21(WAF1/CIP1) and cancer: A shifting paradigm? Biofactors 2009, 35, 161–164. [Google Scholar] [CrossRef] [PubMed]
  184. Lee, J.; Lim, J.W.; Kim, H. Astaxanthin inhibits matrix metalloproteinase expression by suppressing PI3K/AKT/mTOR activation in Helicobacter pylori-infected gastric epithelial cells. Nutrients 2022, 14, 3427. [Google Scholar] [CrossRef] [PubMed]
  185. Chicoine, M.R.; Silbergeld, D.L. Invading C6 glioma cells maintaining tumorigenicity. J. Neurosurg. 1995, 83, 665–671. [Google Scholar] [CrossRef]
  186. National Health Service (NHS). Overview—Brain Tumour. Available online: https://www.nhs.uk/conditions/brain-tumours/ (accessed on 7 October 2022).
  187. National Health Service (NHS). Overview—Breast Cancer. Available online: https://www.nhs.uk/conditions/breast-cancer/ (accessed on 7 October 2022).
  188. Yu, S.; Kim, T.; Yoo, K.H.; Kang, K. The T47D cell line is an ideal experimental model to elucidate the progesterone-specific effect of a luminal A subtype of breast cancer. Biochem. Biophys. Res. Commun. 2017, 486, 752–758. [Google Scholar] [CrossRef] [PubMed]
  189. National Health Service (NHS). Overview—Cervical Cancer. Available online: https://www.nhs.uk/conditions/cervical-cancer/ (accessed on 9 October 2022).
  190. National Health Service (NHS). Overview—Bowel Cancer. Available online: https://www.nhs.uk/conditions/bowel-cancer/ (accessed on 9 October 2022).
  191. National Health Service (NHS). Overview—Stomach Cancer. Available online: https://www.nhs.uk/conditions/stomach-cancer/ (accessed on 9 October 2022).
  192. World Cancer Research Fund International. Liver Cancer Statistics. Available online: https://www.wcrf.org/cancer-trends/liver-cancer-statistics/ (accessed on 9 October 2022).
  193. National Health Service (NHS). Overview—Skin Cancer (Melanoma). Available online: https://www.nhs.uk/conditions/melanoma-skin-cancer/ (accessed on 13 October 2022).
  194. American Cancer Society. About Osteosarcoma. Available online: https://www.cancer.org/cancer/osteosarcoma/about/what-is-osteosarcoma.html (accessed on 13 October 2022).
  195. National Health Service (NHS). Overview—Bone Cancer (Melanoma). Available online: https://www.nhs.uk/conditions/bone-cancer/ (accessed on 13 October 2022).
  196. Prostate Cancer UK. Available online: https://prostatecanceruk.org/prostate-information/about-prostate-cancer (accessed on 14 October 2022).
  197. Novak, J. Arbutin—A risk substance in herbs? J. Med. Spice Plants 2010, 15, 170–173. [Google Scholar]
  198. Cheng, S.L.; Liu, R.H.; Sheu, J.N.; Chen, S.T.; Sinchaikul, S.; Tsay, G.T. Toxicogenomics of A375 human malignant melanoma cells treated with arbutin. J. Biomed. Sci. 2007, 14, 87–105. [Google Scholar] [CrossRef] [PubMed]
  199. O’Donoghue, J.L. Hydroquinone and its analogues in dermatology—A risk-benefit viewpoint. J. Cosmet. Dermatol. 2006, 5, 196–203. [Google Scholar] [CrossRef]
  200. De Arriba, S.G.; Naser, B.; Nolte, K.-U. Risk assessment of free hydroquinone derived from Arctostaphylos Uva-ursi folium herbal preparations. Int. J. Toxicol. 2013, 32, 442–453. [Google Scholar] [CrossRef]
  201. Drug Bank Online. Arbutin. Available online: https://go.drugbank.com/drugs/DB11217 (accessed on 1 November 2022).
  202. Seyfizadeh, N.; Mahjoub, S.; Zabihi, E.; Moghadamnia, A.; Pouramir, A.; Mir, H.; Khosravifarsani, M.; Elahimanesh, F. Cytoprotective effects of arbutin against tert-butyl hydroperoxide induced toxicity in Hep-G2 cell line. World Appl. Sci. J. 2012, 19, 163–167. [Google Scholar]
  203. Pecivova, J.; Nosal, R.; Svitekova, K. Arbutin and decrease of potentially toxic substances generated in human blood neutrophils. Interdiscip. Toxicol. 2014, 7, 195–200. [Google Scholar] [CrossRef] [PubMed]
  204. Khadir, F.; Pouramir, M.; Joorsaraie, G.; Feizi, F.; Sorkhi, H.; Yousefi, F. A study of arbutin protective effect on cyclosporin A induced oxidative damage. Casp. J. Intern. Med. 2015, 6, 196–200. [Google Scholar]
Figure 1. Arbutin (1).
Figure 1. Arbutin (1).
Molecules 27 08786 g001
Scheme 1. Biosynthesis of arbutin [164,165].
Scheme 1. Biosynthesis of arbutin [164,165].
Molecules 27 08786 sch001
Figure 2. A schematic summary of the anticancer potential of arbutin, obtained from different plant families.
Figure 2. A schematic summary of the anticancer potential of arbutin, obtained from different plant families.
Molecules 27 08786 g002
Table 1. Distribution of arbutin (1) in the plant kingdom.
Table 1. Distribution of arbutin (1) in the plant kingdom.
SpeciesFamilyCommon NamePlant PartGeographical SourceReference
Aesculus californica Nutt.HippocastanaceaeCalifornia buckeyeFruit endospermUSA[4]
Afgekia mahidolae B.L. Burtt & Chermsir.FabaceaeKan Pai MahidolLeavesThailand[5]
Ailanthus altissima (Mill.) SwingleSimaroubaceaeVarnish treeFruitsChina[6]
Ainsliaea bonatii BeauverdAsteraceaeChinese daisyLeavesChina[7]
Amaranthus spp. AmaranthaceaeAmaranthLeavesBangladesh[8]
Amaranthus tricolor L.AmaranthaceaeAmaranthLeavesRussia[9]
Antidesma thwaitesianum Muell. Arg.PhyllanthaceaeMao treeFruits and leavesThailand[10]
Arbutus andrachne L.EricaceaeGreek strawberry treeLeavesGreece and Turkey[11]
Arbutus pavarii Pamp.EricaceaeLibyan strawberry treeLeavesLibya[12]
Arbutus unedo L.EricaceaeStrawberry treeLeavesMediterranean region and western Europe[1,13]
Fruits [14]
Arctostaphylos pungens Kunth.EricaceaePoint leaf manzanitaLeavesItaly, Mexico and USA[15]
Arctostaphylos spp. EricaceaeBearberryLeavesScotland and Scandinavia[16,17]
Arctostaphylos uva-ursi (L.) Spreng.EricaceaeBearberryLeavesBulgaria, Turkey[18]
Arctous alpina (L.) Nied.EricaceaeAlpine bearberryLeavesRussia[19]
Artemisia pallens Wall. Ex. DC.AsteraceaeDamanakaLeavesIndia[20]
Artocarpus lacucha L.MoraceaeMonkey fruitLeavesSouth-east Asia[21]
Astilbe rivularis L.SaxifragaceaeFalse spireaLeavesNepal and UK[22]
Atriplex littoralis L.AmaranthaceaeGrass leaf oracheAerial partsSerbia[23]
Bacopa procumbens (Mill.) Greenm.PlantaginaceaeBaby jump-upAerial partsTropical and subtropical areas of North and South America[24]
Bellendena montana R. Br.ProteaceaeMountain rocketLeavesTasmania[25]
Benincasa hispida (Thunb.) Cogn.CucurbitaceaeWax gourdFruitsChina[26]
Bergenia ciliata (Haw.) Sternb.SaxifragaceaeFringed elephant’s earsRhizomeNepal[27]
Bergenia cordifolia L.SaxifragaceaeHeartleaf BergeniaLeavesRussia[28]
Bergenia crassifolia (L.) Fritsch.SaxifragaceaeHeart-leaved BergeniaAerial partsRussia[29]
LeavesRussia[30]
LeavesRomania[31]
Bergenia purpurascens (Hook. f. & Thomson) Engl.SaxifragaceaePurple BergeniaLeavesChina[32]
Bergenia spp. SaxifragaceaeElephant’s earsAerial partsAfghanistan to China and the Himalayan region[17,33,34,35]
Bergenia stracheyi (Hook. F. & Thoms.) Engl.SaxifragaceaeElephant’s earsAerial partsThe Himalayas[36]
Betula pendula Roth.BetulaceaeSilver birchLeavesEurope and Asia[37]
Betula platyphylla Sukatchev var. japonica HaraBetulaceaeShirakambaLeavesChina[38]
Betula schmidtii Regel.BetulaceaeSchmidt’s birchBarkChina, Japan, Korea and Russia[39]
Breynia officinalis Hemsl.PhyllanthaceaeChi R YunLeavesChina and Japan[40]
Breynia rostrata Merr.PhyllanthaceaeHui Guo Hei Mian ShenAerial partsChina and Vietnam[41]
Calluna spp. EricaceaeHeatherLeavesEurope and Asia Minor[17]
Calluna vulgaris L. Hull.EricaceaeHeatherAerial partsAsia Minor[42]
LeavesRussia[43]
Careya arborea Roxb.LecythidaceaeSlow match treeBark, leaves and seedsIndia[44]
Casearia multinervosa C.T.White & SleumerSalicaceaeCaseariaStemAustralia[45]
Cenarrhenes nitida R. Br.ProteaceaePort Arthur plumLeavesTasmania[25]
Centaurea urvillei DC. subsp. urvilleiAsteraceaeStar thistleLeavesTurkey[46]
Chamaecyparis lawsonianaCupressaceaeLawson cypressGallsIran[47]
Clausena indica (Datz.) OliverRutaceaeIndian wampiFruit pericarpIndia and Sri Lanka[48]
Coriandrum sativum L.ApiaceaeCorianderAerial partsWestern Asia, Southern Europe and Russia[49]
Cotoneaster simonsii BakerRosaceaeHimalayan cotoneasterAerial partsThe Himalayas[50]
Cuscuta sinensis Lam.ConvolvulaceaeChinese cuscutaSemenChina, Japan and Korea[51]
Dryopteris sublaeta Ching & Y. P. HsuDryopteridaceaeChinese male fernRhizomeChina[52]
Eriobotrya fragrans Champ. Ex. Benth.RosaceaeXiang hua pi baLeavesChina and Vietnam[53]
Eryngium bourgatii Gouan.ApiaceaeSea hollyFlowers and leavesSpain[54]
Eugenia hyemalis L. CambessMyrtaceaeHyemalisAerial partsArgentina, Bolivia and USA[55]
Flammulina velutipes (Curtis) SingerPhysalacriaceaeVelvet shankLeavesChina[56]
Fragaria spp. RosaceaeStrawberryRootsEurope, North America and China[57]
Gentiana pyrenaica L.GentianaceaePyrenian gentianLeavesUnited Kingdom[58]
Grevillea banksii R. Br.ProteaceaeDwarf silky oakLeavesAustralia[59]
Grevillea robusta A. Cunn. Ex R. Br.ProteaceaeSilk oakLeavesAustralia and India[60]
Bark and leaves [61]
Hakea saligna L.ProteaceaeHakeaLeavesAustralia and India[60]
Halocarpus biformis (Hook.) C.J. QuinnPodocarpaceaeYellow pineLeavesNew Zealand[62]
Heliciopsis lobata (Merr.) SleumerProteaceaeHeliciaLeavesChina and Vietnam[63]
Herpetospermum caudigerum Wall.CucurbitaceaeHerpetospermumLeavesChina, India and Tibet[64]
Homalium zeylanicum (Gardner) Benth.FlacourtiaceaeKalavaramLeavesIndia[65]
Huperzia serrataLycopodiaceaeToothed clubmossWhole plantChina, Japan, Korea, Russia and Tibet[66]
Ilex brasiliensis (Spreng.) Loes.AquifoliaceaeBrazilian hollyLeavesBrazil[67]
Ilex integerrima Reiss.AquifoliaceaeHollyLeavesBrazil[67]
Ilex latifolia Thunb.AquifoliaceaeTarajo hollyLeavesJapan[68]
Ilex pseudobuxus Reiss.AquifoliaceaeBrazilian hollyLeavesBrazil[67]
Ilex theezans Mart.AquifoliaceaeCongonhaLeavesBrazil[67]
Jamesia americana Torr. & A. GrayHydrangeaceaeCliffbushAerial partsUSA[69]
Juglans regia L.JuglandaceaeWalnutsNutsThe Balkans, the Himalayans and China[70]
Larix leptolepisPinaceaeJapanese LarchNeedlesJapan[71]
Lens culinaris Medik.FabaceaeLentilSeedsIndia[72]
Leucadendron spp. ProteaceaeConebushesLeavesSouth Africa[73]
Lysiloma latisiliquum (L.) Benth.FabaceaeWild tamarindLeavesUSA[74]
Madhuca latifolia (J. Konig) J.F. Macbr.SapotaceaeMahuaSeedsIndia, Nepal, Pakistan and Sri Lanka[75]
Magnifera indica L.AnacardiaceaeMangoLeavesIndia[76]
Malus sylvestris (L.) Mill.RosaceaeCrab appleLeavesUnited Kingdom & Russia[77]
Crab appleFruitsRussia[78]
Morus alba L.MoraceaeMulberryLeavesChina and India[79]
Mutisia acuminata var. acuminata Ruiz & Pav.AsteraceaeBolivian MutisiaAerial partsPeru and Bolivia[80]
Mutisia acuminata var. hirsuta (Meyen) CabreraAsteraceaeMutisiaLeavesPeru[81]
Myrsine seguinii H. Lev.Myrsinaceae alt. PrimulaceaeMyrsineLeavesChina, Japan and New Zealand[82]
Myrothamnus flabellifolia Welw.MyrothamnaceaeResurrection plantLeavesSouth Africa[83]
Aerial partsGermany[84]
Onobrychis kachetica Boiss. & BuhseFabaceaeEspartzet KakhetinskiLeavesTrans-caucasus, and Russia[85]
Onobrychis viciifolia Scop.FabaceaeSainfoinPetalsEuro Siberian temperate region[86]
Origanum dubium Boiss.LamiaceaeRouvanosAerial partsCyprus[87]
Origanum majorana L.LamiaceaeSweet majoramLeavesEgypt[88]
Origanum vulgare L.LamiaceaeOregano or wild majoramAerial partsMediterranean region[89]
Paederia scandens (Loir.) Merr.RubiaceaeGandheliAerial partsChina and India[90]
Paulownia fortune (Seem.) Hemsl.PaulowniaceaeDragon treeFlowersChina[91]
Persoonia gunnii Hook. f.ProteaceaePersooniaLeavesTasmania[25]
Petasites tricholobus Franch.AsteraceaeButterbursAerial partsChina, Nepal, Pakistan and Vietnam[92]
Phellinus linteus (Berk. & M.A. Curtis) TengHymenochaetaceaeMeshimakobuAerial partsChina, Korea and Japan[93]
Phellodendron chinense var. glabriusculum C.K. Schenid.RutaceaeCork treeAerial partsChina[94]
Phyllostachys heterocycla Mitf.PoaceaeMousouchiku or tortoise shell bambooBamboo-sheathJapan[95]
Picrorhiza scrophulariiflora Pennell.ScrophulariaceaeXizang HuhuanglianRootsChina, India and Tibet[96]
Platycodon grandiflorum L.CampanulaceaeBalloon flowerLeavesChina[97]
Podospermum canum C. A. MeyAsteraceaeKarakokAerial partsCaucasia, Iran, Iraq, Syria and Turkey[98]
Prunophora salicina Linn.RosaceaeChinese PlumFruit peelsChina and Korea[67]
Psophocarpus tetragonolobus (L.) DCFabaceaeWinged beanLeavesIndia[99]
Pyrola calliantha AndresEricaceaeWintergreenLeavesEastern Himalaya to China[100]
Pyrola incarnata Fisch.EricaceaeLu Shou ChaLeavesChina[101]
Pyrus anatolica BrowiczRosaceaeTurkish pearFruits, leaves and stemTurkey[102]
Pyrus biossieriana BuhseRosaceaeWild pearLeavesIran[103]
Pyrus bretschneideri RehderRosaceaeYa pearLeavesChina[104]
Pyrus bourgaeana Decne.RosaceaeIberian pearAerial partsIberian Peninsula and Morocco[105]
Pyrus communis L.RosaceaePear or Rocha pearLeavesCentral and eastern Europe and western Asia[106,107]
Aerial parts and seeds [108]
FlowersPoland[109]
Pyrus communis L. var. sativa (DC.)RosaceaePearTwigsChina[110]
Pyrus communis L. cv. WujiuxiangRosaceaeWujiuxiang pearFruit peelsChina[111]
Pyrus elaeagrifolia Pall.RosaceaeWild pearLeavesAlbania, Bulgaria, Romania and Turkey[112]
Pyrus pashia Buch ham ex D. DonRosaceaeKainthFruitsThe Himalayas[113]
Pyrus pyraster (L.) Burgsd.RosaceaeEuropean wild pearFruit peelsWestern Europe to the Caucasus[114,115]
Pyrus pyrifolia NakaiRosaceaeNiitaka or Asian pearFruitsJapan[104,116]
FruitsKorea[117]
Asian pearFruit peelsChina[118]
Pyrus pyrifolia cv. Kousui NakaiRosaceaeJapanese pearBranches, fruits, leaves and stemJapan[119]
Pyrus serotina Rehder. var. culta Rehdar.RosaceaeJapanese pearLeavesJapan[120]
Pyrus spinosaRosaceaeAlmond-leaved pearTwigsSiberia[115]
Pyrus spp. RosaceaePearStemCentral and eastern Europe and western Asia[121]
Pyrus ussuriensis Maxim.RosaceaeUssurian pearLeavesChina[104]
Rhodiola coccinea (Royle) Boriss.CrassulaceaeRhodiolaAerial partsCentral Asia, south-western Siberia and central China[122]
Rhodiola crenulata LLLCrassulaceaeArctic rootAerial partsChina[123]
Rhodiola rosea L.CrassulaceaeGolden rootAerial partsChina[124]
Rhododendron adamsii RehderEricaceaeSagaan daliLeavesRussia[125]
Rhododendron dauricum L.EricaceaeDauriaLeavesChina, Mongolia and Russia[125]
Rhododendron fauriei Franch. var. brachycarpumEricaceaeJapanese RhododendronLeavesJapan, Korea and Russia[125]
Rhododendron luteum SweetEricaceaeYellow azaleaLeavesPoland and Russia[125]
Rhododendron ponticum L.EricaceaeCommon rhododendronLeavesIberian Peninsula and Russia[125]
Rosa roxburghii Tratt.RosaceaeRoxburgh roseLeavesChina[126]
Salix acmophylla Boiss.SalicaceaeBrook willowAerial partsPakistan and central Asia[127]
Salvia hispanica L.LamiaceaeChiaFlowers and stemCentral America[128]
Salvia mexicana var. Mexicana L.LamiaceaeMexican sageAerial partsMexico[129]
Sambucus nigra L.AdoxaceaeElderberry or black elderFruitsSerbia[130]
Saxifraga stolonifera CurtisSaxifragaceaeCreeping sailorLeavesChina, Japan and Korea[131]
Scrofella chinensis Maxim.PlantaginaceaeScrofellaWhole plantChina[132]
Sedum purpureum L.CrassulaceaePurple spoon-leaved stonecropLeavesUnited Kingdom[133]
Sedum spp. CrassulaceaeStonecropsLeavesNorthern hemisphere[134]
Selaginella tamariscina (Beauv.) SpringSelaginellaceaeSelaginellaAerial partsChina, India, Japan, Korea, Russia and Thailand[135]
Serratula komaroviilljin L.AsteraceaeSaw-wortLeavesRussia[136]
Serratula quinquefolia M. Bieb. ex. Willd.AsteraceaeFive-leaved saw-wortLeavesPoland[137]
Serratula sogdiana (Bunge) L. MartinsAsteraceaePlumeless saw-wortLeavesEurasia[138]
Sonneratia alba Sm.LythraceaePerepatLeavesEast Africa and south-east/far east Asia[139]
Sorbaria arborea Schneid.RosaceaeFalse spireaStemChina[140]
Stachys alopecuros (L.) Benth. Subsp. divulsa (Ten.) GrandeLamiaceaeYellow betonyAerial partsItaly[141]
Stachys germanica L subsp. Salviifolia (Ten.) Gams.LamiaceaeDowny woundwortAerial partsItaly and Germany[142]
Stachys lavandulifolia Vahl.LamiaceaeWood betonyAerial partsIran[143]
Teucrium chamaedrys L.LamiaceaeWall germanderLeavesMediterranean region[144]
Turnera diffusa Willd.PassifloraceaeDamianaLeaves and stemMexico and USA[145]
Vaccinium arctostaphylos L.EricaceaeCaucasian whortleberryLeavesArmenia, Azerbaijan, Bulgaria, Georgia, Iran, Russia and Turkey[146]
Vaccinium dunalianum WightEricaceaeChinese blueberryFlower buds, fruits and leavesAssam, China South-Central, China Southeast, East Himalaya, Myanmar, Nepal, Taiwan, Tibet and Vietnam[147]
Vaccinium myrtillus L.EricaceaeEuropean blueberryLeaves and fruitsEurope[148]
Leaves and stemEurope[149]
Vaccinium vacillans Torr.EricaceaeBlueberryLeavesRhode Island[150]
Vaccinium vitis-idaea L.EricaceaeCowberryLeaves and berriesAlaska, Canada, Poland, Russia and Eurasia[151,152]
Aerial partsChina[153]
Veronica austriaca L.PlantaginaceaeBroadleaf speedwellLeavesBulgaria[154]
Veronica turrilliana Stoj. & Stef.PlantaginaceaeSpeedwellAerial partsBulgaria[155]
Viburnum fordiae HanceViburnaceaeBright red berryStemChina[156]
Viburnum opulus L.ViburnaceaeGuelder roseLeavesEurope, northern Africa and central Asia[68,157]
Viburnum phlebotrichum Siebold & Zucc.ViburnaceaeJapanese viburnumLeavesJapan[68,158]
Viola arvensis L.ViolaceaeField PansyAerial partsRussia[159]
Wulfeniopsis amherstiana (Benth.) D.Y. HongPlantaginaceaeHimalyan WulfeniaLeavesThe Himalayas[160]
Xanthoxylum piperitum DCRutaceaeSichuan pepper or Japanese pepperPericarp and seedsJapan[161]
Zanthoxylum bungeanum Maxim.RutaceaeJapanese pepper treePericarpsChina and Japan[162]
Table 2. Cytotoxicity of arbutin (1) against various cancer and tumor cell lines.
Table 2. Cytotoxicity of arbutin (1) against various cancer and tumor cell lines.
Type of Cancer/TumourBrief Description of Anticancer Activity of Arbutin (1)In Vivo/In VitroReferences
Bladder cancerInhibition of TCCSUP (anaplastic transitional cell carcinoma in the neck of the urinary bladder) bladder cancer cell proliferation.In vitro[166]
Brain tumourActivity against rat C6 glioma cells.In vitro[167,168]
Breast cancerCytotoxicity of arbutin containing methanolic extract against MDA-MB-231 and T-47D breast cancer cells.In vitro[145]
Cytotoxicity towards the MCF-7 (breast cancer) cell line.In vitro[169]
Cytotoxicity against adriamycin-resistant MCF-7 and wild-type MCF-7.In vitro[170]
Cervical cancerAntiproliferative activity against HeLa cells.In vitro[168]
Activity against human cervical carcinoma HPV-16 positive (SiHa) and HPV negative (C-33) cell lines.[145]
Colon cancerAssessed for cytotoxicity against HCT-15 cells derived from human colon carcinoma.In vitro[171]
Gastric cancerInhibition of gastric carcinoma MGC-803 cells invasion.In vitro[63]
Liver cancerAntioxidant, anti-inflammatory and anticancer activities against diethylnitrosamine-induced liver carcinoma in rats.In vivo[172]
Inhibition of DNA-reactive carcinogen acetylaminofluorene induction of initiation of rat liver carcinogenesis.In vivo[173]
Anticarcinogenic activity against hepatocellular carcinoma cells (HepG2).In vitro[169,174]
Cytotoxicity against HepG2 cells.In vitro[175]
Skin cancerPro-apoptotic activity on B16 murine melanoma cells.In vitro[176]
Action on the toxic trans-crotonaldehyde.In vitro[177]
OsteosarcomaSuppression of osteosarcoma progression.In vitro[178]
Prostate cancerInduction of apoptosis in human prostrate adenocarcinoma (LNCaP) cells.In vitro[179,180]
Cytotoxicity against the prostate cancer cell line PC3.[12]
MiscellaneousPromotion of expression of miRNA-338-3p in suppressing cancer progression.In vitro[181]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nahar, L.; Al-Groshi, A.; Kumar, A.; Sarker, S.D. Arbutin: Occurrence in Plants, and Its Potential as an Anticancer Agent. Molecules 2022, 27, 8786. https://doi.org/10.3390/molecules27248786

AMA Style

Nahar L, Al-Groshi A, Kumar A, Sarker SD. Arbutin: Occurrence in Plants, and Its Potential as an Anticancer Agent. Molecules. 2022; 27(24):8786. https://doi.org/10.3390/molecules27248786

Chicago/Turabian Style

Nahar, Lutfun, Afaf Al-Groshi, Anil Kumar, and Satyajit D. Sarker. 2022. "Arbutin: Occurrence in Plants, and Its Potential as an Anticancer Agent" Molecules 27, no. 24: 8786. https://doi.org/10.3390/molecules27248786

Article Metrics

Back to TopTop