Review article

Rhoifolin: A promising flavonoid with potent cytotoxic and anticancer properties: molecular mechanisms and therapeutic potential

Ceyda Sibel Kiliç1, Mehmet Murat Kisla2, Gülin Amasya3, Ceyda Tugba Sengel-Türk3, Zeynep Ates Alagöz2, Ayse Mine Gençler Özkan1, Ilker Ates4, Safa Gümüsok1, Jesús Herrera-Bravo5[*], Javad Sharifi-Rad6,7, Daniela Calina8

1Department of Pharmaceutical Botany, Faculty of Pharmacy, Ankara University, Tandogan, Türkiye

2Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Tandogan, Türkiye

3Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Tandogan, Türkiye

4Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ankara University,Tandogan, Türkiye

5Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago, Chile

6Universidad Espíritu Santo, Samborondón 092301, Ecuador

7Department of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea

8Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania

EXCLI J 2025;24:Doc289

 

Abstract

Rhoifolin is a flavonoid found in various plant species, especially within the Rutaceae family, and is considered a dietary component due to its presence in edible plants. Its bioactive properties, such as cytotoxic and anticancer activities, have gained significant attention. This review aims to highlight the general properties and diverse bioactivities of rhoifolin, with a particular focus on its cytotoxic and anticancer effects. This is based on a comprehensive literature search, focusing on the presence of rhoifolin in different plant species and its biological activities, particularly its anticancer properties. Rhoifolin is widely distributed in the plant kingdom, especially in Citrus species. It exhibits a variety of bioactivities, including strong cytotoxic and anticancer effects. Recent studies have shown that rhoifolin can induce apoptosis and inhibit cancer cell proliferation, making it a promising candidate for anticancer therapies. Rhoifolin's diverse bioactivities, particularly its cytotoxic and anticancer properties, position it as a potential therapeutic agent. Further detailed investigations into its molecular mechanisms and well-designed clinical studies are needed to fully understand and utilize its therapeutic potential.

See also the graphical abstract(Fig. 1).

Keywords: rhoifolin, apigenin-7-O-neohesperidoside, flavonoid, Citrus, anti-cancer activity

Introduction

Flavonoids, a group of omnipresent plant metabolites, have been recognized as phytochemicals with specific pharmacological activities associated with human health. The therapeutic efficacy of fully characterized flavonoids such as quercetin, curcumin, berberin, rutin, apigenin, naringenin, catechin etc. on various chronic diseases, especially cancer, has been demonstrated in many studies (Rasouli et al., 2019[145]). Flavonoids are important secondary metabolites that are known to be present in fruits and vegetables, having a wide spectrum of bioactivities like antiviral, antiplatelet, anti-allergic, antioxidant, anti-inflammatory, hepatoprotective, insulin mimetic, protective in cardiovascular diseases, antitumor and highly selective cytotoxic activities that might be beneficial for human health (Tanwar and Modgil, 2012[175]; Refaat et al., 2015[147]). However, the safe use of herbal medicines poses a significant barrier to their development due to insufficient data on the incidence of side effects (Dores et al., 2023[41]). Numerous studies have been carried out in the recent past related to the anti-proliferative activities of flavonoids in general. Furthermore, numerous studies focused on the anticancer activity of Citrus flavonoids, Citrus juices and extracts, in which rhoifolin was also found to be present (Benavente-Garcia and Castillo, 2008[25]; Visalli et al., 2014[182]; Song et al., 2022[166]). Citrus aurantium L. (bitter orange) is among these species having rhoifolin and also having anti-cancer usage (Suryawanshi, 2011[172]). There are other plants with cytotoxic and/or anti-cancer activity having this important secondary metabolite within their compositions, as well (Hasibuan et al., 2020[69]; Persia et al., 2020[134]). Thus, this review is planned to focus on the anti-cancer effect of rhoifolin which is found to be present in different plant species belonging to different families. Rhoifolin is a flavone glycoside that is also known as apigenin-7-O-neohesperidoside (Lefort and Blay, 2011[96]), isolated firstly from fresh Rhus succedanea L. leaves, a plant species belonging to the Anacardiaceae family (Hattori et al., 1952[70]; Huang et al., 2014[75]). One of the earliest records related to rhoifolin that is found in many different plant species, dates back to 1952. When the compound was first isolated, it was reported to be a new flavone glycoside of apigenin (apigenin-7-rhamnoglucoside) (Hattori et al., 1952[70]), however in some studies, the compound is named as apigenin-7-O-neohesperidoside, as well (Hafez et al., 2003[66]; Abd Elhameid et al., 2006[2]). When we look at the plant kingdom, various Citrus plants are known to possess this compound in general, such as oranges, bergamot, lemon, kumquat, mandarin, tantor etc. (Ramful et al., 2010[143]; Refaat et al., 2015[147]).

Methodology

A comprehensive literature search was conducted to identify relevant studies on the anticancer properties of rhoifolin. The search was performed across multiple electronic databases, including PubMed/MedLine, Scopus, Web of Science, and Google Scholar. The search strategy involved the use of Medical Subject Headings (MeSH) terms and Boolean operators to ensure a thorough and precise retrieval of relevant articles. The primary MeSH terms used included "r," "Flavonoids," "Antineoplastic Agents," "Cytotoxicity," and "Nanoparticles." These terms were combined using Boolean operators such as "AND," "OR," and "NOT" to refine the search. For example, the search query "rhoifolin AND (Antineoplastic Agents OR Cytotoxicity) AND Nanoparticles" was used to capture studies focusing on the anticancer properties of rhoifolin and its delivery via nanoparticles. The search was limited to articles published in English. Studies were included if they were original research articles, reviews, or meta-analyses focusing on the anticancer properties of rhoifolin. Both in vitro and in vivo models were considered. Studies were required to report on the anticancer activity of rhoifolin, including its cytotoxic effects, mechanisms of action, and potential for clinical application. No restrictions were placed on the publication date, allowing the inclusion of both historical and recent studies. Studies that were not peer-reviewed, such as conference abstracts, editorials, or opinion pieces, were excluded. Articles that did not specifically address the anticancer properties of rhoifolin or focused on unrelated bioactivities were excluded. Additionally, articles published in languages other than English were excluded. Data from the selected studies were extracted independently by two researchers using a standardized data extraction form. The extracted data included study design, model systems used (e.g., cell lines, animal models), outcomes measured (e.g., cell viability, apoptosis induction, tumor growth inhibition), and key findings related to the anticancer properties of rhoifolin. The most important data were synthesized and summarized in tables and figures to provide a clear and concise overview of the findings. These visual aids include details on the types of cancer cells affected by rhoifolin, its mechanisms of action, and the efficacy of different delivery systems, particularly nanoparticle-based approaches. Furthermore, the chemical structures and taxonomy of the plants associated with rhoifolin were validated using PubChem and the World Flora Online (WFO) database to ensure accuracy and consistency in the scientific details reported (PubChem[124], WFO, 2023[189]).

Rhoifolin: a Brief Overview

Natural sources

When we search the literature, we can see that this compound is quite common in the nature and is present in many species of different plant families. A tabulated list of some of these plants is presented in alphabetical order in Table 1(Tab. 1) (References in Table 1: Abbas et al., 2021[1]; Abd Elhameid et al., 2006[2]; Abiri et al., 2021[3]; Agogbua et al., 2022[6]; Alfarisi et al., 2020[8]; Alreshidi et al., 2020[9]; Amoroso et al., 2021[12]; Andrade-Pinheiro et al., 2023[13]; Awad et al., 2014[15]; Barberis et al., 2020[19]; Barreca et al., 2011[22][21][23]; Bindu and Udayan, 2018[26]; Brinza et al., 2020[27]; Burlando et al., 2017[30]; Cai et al., 2020[31]; Casabuono and Pomilio, 1990[32]; Chen et al., 2021[35], 2023[34]; Cheng et al., 2017[36]; Cicero et al., 2017[37]; Dai et al., 2021[38]; Dhanabal et al., 2006[40]; Dormousoglou et al., 2023[42]; Egger and Keil, 1969[43]; Ekeke et al., 2019[44]; Eldahshan and Azab, 2012[46]; Eldahshan, 2013[45]; Elma et al., 2019[47]; El-Shawi and Eldahshan, 2014[48]; El-Shawy, 2014[49]; Eroğlu Özkan et al., 2019[50]; Fahim et al., 2015[52]; Fan et al., 2019[53]; Fenglin et al., 2004[54]; Gattuso et al., 2006[57]; Ge, 2014[58]; Giambanelli et al., 2018[60]; Guetchueng et al., 2020[65]; Hafez et al., 2003[66]; Han et al., 2023[67]; Hasibuan et al., 2020[69]; Hattori et al., 1952[70]; He et al., 2003[71]; Hong et al., 2021[73]; Huang et al., 2014[75]; Huo et al., 2021[77]; Huynh et al., 2016[78]; Jaiswal et al., 2019[80]; Juee, 2022[83]; Kanao and Matsuda, 1978[84]; Kaneko et al., 1995[85]; Kanes et al., 1993[86]; Kayahan and Saloğlu, 2022[88]; Kiem et al., 2010[90]; Kumar et al., 1999[93]; Kuo et al., 2017[94]; Lee et al., 2010[95], Liang et al., 2007[97]; Lin et al., 1994[100], 1998[99], 2023[101]; Liu et al., 2012[105], 2013[102], 2021[103], 2023[106]; Lopez-Gutierrez et al., 2015[109], 2016[110]; Ma et al., 2013[113], 2014[114], 2018[112]; Mencherini et al., 2013[122]; Nakajima et al., 2014[123]; Negm et al., 2022[125]; Neretina et al., 2005[126]; Okonwu and Muonekwu, 2019[129]; Ozkan et al., 2022[130]; Papalia et al., 2017[131]; Pernice et al., 2009[133]; Persia et al., 2020[134]; Plioukas et al., 2016[135]; Prashar and Patel, 2020[137]; Qiaoyu and Lingsheng, 1996[140]; Rahmouni et al., 2022[141]; Rajkumar and Jebanesan, 2008[142]; Rao et al., 2011[144]; Sawikowska, 2020[150]; Saxena et al., 2014[152], 2016[151]; Sayeed et al., 2023[153]; Senizza et al., 2021[154]; Seukep et al., 2020[155]; Sharifi-Rad et al., 2021[156]; Shen et al., 2017[158]; Shimokoriyama, 1966[159]; Son et al., 1992[165]; Song et al., 2022[166]; Souilah et al., 2020[169], 2021[168]; Sultana et al., 2018[171]; Suryawanshi, 2011[172]; Taheri et al., 2023[173]; Tsujimoto et al., 2019[177]; Uddin et al., 2011[178]; Uysal et al., 2019[179]; Vega-Ruiz et al., 2021[181]; Visalli et al., 2014[182]; Vo et al., 2022[183]; Wang et al., 2010[187], 2016[185], 2017[184]; Xie et al., 2021[191]; Yadav et al., 2009[193]; Yang et al., 2021[195]; Ye et al., 2014[197]; Yıldız Turgut et al., 2019[198]; Zengin et al., 2021[199]; Zhang et al., 2005[201], 2009[200], 2022[202]; Zhang, 2007[204]; Zheng et al., 2022[205]; Zhou et al., 2023[207][208]). In time, taxonomical hierarchy and nomenclature of some of these species have changed; therefore, synonyms for these species (if present) are also provided.

Today, biotechnological studies on plants use plant in vitro culture strategies to produce therapeutically effective phytochemicals with a completely safe efficacy profile without metabolite variations due to different geographical and climatic conditions (Thorpe, 2007[176]; Khan et al., 2021[89]). Although in vitro culture technologies have an increasing interest and importance due to their potential to grow any plant anywhere, to provide a sophisticated production platform for phytochemicals, and to enhance new plant-based medicinal compounds, there has been still no biotechnological-based in vitro cell culture study for the production of rhoifolin in the literature.

Chemical characterization

When the importance of flavonoids that constitute a diverse group of polyphenolic compounds having various and significant biological activities including antiinflammatory, cardioprotective, antidiabetic, and anti-cancer effects were understood, many researchers started to investigate their health-related benefits (Nijveldt et al., 2001[127], Refaat et al., 2015[147]). Rhoifolin (apigenin 7-O-neohesperidoside) is a flavonoid and an apigenin derivative that bears an alpha-(1->2)-L-rhamnopyranosyl)-beta-D-glucopyranosyl moiety that is attached to the 7-hydroxy group. Several studies on the diverse activity potentials of the mixtures having this compound have been reported in the literature (Lou et al., 2016[111]; Burlando et al., 2017[30]; Kuo et al., 2017[94]; Brinza et al., 2020[27]; Seukep et al., 2020[155]; Qi and Liu, 2022[139]; Zheng et al., 2022[206]). The chemical structure, nomenclature, and spectroscopic properties of rhoifolin are given in Table 2(Tab. 2). In respect to chemical nomenclature, rhoifolin is apigenin 7-O-β-neohesperidoside having C27H30O14 chemical formula that has a molecular weight of 578.53 (exact mass: 578.1636). The compound is usually isolated as yellow amorphous powder or yellow needles after crystallization from methanol (melting point: 245-253 °C). It is soluble in hot ethanol, methanol, water; sparingly soluble in cold ethanol, ethyl acetate, and insoluble in chloroform and n-hexane (Refaat et al., 2015[146]).

Semi-synthetic derivatives

Studies regarding the derivatization of rhoifolin are limited in the literature. One of them is the transglycosylation route conducted by Aoki et al., in which alpha-glucosyl rhoifolin was synthesized and its 1H and 13C-NMR signals were identified by using a variety of NMR methods (HSQC, COSY, HMBC, and 1D TOCSY) and mass spectrometry. Accordingly, the assignment of the signals for rhoifolin and sugar moieties was completed. With the help of correlations in the HMBC spectra, it was understood that intramolecular hydrogen bonds were present within the flavone and the flavonol skeletons of Rhf-G, as shown in Figure 2(Fig. 2) (Aoki et al., 2017[14]).

The usage of rhoifolin as an intermediate was a strategy implemented for the chemo-enzymatic synthesis of acacetin (Hanamura et al., 2016[68]). To achieve the end product, rhoifolin (1b) was prepared from naringin (3a), which is essentially an oxidation process with I2 and pyridine at 95 ℃. After cooling down, forming 1b was acetylated by adding acetic anhydride into the mixture. Then, this mixture was heated for 6 h at 95 ℃ to yield 1c using the process given in the literature. For the enzymatic deacetylation of R2, Candida antarctica lipase B was used at given conditions to afford 1d with excellent yield (98 %). An in-situ formed diazomethane solution was used to methylate 1d at the same position, to achieve 1e. To prepare this solution beforehand, N-methyl-N-nitroso-p-toluene-sulfonamide was added to the solution of KOH in water and EtOH. For the total deacetylation of 1e to obtain 1f, this compound was dissolved in methanol and added to a solution of sodium methoxide. Finally, sugar side chain of 1f was cleaved to obtain 1a with high yield and regioselectivity, using concentrated H2SO4 and a reflux set-up (Figure 3(Fig. 3); Reference in Figure 3: Hanamura et al., 2016[68]).

Anticancer Activities of Rhoifolin

Mechanism of antitumor action of rhoifolin

Rhoifolin is present in many plant species and one of them is Callicarpa nudiflora Hook & Arn. which grows widely in southern China. Xiong and colleagues (2021[192]) investigated the anti-motile effects of rhoifolin in this plant, to evaluate if this effect on cell motility would produce anti-cancer effect. Ezrin and podocalyxin (PODXL) are the main actors that would organize membrane proteins and signal transduction mechanisms, thus eventually modulating the cytoskeleton rearrangement in cell motility.The latter regulates cell motility by interacting with actin polymerization complex composed of ezrin, and PDZ proteins NA+/H+ exchanger regulatory factor isoforms 1 and 2 (NHERF-1/2) (Sizemore et al., 2007[163]). Ezrin and NHERF-1/2 are adaptor proteins facilitating the interaction of PODXL with the cytoskeleton within epithelial cells. PODXL, ezrin, and NHERF-1/2 interactions result in metastasis induction via Cdc42, MAPK, PI3K, Rac1, and RhoA (McNagny et al., 2012[120]; Flores-Tellez et al., 2015[56]). RFL's activity on breast cancer cell viability was assessed via MTT assay. Antimigratory properties were elucidated by ORISTM cell migration assay and immuno-precipitation was performed to assessing the effects of RFL on the interaction between ezrin and PODX. Results demonstrated that RFL resulted in remarkable inhibitions on cell migration and alterations in the location and organization of actin cytoskeleton in breast cancer cells. Secondly, RFL suppressed ezrin phosphorylation and its interaction with PODXL, thus exerting anti-motile effect. Moreover, it inhibited TGF-β1-induced EMT in MDA-MB-231 cells. As a conclusion, it was deduced that the anti-motile action of RFL was due to its potential downregulatory effect on PODXL-Ezrin interaction during EMT (Xiong et al., 2021[192]). In another study, Zheng and colleagues investigated the potential anticancer effects of flavonoids in Plumula nelumbinis, which is the green embryo of the plant named Nelumbo nucifera Gaertn. against pancreas cancer. According to high-performance liquid chromatography (HPLC) and mass spectrometry (MS) results, rhoifolin, apiin, and vitexin were found to be the most abundant compounds (Figure 4(Fig. 4) and Figure 1(Fig. 1): Graphical abstract). Cell viability tests showed that these three compounds have the potential to inhibit PANC-1 and ASPC-1 cell lines' proliferation. Among these compounds, rhoifolin was the most potent compound, which also promoted apoptosis of pancreatic cancer cells via up-regulation of JNK and p-JNK with the down-regulation of p-Akt. Furthermore, this compound led to the inhibition of cell migration and invasion while it effectively enhanced antioxidant capacity of PANC-1 and ASPC-1. AKT activator (SC79) and JNK inhibitor (SP600125) that both reversed the anticancer effects of rhoifolin on pancreatic cancer cells, thus validating the mechanism of this compound. According to quantitative proteomics analysis, it also modified proteomic profiles in these cancer cell lines. Western blot analysis results demonstrated the downregulation of transforming growth factor beta 2 (TGF-β2) and phosphorylated SMAD family member (SMAD2). All these findings suggest that rhoifolin may exert its anti-pancreatic cancer activities through signaling pathways of Akt/JNK/ caspase-3 and TGF-β2/SMAD2 (Zheng et al., 2022[205]).

Evidence from preclinical studies confirmed the anticancer properties

Anticancer action of rhoifolin in the plant species has been evidenced by a few assays according to the existing literature (Table 3(Tab. 3); References in Table 3: Eldahshan, 2013[45]; Koch et al., 2005[91]; Ma et al., 2014[115]; Xiong et al., 2021[192]; Zheng et al., 2022[205]). For instance, Ma and co-workers assessed the cytotoxicity of the EtOH extracts prepared from Callicarpa nudiflora by the MTT assay. The flavonoid composition of this plant was isolated and purified by HP-20 macroporous resin, silica gel and Sephadex LH-20 column chromatography methods. The structures of the twelve isolated compounds were elucidated by spectroscopic data and one of them was identified as rhoifolin. Among these compounds, luteoloside, luteolin-4'-O-β-D-glucoside, 6-hydroxyluteolin-7-O-β-glucoside, luteolin-7-O-neohesperidoside, rhoifolin, luteolin-7,4-di-O-glucoside (Figure 5(Fig. 5)) exerted proliferation inhibitory activities, in various concentrations, against HeLa, A549, and MCF-7 cells. Meanwhile, 6-hydroxyluteolin-7-O-β-glucoside, rhoifolin, and nudifloside produced much higher cytotoxic activities (Ma et al., 2014[115]).

Eldahshan studied the antitumor activity of rhoifolin against human epidermoid larynx (Hep 2), human cervical (HeLa), hepatocellular (HepG2), colon (HCT-116) and fetal human lung fibroblast (MRC-5) carcinoma cell lines. According to the assay results, the compound exhibited high cytotoxicity against Hep 2 with IC50=5.9 μg/mL whereas the value for the standard vinblastine was 4.6 μg/mL. The IC50 value for HeLa was 6.2 μg/mL and for vinblastine it was 5.2 μg/mL. The IC50 of rhoifolin was 22.6 μg/mL for HepG2, 34.8 μg/mL for HCT-116 and 44 μg/mL for MRC-5. Moreover, calculated selectivity index (SI) data for rhoifolin was greater than 8.47 for Hep 2, followed by 8.06 for HeLa and 2.21 for HepG2.

The compound was found to be safe for these cells, and toxic for colon and fetal human lung fibroblast cell lines, since an SI value less than 2 indicates the general toxicity of the compound (Koch et al., 2005[91]; Eldahshan, 2013[45]).

Other Biological Activities of Rhoifolin

Today, the tendency to use natural and traditional medicines to cope with chronic diseases that have difficulties in diagnosis and treatment has increased (Sharma et al., 2024[157]). In this context, it is essential to consider rhoifolin and other natural bioactive compounds in terms of their scientific, medical, and traditional applications, particularly given the growing interest in flavonoid chemical structures and their pharmacological properties. The literature search result showed that this compound is widely distributed in many families of the plant kingdom and can be obtained in significant quantities especially from Citrus and Chorisia species. Moreover, in vitro and in vivo studies have shown that rhoifolin has many biological activities. According to these pharmacological findings, rhoifolin is on its way to being among the most preferred drugs, especially due to its strong anti-inflammatory, hepatoprotective, hypoglycemic and highly selective cytotoxic effects. Therefore, in the future, a detailed investigation of the molecular mechanisms of these activities will be necessary, along with well-designed clinical studies (Eldahshan, 2013[45]; Refaat et al., 2015[146]).

Antioxidant properties

The dynamic interplay of oxidative stress, cellular communication and signaling pathways is fundamental to understanding the initiation, progression, and therapeutic resistance of cancer (Alshehri et al., 2022[11]; Iqbal et al., 2024[79]). Since the health benefits of flavonoids are accredited to their antioxidant activities, Citrus grandis L. Osbeck (Shatianyu) was investigated by Mei Deng and co-workers for its chemical composition of flavonoids. In their study, they isolated and identified 11 flavonoids from Shatianyu pulp flavonoid extracts (SPFEs). They also evaluated the cellular antioxidant activity (CAA) and oxygen radical absorbance capacity (ORAC) of the isolated compounds. Naringin and rhoifolin showed the highest ORAC activities while SAR results suggested that 3-hydroxy-3-methylglutaryl or 4'-glucose decreases the ORAC activity of flavonoids. The contribution to the holistic antioxidant activity of these flavonoids was evaluated by an online knockout method in Table 4(Tab. 4) (Reference in Table 4: Deng et al., 2022[39]). ORAC activity was mostly influenced by melitidin, bergamjuicin and naringin (Figure 6(Fig. 6)) (Deng et al., 2022[39]).

Another example of ORAC analysis to determine antioxidant activity was conducted by Zhang et al. In their study, they separated and identified the components of Piper nigrum L. leaf and fruit extracts. According to the obtained results, the ORAC value of leaf extracts was 3639.05 μmol TE/g, which was greater than that of the fruit extracts. Among all the solvents, the ethanol extract was determined to exert the highest ORAC value. Consequently, active components were isolated from ethanol extracts of Piper nigrum leaves with silica gel chromatography (Sephadex LH-20 resin), reversed-phase chromatography, gel-filtration chromatography, thin-layer chromatography and HPLC. Structural confirmations of hinokinin (Figure 7(Fig. 7)) and rhoifolin were performed by utilizing nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) techniques. ORAC values for these compounds were found to be 16070 μmol TE/g and 10823 μmol TE/g, respectively (Zhang et al., 2015[203]).

Wang and co-workers evaluated the DPPH radical scavenging activity of the n-butanol extract of Lonicera japonica Thunb. leaves. In this assay, they assessed the extract and the compounds which were identified with HPLC present in the extract. Ascorbic acid and Vitamin E were used as standards (Table 3(Tab. 3)). Authors concluded that this crude drug had an antioxidant potential value (11.2 µg/mL) close to that of Vitamin E (9.5 µg/mL) (Table 5(Tab. 5); Reference in Table 5: Wang et al., 2017[184]). Moreover, they isolated the compounds in the extract and also identified their DPPH scavenging activities. Based on the table below, compounds 1-9 (rhoifolin being 4) exhibited high radical scavenging activity which eventually translate to high antioxidant activity (Wang et al., 2017[184]).

Anti-inflammatory effect

Inflammation, arising as local response of living mammalian tissues when injured, is also a fundamental protective process that aims to preserve the general structure of the organism against infectious, physical and chemical attacks (Eldahshan and Azab, 2012[46]). However, when the body is exposed to toxins and other agents like chronic stress, obesity, and autoimmune disorders, unwanted body responses can sometimes be triggered. In this case, instead of curing the problem and then restoring the normal functioning, inflammation might persist; proinflammatory cytokines, chemokines, adhesion molecules and inflammatory enzymes come into play. Today, it is thought that this chronic inflammatory condition can result in various health problems, including arthritis, depresssion, cardiovascular problems, Alzheimer's Disease (AD) and even cancer (Singh et al., 2019[161]). Steroids, nonsteroidal anti-inflammatory drugs (NSAIDs) and immunosuppressants that are used to control and/or suppress the inflammatory crisis might result in various side effects, as well. In practice, the aim is to administer the minimum effective dose with the highest efficiency and the fewest side effects. Therefore, it is necessary to incorporate natural anti-inflammatory factors in drug therapy to achieve an enhanced pharmacological response, and the lowest extent of adverse effects. In this context, herbal medicines are prominent agents in medicine (Ghasemian et al., 2016[59]). Throughout history, various herbs have been used in the treatment of inflammation and associated ailments like rheumatism all over the world. Flavonoids found in plants used in many traditional medicines have been associated with this activity. In this context, apigenin, a phytopolyphenol commonly found in the human diet, is an important example. It was confirmed by Sawatzky et al. that apigenin, like many other flavonoids, exerts anti-inflammatory effects like reducing oxidative stress and preventing the expression of various inflammatory factors (Sawatzky et al., 2006[149]). With the knowledge provided by the apigenin glycoside study; a different study group was motivated to investigate rhoifolin's anti-inflammatory activity that has not been tested previously. Rhoifolin administered at doses of 2.5, 25, and 250 mg/kg resulted in a significant inhibition of rat paw edema of 14 %, 25 %, and 45 %, respectively, after 4 hours of treatment compared to the control group (74 %). In addition to the significant abolition of prostaglandin E2 levels with increasing rhoifolin doses, TNF-α release in inflammatory exudates was significantly reduced. In this study, rhoifolin was found to have potent anti-inflammatory activity at low doses (Eldahshan and Azab, 2012[46]). The study by Peng et al. (2020[132]) aimed to reveal the effect of rhoifolin on arthritis induced by complete Freund's adjuvant (CFA) in rat models. Significant improvement was observed in paw edema and weight loss parameters with the administration of Rholifolin at doses of 10 and 20 mg/kg. These improvements were also confirmed with the data obtained as a result of histopathological observations. Additionally, significant decrease in oxidative stress was observed with rhoifolin administration, as evidenced by the changes in intracellular glutathione, glutathione peroxidase, superoxide dismutase and malondialdehyde levels in the tissue of articular cartilage. Besides, proinflammatory cytokines, tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 gene expression exhibited a significant downregulation of gene expression. According to this group, antioxidant and anti-inflammatory effects of the compound were probably via the NF-κB pathway, however, the exact compounds responsible for this action need to be determined in future studies. Osteoarthritis (OA) or degenerative arthritis, which reduces the quality of life of approximately 250 million people worldwide, is an important public health problem today. It is a chronic musculoskeletal disease, especially seen in the elderly population and affects mobile connections between two bones such as the knee and hip joints (Kraus et al., 2015[92]). OA affects all structures of the joints and is characterized by cartilage destruction, remodeling of subchondral bone, osteophyte formation, and changes in the synovium and joint capsule (Goldring and Goldring, 2010[61]). In some studies, it has been shown that proinflammatory cytokines can trigger cartilage destruction by directing the production of catabolic degrading enzymes in chondrocytes. Therefore, inhibition of synovial joint inflammation might be an effective form of treatment for OA (Meliconi and Pulsatelli, 2019[121]). A study based on this information aimed to address the protective effects of rhoifolin on OA with some in vitro and in vivo experiments. Results showed that rhoifolin suppressed senescence-associated secretory phenotype factors' expression and the senescence phenotype in IL-1β-treated chondrocytes. Additionally, rhoifolin inhibited IL-1β-induced activation of the NF-κB pathway. Molecular docking and knock-down studies demonstrated that rhoifolin might also bind to Nrf2 to suppress the NF-κB pathway. Finally, rhoifolin was shown to ameliorate the OA process in an in vivo ACLT rat model (Chen et al., 2022[33]). Another study conducted by Yan et al. also contributed to the knowledge of the therapeutic effects of rhoifolin and provided a new perspective for prospective treatment of OA. They clearly reported the anti-inflammatory, anti-cartilage degradation and autophagy promoting properties of rhoifolin, which was confirmed to function by regulating autophagy. Furthermore, P38/JNK and PI3K/AKT/ mTOR pathways were also involved in the process (Yan et al., 2021[194]).

Neuroprotective

Neurodegeneration is a complication of incurable age-related diseases that have a toll on the nervous system and significantly reduces the life qualities of both elderly patients and their families (Azzini et al., 2024[16]). With the prolongation of life expectancy in modern societies, the prevalence of neurodegenerative diseases has also increased, and this issue has turned into a globally recognized public health problem and it is well-known that one of the most important causes of age-related dementia is AD. Today, there is no effective treatment for this disease. Recently, nutrition has been considered a very important factor in the protection of the body against chronic inflammation and oxidative stress that lead to chronic degenerative diseases (Ezzat et al., 2024[51]). Numerous bioactive food components might affect the pathological mechanisms underlying AD. Among these, phenolic compounds, omega-3 fatty acids, isothiocyanates, fat-soluble vitamins and carotenoids are promising agents in this respect (Grodzicki and Dziendzikowska, 2020[63]). In a study, ameliorative effects of rhoifolin on zebrafish anxiety induced by scopolamine, amnesia, brain oxidative stress and the mechanisms underlying these disorders were investigated. For nine consecutive days, rhoifolin (1, 3 and 5 μg/L) and then scopolamine (100 μM) were administered to Zebrafish 30 minutes before behavioral tests (novel tank diving test, Y-maze and novel object recognition tests). Rhoifolin, isolated from the leaves of Chorisia crispiflora Kunth (Malvaceae) can alleviate memory deficits, anxiety, brain oxidative stress in scopolamine-treated zebrafish and regulate cholinergic function by inhibiting AChE activity. The results of this study showed that rhoifolin was a promising compound against amnesia and anxiety via restoration of cholinergic activity and improving brain oxidative stress (Brinza et al., 2020[27]). Spinal cord injury usually results from physical damage that results in infiltration of inflammatory cells and secondary degeneration. It leads to detrimental impairment of neurological dysfunctions (autonomic, motor and sensory) (Ahn et al., 2015[7]). In a study designed with the thought that the widely known antioxidative property of rhoifolin may be effective in spinal cord injury, rhoifolin inhibited proinflammatory cytokines and NF-κB pathways, and significantly reduced intracellular oxidative stress in the spinal cord-injured rat model. Overall, there were significant improvements in motor function in rats treated with rhoifolin. Thus, the study evidently confirmed that rhoifolin could be a safer alternative than current treatments for spinal cord injury. It has been reported that the effect may be due to a decrease in apoptotic signal, as observed in reduced levels of p38MAPK and caspases (Long et al., 2023[108]). Rhoifolin was also investigated in the experimental rat model of AD, induced by streptozotocin administration. Rhoifolin's effect on the thickness of the CA1 pyramidal layer and spatial learning in the AD model were examined, as well. The Morris water maze test and novel object recognition test were used to demonstrate the effects on relieving Alzheimer's symptoms. In the hippocampus and cerebral cortex, the free-radical scavenging activity of rhoifolin was estimated by calculation of levels for key enzymes (GPx, GRX, SOD and CAT) belonging to the antioxidant defense system. Obtained results showed that rhoifolin could be an promising agent for the management of AD (Huang et al., 2021[76]).

Hepatoprotective

The liver is one of the most important organs in the body that plays an important role in metabolism, detoxification and storage of endogenous and exogenous substances. Due to the vital functions of this organ, liver diseases are among the leading public health problems all over the world. Despite the high level reached by modern medicine, there is not yet a defined drug that supports liver functions, fully protects the organ or helps regenerate liver cells. Thus, it is important to find new pharmaceutical alternatives to be used in the prevention and treatment of liver disorders. Due to the compounds, they contain, some plants play fundamental roles in human health regarding their hepatoprotective potentials that have also been the subject of many scientific studies (Madrigal-Santillan et al., 2014[116]). Alcoholic liver disease (ALD) refers to the spectrum of liver damage due to excessive alcohol consumption (Liu et al., 2019[104]) and its prevalence is increasing worldwide as an important public health problem (Wang and Liu, 2021[186]). Citrus grandis is a valuable traditional medicine dating back to hundreds of years in China, and it has been proven to have many pharmacological properties with activity studies. Tea prepared with C. grandis is traditionally used to sober up a drunk person due to its hangover-preventing effects. A group that had previously investigated the therapeutic effect of total flavones from C. grandis in alcoholic liver disease (Xiao et al., 2012[190]) evaluated the hepatoprotective effects of rhoifolin along with its potential mechanisms. In this study, rhoifolin showed protective effects on a mouse model of ALD and ethanol-treated LO2 cells. Downregulation of CYP2E1, TLR4 expressions, and NF-κB phosphorylation were proposed as the basic mechanisms. Providing an experimental ground for the traditional use of Citrus grandis, these results underlined that rhoifolin is a promising herbal remedy for alcoholic liver disease. Additionally, it was also highlighted that further clinical studies should be conducted to confirm the efficacy and safety of this herbal remedy in patients with ALD (Mai et al., 2022[118]).

Antidiabetic

Chronic hyperglycemia in diabetic patients leads to an increase in oxidative stress in different tissues and plays an important role in the progression of various related complications including nephropathy, neuropathy, retinopathy, and cardiovascular disorders (Mahmood et al., 2015[117]). The leaves of Citrus grandis (L.) Osbeck, also known as red wedun with a high content of rhoifolin (1.1 %, w/w) have been used in Traditional Chinese Medicine (TCM) in the treatment of diabetes. Rhoifolin has the potential to be among the effective precursors for the treatment of diabetes and exerts its antidiabetic effects via increased adiponectin secretion, GLUT4 translocation and phosphorylation of insulin receptor-β in 3T3-L1 fat cells, which explains the traditional use of C. grandis in diabetes. Critical genes in the context of the rhoifolin effect may lead to the emergence of new targets in the treatment of diseases that occur as a result of insulin resistance (Rao et al., 2011[144]).

Regulatory effect on bone metabolism

Today, the incidence of aseptic loosening after joint-prosthesis replacement operations is increasing. Aseptic loosening can be defined as the failure of joint prostheses without a mechanical cause or any infection. This condition is most often caused by osteolysis (bone resorption) and an inflammatory cellular response in the joint (Hench, 2019[72]). It was revealed that the bone resorption of hyperactive osteoclasts plays a very important role in osteolysis. A study showed that rhoifolin reduced RANKL-induced osteoclastogenesis and bone resorption in vitro. In addition, analyses made on titanium particle-induced osteolysis mouse models approved that rhoifolin well ameliorated osteoclast‐stimulated calvarial osteolysis. In this study, it was demonstrated that rhoifolin potently suppressed receptor activators of nuclear factor-κB (NF-κB) ligand-induced osteoclastogenesis, hydroxyapatite resorption, F-actin formation, and expression of osteoclast-related genes. These results especially highlighted the potential of rhoifolin as an important agent for the improvement of prosthesis loosening (Liao et al., 2019[98]).

Nephroprotective

Therapeutic use of cisplatin in tumor chemotherapy is limited due to dose-related nephrotoxicity. The effects of rhoifolin on cisplatin-induced nephrotoxicity were investigated by using rats, in which renal damage was demonstrated by decrease in body weight, increase in blood urea, nitrogen and creatinine, and the destruction of histological integrity. Nevertheless, rhoifolin administration ameliorated cisplatin induced nephrotoxicity. Furthermore, rhoifolin administration resulted in alleviation of cisplatin-induced oxidative stress and inflammatory response. Finally, rhoifolin administration inhibited nuclear translocation of NF-κB via downregulation of phospho-IκBα and phospho-p65, and it also up-regulated IκBα, suggesting that it could be a promising adjunct for cisplatin in tumor treatment (Song et al., 2020[167]).

Cardioprotective

Hypertension is a fairly common condition including various health risks, and studies have mostly focused on cardiovascular diseases and related problems (Buford, 2016[29]). As overexpression of angiotensin-converting enzyme (ACE), which is a fundamental component in the renin-angiotensin-aldosterone system (RAAS) regulating blood pressure, is associated with vascular hypertension, inhibition of ACE is of great importance in respect to hypertension. Recently, research on potential ACE inhibitors started to include natural product derivatives such as peptides, polyphenolics and terpenes (Balasuriya and Rupasinghe, 2011[18]). In a study conducted within this context, 17 flavonoids belonging to five structural subtypes were evaluated in vitro for their abilities to inhibit ACE. As a result of this study, which was carried out with two different concentrations (500 µM and 100 µM) using the fluorimetric method, it was determined that the inhibitory potential ranged from 17 % to 95 % at 500 µM and from 0 to 57 % at 100 µM. The IC50 value for rhoifolin was determined to be 183 µM. In this study, which showed that flavonoids are an excellent source of functional antihypertensive products, the structural features that increase the activity on the flavonoid skeleton were determined as follows: presence of (a) a catechol group in the B ring, (b) double bond between C2 and C3 in the C-ring, and (c) ketone group at C4 in the C-ring (Guerrero et al., 2012[64]). In another study conducted in the 1990s, acute effects of luteolin, apiin, and rhoifolin on pulmonary vascular circuit were compared with nifedipine in two pulmonary hypertension experimental models during hypoxia and PGF-induced pulmonary vasoconstriction in dogs under anesthesia. Although 5 mM/kg/ i.v. rhoifolin did not cause any change in hypoxic pulmonary vasoconstriction, it led to decrease in cardiac output and aortic pressure (Occhiuto and Limardi, 1994[128]).

Antimicrobial

Antibiotic resistance is an important and growing phenomenon and constitutes a significant public health problem of the 21st century. Though a high number of antibiotics are being used in modern medicine, microbial resistance to these antibiotics has increased significantly due to their inappropriate and widespread usage and the rapid genetic transmission of resistance. The discovery of new drugs that can overcome microbial resistance is of great importance to save humanity from entering a critical era in which minor injuries and infections can become life-threatening (Vadhana et al., 2015[180]). Plants have the potential to provide an unlimited number of antimicrobial compounds due to their rich phytochemical profiles and the fact that they have been used in many different traditional therapeutic applications for centuries (Silva et al., 2013[160]). As one of the largest classes of secondary metabolites that are formed in different parts of the plant, flavonoids exhibit a wide range of pharmacological and beneficial health effects for humans. It is known that plants synthesize flavonoids, especially in response to microbial threats. Scientific studies have shown that these compounds are powerful antimicrobial agents against a variety of pathogenic microorganisms (Gorniak et al., 2019[62]).

Sepsis is a life-threatening organ dysfunction due to microbial infection and is responsible for systemic inflammation leading to organ dysfunction. According to the data reported by the World Health Organization (WHO), mortality due to sepsis is quiet high, about six million deaths occur on a a yearly basis (Hotchkiss et al., 2016[74]). Within the scope of search for alternative treatment methods for this serious health problem, a study on the protective effect of rhoifolin against sepsis was planned. After cecal ligation and puncture method induced sepsis was obtained, rhoifolin was administered to mice at doses of 20 and 40 mg/kg, i.p. for one week. Food intake, survival rates of the mice, and liver function test results and cytokines, were examined. Oxidative stress parameters were also determined in lung tissue homogenate, histopathological analyses were carried out in the liver and lung tissues. The results of the study showed that rhoifolin administration reduced oxidative stress and inflammation in sepsis of the mice via regulation of the TLR4/ MyD88/NF-κB pathway. For this reason, it has been reported that rhoifolin would be beneficial against sepsis and could be used clinically for the management of sepsis (Wen et al., 2023[188]).

Antiviral

COVID-19 pandemic, having severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the causative agent, has become the most important global public health problem in recent years (Popescu et al., 2022[136]). Despite administration of different vaccine types, mutation of new strains and the problems arising from universal immunity have shifted attention to the search for more effective solutions. Flavonoids are the most studied group in terms of antiviral effect, with their success in in silico, in vitro, in vivo and recently clinical studies (Kaul et al., 2021[87]). It was reported that the flavonol herbacetin and the flavones rhoifolin and pectolinarin inhibited the enzymatic activity of SARS-CoV 3CLprotease enzyme, which is one of the most studied targets for flavonoid inhibition and these three flavonoids were reported as the best inhibitory compounds against SARS-CoV 3CLpro. They attached to the active sites of proteins and thus inactivated these proteins, resulting in the neutralization of the virus. It has been suggested that hydrophobic aromatic rings and hydrophilic hydroxyl groups are effective in binding affinity. The IC50 values of herbacetin, rhoifolin, and pectolinanrin were calculated by the dose-dependent inhibitory curve and found to be 33.17 µM, 27.45 µM, and 37.78 µM, respectively (Jo et al., 2019[82]; Sawikowska, 2020[150]). With regard to SARS-CoV, some studies report that rhoifolin inhibits the 3CL protease enzyme of SARS-CoV (Benarba and Pandiella, 2020[24]; Russo et al, 2020[148]; Abiri et al, 2021[3]; Badshah et al, 2021[17]; Budak et al, 2022[28]; Singh et al, 2022[162]; Sruthi et al, 2023[170]), or show bioactivity; other studies report that it is also bioactive against the main protease and spike glycoprotein of SARS-CoV-2 (Sawikowska, 2020[150]; Tallei et al, 2020[174]; Adhikari et al, 2021[5]; Yantih et al, 2021[196]). Table 6(Tab. 6) (References in Table 6: Abiri et al., 2021[3]; Brinza et al., 2020[27]; Chen et al., 2022[33]; Deng et al., 2022[39]; Eldahshan and Azab, 2012[46]; Guerrero et al., 2012[64]; Huang et al., 2021[76]; Jo et al., 2019[82]; Liao et al., 2019[98]; Long et al., 2023[108]; Mai et al., 2022[118]; Occhiuto and Limardi, 1994[128]; Peng et al., 2020[132]; Rao et al., 2011[144]; Sawikowska, 2020[150]; Song et al., 2020[167]; Wang et al., 2017[184]; Wen et al., 2023[188]; Zhang et al., 2015[203]) summarizes the biological activities of rhoifolin.

Pharmacokinetic Data and Strategies to Increase the Bioavailability of Rhoifolin

Many flavonoids, especially rhoifolin, having therapeutic efficacy confirmed by in vitro/in vivo experiments, have a limited role as therapeutic agents in clinical use due to their physicochemical properties such as low solubility in water or low permeability (Abou Baker, 2022[4]; Ferreira et al., 2022[55]; Maity et al., 2022[119]; Zheng et al., 2022[205]). As a general rule, the pharmacokinetic features of the active substances such as absorption, distribution, metabolism and elimination are mainly affected by their physicochemical properties. In other words, all features of the active molecule such as lipophilicity, solubility in aqueous solutions, molecular weight, size, structure, polar surface area, ability to form hydrogen bonds, and resistance to enzymatic reactions are effective parameters for the bioavailability of the active ingredient (Smith, 1997[164]; Loftsson, 2015[107]). On the other hand, unpleasant odor or taste; stability problems caused by many factors such as light or heat during the storage stage, as well as body pH differences may also result in less or no therapeutic effect of the active ingredient. For example, quercetin has very limited clinical use due to its low water solubility, while epigallocatechin gallate is known to be unstable in neutral or alkaline solutions, and naringin is poorly absorbed when administered orally (Barras et al., 2009[20]). Hence modern drug delivery strategies with different methods and processes can come into play to overcome the physicochemical drawbacks of phytochemicals and to convert a biologically active compound into a therapeutically effective drug (Puglia et al., 2017[138]). Phytochemicals can be found on the market as dietary supplements, conventional drugs or cosmetics. Nanotechnology-based carriers, called colloidal drug carrier systems, are one of the most up-to-date approaches for biopharmaceutical applications of phytochemicals since they provide a solution for an active molecule which is insufficient in treatment alone. Colloidal drug delivery systems including polymeric nanoparticles, lipidic nanoparticles, liposomes, niosomes or phytosomes etc. are particles and vesicles with three external nanoscale dimensions. They should have a size range of about 1 to 1000 nm. Due to the unique and tunable size-dependent properties of colloidal drug delivery systems and their ability to trap phytochemicals in nanocarriers, both increased efficacy and stability can be achieved while offering controlled and site-specific drug delivery. When an active molecule, in this case, flavonoids, is embedded or entrapped in the matrix of nanocarriers, controlled and sustained release profiles can be obtained and prolongation of the systemic circulation lifetime of the molecule can be achieved. In addition, significant improvements in the pharmacokinetics and therapeutic index of the flavonoid can be achieved in the presence of a colloidal carrier system. One of the effective parameters here is particle size and so the increased surface area.

The colloidal size causes an increase in interaction with the biological environment and provides ease of overcoming many biological barriers. Furthermore, the safety and effectiveness of the phytochemical can be increased by reducing the dose and thus its side effects. By another way of explanation, colloidal drug delivery systems have been developed for the purpose of obtaining maximum therapeutic efficiency with minimum side effects for phytochemicals that are insufficient in treatment alone. Both topical and systemic applications such as oral, nasal, dermal, parenteral or ocular routes can be used for the bioactive molecule. As a result, colloidal carrier systems and nanomedicine offer a new perspective on therapeutic quality for various flavonoids, especially rhoifolin, as well as patient compliance (Jeevanandam et al., 2018[81]). Rhoifolin is a type of flavone glycoside belonging to the apigenin family known also as “apigenin 7-O-neohesperidoside” and preclinical studies have demonstrated that rhoifolin exhibits many potent therapeutical activities such as anti-inflammatory, antioxidant, antibacterial, antiviral (Eldahshan, 2013[45], Negm et al., 2022[125]); anticancer (Eldahshan, 2013[45], Zheng et al., 2022[205]); antidiabetic (Brinza et al., 2020[27]); hepatoprotective (Refaat et al., 2015[146]); antirheumatic (Peng et al., 2020[132]) properties. Despite the potential therapeutic efficacy of rhoifolin or similar phenolic compounds, their easy degradation by environmental stress and low water solubility are the main limitations of their application as drugs. Considering the aforementioned limitations, many colloidal drug delivery system studies have been carried out on fully characterized flavonoids such as apigenin and naringin. In a recent study, rhoifolin isolated from Jordanian Teucrium polium, then rhoifolin-loaded poly(lactide-co-glycolide)-PLGA nanoparticles were obtained by single emulsion (O/W) solvent evaporation technique (Al-Shalabi et al., 2022[10]). Optimum polymeric nanoparticle formulation was selected by evaluating the loading capacity results after pharmaceutical development studies and tannic acid-mediated surface modification was carried out with poly(ethylene glycol)-PEG. While optimum rhoifolin-loaded PLGA nanoparticles have a particle size of 182 ± 8 0 nm, a PDI value of 0.15 ± 0.01, −27 ± 8 mV surface charge, and an encapsulation efficiency percentage of 45.0 ± 4.3 %, after PEGylation process; particle size, PDI and zeta potential were measured as 204 ± 2 nm, 0.14 ± 0.02 and −28 ± 3 mV, respectively. One of the most important data obtained in this study is that the surface modification process of rhoifolin-loaded PLGA nanoparticles with hydrophilic polymer-PEG does not cause a statistically significant change in the physicochemical properties of these nanoparticles. It has been emphasized that the advantage of the PEGylation process is to increase the water solubility of rhoifolin-loaded polymeric nanoparticles, as well as to prolong their systemic circulation, reduce their immunegenicity and reduce their accumulation in reticuloendothelial system. On the other hand, surface modified nanoparticles maintained their colloidal stability both during the storage stage and in the presence of serum. Another remarkable finding obtained in the study was rhoifolin nanoparticles' antioxidant and free radical scavenging capacity. Cell-based assay with RAW 264.7 murine macrophage cells as well as the in vitro cell-free ABTS assay results show that it exhibits enhanced antioxidant activity and cellular uptake when bioactive flavonoid is entrapped into nanoparticles. In vivo efficacy tests with in vivo paw edema test and histopathological studies in rats confirmed the role of rhoifolin-loaded polymeric nanoparticles in alleviating paw edema and also confirmed that both free rhoifolin and rhoifolin-loaded polymeric nanoparticles possessed potent anti-inflammatory activity.

Limitations

The current research on the anticancer properties of rhoifolin, while promising, is hindered by several significant limitations and clinical gaps that must be addressed to facilitate its advancement into clinical use. Primarily, the majority of studies have been conducted in vitro or in animal models, with a noticeable lack of clinical trials evaluating rhoifolin's safety, efficacy, and pharmacokinetics in human subjects. This absence of clinical data significantly impedes the translation of preclinical findings into practical therapeutic applications. Additionally, rhoifolin's poor water solubility and low bioavailability present substantial challenges for its clinical application. Although recent developments in nanotechnology-based delivery systems have shown potential in enhancing its bioavailability, further research is necessary to optimize these systems and validate their effectiveness in humans. Moreover, the variability in rhoifolin concentration depending on plant sources and environmental conditions poses challenges for the standardization of its extracts, which is fundamental for consistent therapeutic outcomes. The current understanding of the molecular mechanisms underlying rhoifolin's anticancer activity remains incomplete, with only a partial elucidation of its pathways, limiting the ability to develop targeted and effective therapies. Furthermore, there is a lack of comprehensive toxicological studies, which are essential to determine the safe dosage ranges and potential side effects of rhoifolin, particularly for its use in long-term cancer treatments. Another significant gap is the limited exploration of rhoifolin in combination with other anticancer agents, which could potentially enhance or synergize with existing treatments, offering new therapeutic strategies. Finally, the development of rhoifolin as a viable therapeutic agent faces regulatory challenges due to the limited clinical data and the need for standardized formulations, which necessitates coordinated efforts between researchers, clinicians, and regulatory authorities. Addressing these limitations through rigorous research and clinical trials is essential to fully realize the therapeutic potential of rhoifolin in oncology.

Conclusion

Rhoifolin, apigenin-7-O-neohesperidoside is a flavonoid derivative that is found in many plant species belonging to various plant families. Within these plant species, we can see that the compound is not limited to certain plant parts, and thus, can be found in every part of different species such as seeds, flowers, roots, leaves, fruits, and stems. Having been found in hundreds of plant species in varying quantities, this flavonoid has important bioactivities which deserve more research, especially related to its anticancer activity since novel compounds with low side effects are constantly being sought for a couple of decades. While rhoifolin is an important candidate for the development of anticancer drugs, its low bioavailability due to low water solubility and easy degradation by environmental stress limits its usage for medicinal purposes. This difficulty might be overcome in the future with the help of biotechnology studies performed with nanoparticles in which the compound could be encapsulated in, thus more studies related to anticancer activity focusing on increasing the bioavailability of rhoifolin are needed.

Notes

Jesús Herrera-Bravo, Javad Sharifi-Rad (Universidad Espíritu Santo, Samborondón 092301, Ecuador; E-mail: javad.sharifirad@gmail.com) and Daniela Calina (Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; E-mail: calinadaniela@gmail.com) contributed equally as corresponding author.

Declaration

Competing interests

The authors wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.

Funding

Not applicable.

 

References

1. Abbas MA, Al-Najjar BO, Jaffal SM. Analgesic property of loranthus acaciae studied by molecular docking and biological assays. Sys Rev Pharm. 2021;12:712-9
2. Abd Elhameid M, Shabana MM, Abdel-Sattar E, Salama MM, Sleem AA. Phytochemical and biological investigation of Leptadenia heterophylla Decne. Fruits and its latex. Bull Fac Pharm Cairo Univ. 2006;44:59-74
3. Abiri R, Abdul-Hamid H, Sytar O, Abiri R, de ALmeida EB, Sharma SK, et al. A brief overview of potential treatments for viral diseases using natural plant compounds: the case of SARS-Cov. Molecules. 2021;26(13):3868
4. Abou Baker DH. An ethnopharmacological review on the therapeutical properties of flavonoids and their mechanisms of actions: A comprehensive review based on up to date knowledge. Toxicol Rep. 2022;9:445-69
5. Adhikari B, Marasini BP, Rayamajhee B, Bhattarai BR, Lamichhane G, Khadayat K, et al. Potential roles of medicinal plants for the treatment of viral diseases focusing on COVID-19: A review. Phytother Res. 2021;35:1298-312. doi: 10.1002/ptr.6893
6. Agogbua JU, Okonwu K, Akonye LA, Mensah SI. Phytochemicals of Telfairia occidentalis leaf grown in urea solutions. Res J Phytochem. 2022;16:65-73
7. Ahn M, Moon C, Park C, Kim J, Sim K-B, Shin T. Transient activation of an adaptor protein, disabled-2, in rat spinal cord injury. Acta Histochem. 2015;117:56-61
8. Alfarisi H, Sa'diah S, Wresdiyati T. Polyphenol profile, antioxidant and hypoglycemic activity of Acalypha hispida leaf extract. Indian J Pharm Sci. 2020;82:291-9
9. Alreshidi M, Noumi E, Bouslama L, Ceylan O, Veettil VN, Adnan M, et al. Phytochemical screening, antibacterial, antifungal, antiviral, cytotoxic, and anti-quorum-sensing properties of Teucrium polium L. aerial parts methanolic extract. Plants (Basel). 2020;9(11):1418. doi: 10.3390/plants9111418
10. Al-Shalabi E, Abusulieh S, Hammad AM, Sunoqrot S. Rhoifolin loaded in PLGA nanoparticles alleviates oxidative stress and inflammation in vitro and in vivo. Biomater Sci. 2022;10:5504-19
11. Alshehri MM, Quispe C, Herrera-Bravo J, Sharifi-Rad J, Tutuncu S, Aydar EF, et al. A review of recent studies on the antioxidant and anti-infectious properties of senna plants. Oxid Med Cell Longevity. 2022;2022:6025900. doi: 10.1155/2022/6025900
12. Amoroso VB, Mendez RA, Junio HA, Molino RJEJ, Pescadero IR, Villalobos AP. Characterization of a natural fungicide from an indigenous plant Tasmannia piperita (Hook.f.) Miers extract: stability, metabolomics, and in silico studies. Philippine J Sci. 2021;150:355-70
13. Andrade-Pinheiro JC, de Souza CES, Ribeiro DA, Fonseca VJA, de Macedo DG, da Cruz RP, et al. LC-MS analysis and antifungal activity of Turnera subulata Sm. Plants. 2023;12(2):415
14. Aoki C, Takeuchi Y, Higashi K, Okamoto Y, Nakanishi A, Tandia M, et al. Structural elucidation of a novel transglycosylated compound α-glucosyl rhoifolin and of α-glucosyl rutin by NMR spectroscopy. Carbohydr Res. 2017;443:37-41
15. Awad HM, Abd-Alla HI, Mahmoud KH, El-Youmy SA. In vitro anti-nitrosative, antioxidant, and cytotoxicity activities of plant flavonoids: a comparative study. Med Chem Res. 2014;23:3298-307
16. Azzini E, Peña-Corona SI, Hernández-Parra H, Chandran D, Saleena LAK, Sawikr Y, et al. Neuroprotective and anti-inflammatory effects of curcumin in Alzheimer's disease: Targeting neuroinflammation strategies. Phytother Res. 2024;38:3169-89. doi: 10.1002/ptr.8200
17. Badshah SL, Faisal S, Muhammad A, Poulson BG, Emwas AH, Jaremko M. Antiviral activities of flavonoids. Biomed Pharmacother. 2021;140:111596
18. Balasuriya BWN, Rupasinghe HPV. Plant flavonoids as angiotensin converting enzyme inhibitors in regulation of hypertension. Funct Foods Health Dis. 2011;1:172-88
19. Barberis A, Deiana M, Spissu Y, Azara E, Fadda A, Serra PA, et al. Antioxidant, antimicrobial and other biological properties of pompia juice. Molecules. 2020;25(14):3186
20. Barras A, Mezzetti A, Richard A, Lazzaroni S, Roux S, Melnyk P, et al. Formulation and characterization of polyphenol-loaded lipid nanocapsules. Int J Pharm. 2009;379:270-7
21. Barreca D, Bellocco E, Caristi C, Leuzzi U, Gattuso G. Elucidation of the flavonoid and furocoumarin composition and radical-scavenging activity of green and ripe chinotto (Citrus myrtifolia Raf.) fruit tissues, leaves and seeds. Food Chem. 2011;129:1504-12
22. Barreca D, Bellocco E, Caristi C, Leuzzi U, Gattuso G. Flavonoid profile and radical-scavenging activity of Mediterranean sweet lemon (Citrus limetta Risso) juice. Food Chem. 2011;129:417-22. doi: 10.1016/j.foodchem.2011.04.093
23. Barreca D, Bellocco E, Caristi C, Leuzzi U, Gattuso G. Kumquat (Fortunella japonica Swingle) juice: Flavonoid distribution and antioxidant properties. Food Res Int. 2011;44:2190-7
24. Benarba B, Pandiella A. Medicinal plants as sources of active molecules against COVID-19. Front Pharmacol. 2020;11:1189
25. Benavente-Garcia O, Castillo J. Update on uses and properties of Citrus flavonoids: new findings in anticancer, cardiovascular, and anti-inflammatory activity. J Agric Food Chem. 2008;56:6185-205
26. Bindu TK, Udayan PS. LC-MS profiling of methanolic extract of Pueraria tuberosa (Roxb. Ex Willd.) DC. tubers. Int J Horticult Agric Food Sci (IJHAF). 2018;2:190-8
27. Brinza I, Abd-Alkhalek AM, El-Raey MA, Boiangiu RS, Eldahshan OA, Hritcu L. Ameliorative effects of rhoifolin in scopolamine-induced amnesic zebrafish (Danio rerio) model. Antioxidants (Basel). 2020;9(7):580. doi: 10.3390/antiox9070580
28. Budak GG, Özkan S, Budak M, Şeker T, Meryemoğlu B, Miran ŞS, et al. Preclinical toxicity test results of a new antiviral-immune modulator compound consisting of flavonoid molecules (COVID-19 clinical trial preliminary data). Explor Med. 2022;3:1-21
29. Buford TW. Hypertension and aging. Ageing Res Rev. 2016;26:96-111
30. Burlando B, Pastorino G, Salis A, Damonte G, Clericuzio M, Cornara L. The bioactivity of Hedysarum coronarium extracts on skin enzymes and cells correlates with phenolic content. Pharm Biol. 2017;55:1984-91. doi: 10.1080/13880209.2017.1346691
31. Cai Z, Liao H, Wabg C, Chen J, Tan M, Mei Y, et al. A comprehensive study of the aerial parts of Lonicera japonica Thunb. Based on metabolite profiling coupled with PLS-DA. Phytochem Anal. 2020;31:786-800
32. Casabuono AC, Pomilio AB. Flavonoids of Festuca argentina. Fitoterapia. 1990;61:284-5
33. Chen H, Qin J, Shi H, Li Q, Zhou S, Chen L. Rhoifolin ameliorates osteoarthritis via the Nrf2/NF-κB axis: in vitro and in vivo experiments. Osteoarthritis Cartilage. 2022;30:735-45. doi: 10.1016/j.joca.2022.01.009
34. Chen L, Sun J, Pan Z, Lu Y, Wang Z, Yang L, et al. Analysis of chemical constituents of Chrysanthemum morifolium extract and its effect on postprandial lipid metabolism in healthy adults. Molecules. 2023;28(2):579
35. Chen Q, Wang Y, Ma F, Han M, Wang Z, Xue P, et al. Systematic profiling of the effective ingredients and mechanism of Scabiosa comosa and S. tschilliensis against hepatic fibrosis combined with network pharmacology. Sci Rep. 2021;11(1):2600
36. Cheng L, Ren Y, Lin D, Peng S, Zhong B, Ma Z. The anti-inflammatory properties of Citrus wilsonii Tanaka extract in LPS-induced RAW 264.7 and primary mouse bone marrow-derived dendritic cells. Molecules. 2017;22(7):1213. doi: 10.3390/molecules22071213
37. Cicero AF, Fogacci F, Colletti A. Food and plant bioactives for reducing cardiometabolic disease risk: an evidence based approach. Food Funct. 2017;8:2076-88
38. Dai TD, Thien DD, Tam NT. Flavone glycosides from Uraria crinite. Vietnam J Sci Technol Engin. 2021;63 (2):3-6
39. Deng M, Jia X, Dong L, Liu L, Huang F, Chi J, et al. Structural elucidation of flavonoids from Shatianyu (Citrus grandis L. Osbeck) pulp and screening of key antioxidant components. Food Chem. 2022;366:130605
40. Dhanabal SP, Syamala G, Kumar MNS, Suresh B. Hepatoprotective activity of the Indian medicinal plant Polygala arvensis on D-galactosamine-induced hepatic injury in rats. Fitoterapia. 2006;77:472-4
41. Dores AR, Peixoto M, Castro M, Sá C, Carvalho IP, Martins A, et al. Knowledge and beliefs about herb/supplement consumption and herb/supplement-drug interactions among the general population, including healthcare professionals and pharmacists: a systematic review and guidelines for a smart decision system. Nutrients. 2023;15(10):2298. doi: 10.3390/nu15102298
42. Dormousoglou M, Boti V, Hela D, Vlastos V, Antonopoulou M, Chondrogiannis C, et al. Beneficial properties of Drimia numidica leaf nethanolic extract against the cytogenotoxic effects of mitomycin C on human lymphocytes. Food Chem Toxicol. 2023;173:113626
43. Egger K, Keil M. [Flavoneglycosides in the flowers of Paeonia arborea and P. suffruticosa]. Planta. 1969;88:154-6. doi: 10.1007/bf01391121
44. Ekeke C, Obute GC, Ogazie CA. HPLC evaluation of phenolic compounds in Physalis angulata Linn. And Physalis micrantha Linn. (Solanaceae). Eur J Med Plants. 2019;29(2):1-9
45. Eldahshan OA. Rhoifolin;A potent antiproliferative effect on cancer cell lines. Brit J Pharm Res. 2013;3:46-53. doi: 10.9734/bjpr/2013/1864
46. Eldahshan OA, Azab SS. Anti-inflammatory effect of apigenin-7-neohesperidoside (rhoifolin) in carrageenin-induced rat oedema model. J Appl Pharm Sci. 2012;2(8):74-9
47. Elma SN, Rosli D, Badarusham K, Salvamani S, Hassan MS. Effect of phytochemicals in Phoenix dactylifera L. on human body using LC-QTOF-MS. Ind J Public Health Res Dev. 2019;10:760-4
48. El-Shawi OE, Eldahshan OA. Protective effect of rhoifolin on gamma irradiation induced cardiac dysfunctions in albino mice. Arab J Nucl Sci Appl. 2014;47:198-207
49. El-Shawy OE. Rhoifolin alleviates gamma-irradiation induced toxicity in whole body irradiated swiss albino mice (biochemical and haematological studies). Middle East J Appl Sci. 2014;4:365-72
50. Eroğlu Özkan E, Boğa M, Yılmaz MA, Mataracı Kara E, Yeşil Y. LC-MS/MS analyses of Ziziphora clinopodioides Lam. from Turkey: antioxidant, anticholinesterase, antimicrobial and anticancer activities. İstanbul J Pharm. 2019;50(1):33-41
51. Ezzat SM, Merghany RM, Baki PMA, Abdelrahim NA, Osman SM, Salem MA, et al. Nutritional sources and anticancer potential of phenethyl isothiocyanate: molecular mechanisms and therapeutic insights. Mol Nutr Food Res. 2024;68(8):2400063. doi: 10.1002/mnfr.202400063
52. Fahim JR, Abd El-Moneim Hegazi G, El-Said Abo El-Fadl R, Abd Almegid Abd Alhady MR, Desoukey SY, Ramadan MA, et al. Production of rhoifolin and tiliroside from callus cultures of Chorisia chodatii and Chorisia speciosa. Phytochem Lett. 2015;13:218-27
53. Fan R, Zhu C, Qiu D, Zeng J. Comparison of the bioactive chemical components and antioxidant activities in three tissues of six varieties of Citrus grandis ‘Tomentosa’ fruits. Int J Food Prop. 2019;22:1848-62
54. Fenglin H, Ruili L, Bao H, Liang M. Free radical scavenging activity of extracts prepared from fresh leaves of selected Chinese medicinal plants. Fitoterapia. 2004;75:14-23
55. Ferreira M, Costa D, Sousa A. Flavonoids-based delivery systems towards cancer therapies. Bioengineering (Basel). 2022;9(5):197
56. Flores-Tellez TNJ, Lopez TV, Garzon VRV, Villa-Trevino S. Co-expression of Ezrin-CLIC5-Podocalyxin is associated with migration and invasiveness in hepatocellular carcinoma. PLOS One. 2015;10(7):e0131605
57. Gattuso G, Caristi C, Gargiulli C, Bellocco E, Toscano G, Leuzzi U. Flavonoid glycosides in bergamot juice (Citrus bergamia Risso). J Agric Food Chem. 2006;54:3929-35. doi: 10.1021/jf060348z
58. Ge YB. [Chemical constituents of Fortunella margarita fruits]. Zhong Yao Cai. 2014;37:435-8
59. Ghasemian M, Owlia S, Owlia MB. Review of anti-inflammatory herbal medicines. Adv Pharmacol Sci. 2016;2016:9130979
60. Giambanelli E, D'Antuono LF, Ferioli F, Frenich AG, Romero-Gonzalez R. Sesquiterpene lactones and inositol 4-hyrdoxyphenylacetic acid derivatives in wild edible leafy vegetables from Central Italy. J Food Composition Anal. 2018;72:1-6
61. Goldring MB, Goldring SR. Articular cartilage and subchondral bone in the pathogenesis of osteoarthritis. Ann N Y Acad Sci. 2010;1192:203-37
62. Gorniak I, Bartoszewski R, Kroliczewski J. Comprehensive review of antimicrobial activities of plant flavonoids. Phytochem Rev. 2019;18:241-72
63. Grodzicki W, Dziendzikowska K. The role of selected bioactive compounds in the prevention of Alzheimer’s disease. Antioxidants (Basel). 2020;9(3):229
64. Guerrero L, Castillo J, Quiñones M, Garcia-Vallvé S, Arola L, Pujadas G, et al. Inhibition of angiotensin-converting enzyme activity by flavonoids: structure-activity relationship studies. PLoS One. 2012;7(11):e49493. doi: 10.1371/journal.pone.0049493
65. Guetchueng ST, Nahar L, Ritchie KJ, Ismail FMD, Dempster NM, Nnanga EN, et al. Phenolic compounds from the leaves and stem bark of Pseudospondias microcarpa (A. Rich.) Engl. (Anacardiaceae). Biochem System Ecol. 2020;91:104078
66. Hafez SA, Abdel Ghani AE, El Shazly AM. Pharmacognostical and antibacterial studies of Chorisia speciosa A.St.Hill flower (Bombaceae). Mansoura J Pharm Sci. 2003;19(1):40-59
67. Han F, Zhang Q, Ding R, Wang J, Wu H, Zhao A. Relative quantification of phenolic compounds in exocarp-mesocarp and endocarp of sumac (Toxicodenderon vernicifluum) combined with transcriptone analysis provides insights into glycosylation of flavonoids and biflavonoid biosynthesis. Plant Physiol Biochem. 2023;195:275-87
68. Hanamura S, Hanaya K, Shoji M, Sugai T. Synthesis of acacetin and resveratrol 3,5-di-O-β-glucopyranoside using lipase-catalyzed regioselective deacetylation of polyphenol glycoside peracetates as the key step. J Mol Catalysis B: Enzymatic. 2016;128:19-26
69. Hasibuan PAZ, Harahap U, Sitorus P, Satria D. The anticancer activities of Vernonia amygdalina Delile. Leaves on 4T1 breast cancer cells through phosphoinositide 3-kinase (PI3K) pathway. Heliyon. 2020;6(7):e04449. doi: 10.1016/j.heliyon.2020.e04449
70. Hattori S, Shimokoriyama M, Kanao M. Studies on flavanone glycosides. IV. The glycosides of ripe fruit peel and flower petals of Citrus aurantium L. J Am Chem Soc. 1952;74:3614-5
71. He ZD, Lau KM, But PP, Jiang RW, Dong H, Ma SC, et al. Antioxidative glycosides from the leaves of Ligustrum robustum. J Nat Prod. 2003;66:851-4. doi: 10.1021/np020568g
72. Hench LL. Joint replacement. In: Henh LL, Jones JR (eds). Biomaterials, artifical organs and tissue engineering (pp 379-410). Sawston, UK: Woodhead Publishing, 2019
73. Hong LTT, Huyen TT, Tri LM, Hien TD, Loi H. New iridoid from Valeriana hardwickii Wall. Vietnam Journal of Chemistry. 2021;59(1):12-6. doi: 10.1002/vjch.202000073
74. Hotchkiss RS, Moldawer LL, Opal SO, Reinhart K, Turnbull IR, Vincent J-L. Sepsis and septic shock. Nat Rev Dis Primers. 2016;2:16045
75. Huang Q, Guo Y, Fu R, Peng T, Zhang Y, Chen F. Antioxidant activity of flavonoids from leaves of Jatropha curcas. SciAsia. 2014;40:193-7
76. Huang X, Wu S, Wei Y, Meng S, Jiang R. Effect of the plant flavonoid, rhoifolin, on memory and cognition in a rat model of Alzheimer’s disease. Trop J Pharm Res. 2021;20:1481-7
77. Huo K, Chen Y, Sui Y, Wang Y, Fu Y, Pei X, et al. Transient expression and enzymatic assay identified uridine-diphosphate glucosyltransferases related to flavonoid glycosylation in Vernonia amygdalina leaves. Industr Crops Prod. 2021;172:114005
78. Huynh L, Tran H, Bacher M, Pacher T. Iridoids and flavonoids from Valeriana hardwickii Wall. J Pharmacogn Phytochem. 2016;5:245-9
79. Iqbal MJ, Kabeer A, Abbas Z, Siddiqui HA, Calina D, Sharifi-Rad J, et al. Interplay of oxidative stress, cellular communication and signaling pathways in cancer. Cell Commun Signal. 2024;22(1):7. doi: 10.1186/s12964-023-01398-5
80. Jaiswal Y, Weber D, Yerke A, Xue Y, Lehman D, Williams T, et al. A substitute variety for agronomically and medicinally important Serenoa repens (saw palmetto). Sci Rep. 2019;9(1):4709
81. Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050-74
82. Jo S, Kim S, Shin DH, Kim M-S. Inhibition of SARS-CoV 3CL protease by flavonoids. J Enzyme Inhibit Med Chem. 2019;35:145-51
83. Juee LYM. Phytochemical characterization and mushroom tyrosinase inhibition of different extracts from Salvia officinalis L. leaves. J Pharm Pharmacogn Res. 2022;10:605-15
84. Kanao M, Matsuda H. Studies on the constituents of the leaves of Lespedeza thunbergii (DC.) Nakai, I. The structure of miyaginin. Yakugaku Zasshi. 1978;98:366-8
85. Kaneko T, Sakamoto M, Ohtani K, Ito A, Kasai R, Yamasaki K, et al. Secoiridoid and flavonoid glycosides from Gonocaryum calleryanum. Phytochemistry. 1995;39:115-20
86. Kanes K, Tisserat B, Berhow M, Vandercook C. Phenolic composition of various tissues of Rutaceae species. Phytochem Lett. 1993;32:967-74
87. Kaul R, Paul P, Kumar S, Büsselberg D, Dwivedi VD, Chaari A. Promising antiviral activities of natural flavonoids against SARS-CoV-2 targets: systematic review. Int J Mol Sci. 2021;22(20):11069
88. Kayahan S, Saloğlu D. Investigation of bioactive components of sakiz artichoke (Cynara scolymus L.) consumed in different maturity. J Adv Res Nat Appl Sci. 2022;8:105-13
89. Khan T, Khan MA, Karam K, Ullah N, Mashwani Z-u-R, Nadhman A. Plant in vitro culture technologies;a promise into factories of secondary metabolites against COVID-19. Front Plant Sci. 2021;12:610194
90. Kiem PV, Mai NT, Minh CV, Khoi NH, Dang NH, Thao NP, et al. Two new c-glucosyl benzoic acids and flavonoids from Mallotus nanus and their antioxidant activity. Arch Pharm Res Vol. 2010;2:203-8
91. Koch A, Tamez P, Pezzuto J, Soejarto D. Evaluation of plants used for antimalarial treatment by the Maasai of Kenya. J Ethnopharmacol. 2005;101:95-9
92. Kraus VB, Blanco FJ, Englund M, Karsdal MA, Lohmander LS. Call for standardized definitions of osteoarthritis and risk stratification for clinical trials and clinical use. Osteoarthritis Cartilage. 2015;23:1233-41
93. Kumar JK, Rao MS, Rao PS, Toth G, Balazs B, Duddeck H. Flavone glycosides from Polygala telephoides and polygala arvensis. Nat Prod Lett. 1999;14 (1):35-8
94. Kuo PC, Liao YR, Hung HY, Chuang CW, Hwang TL, Huang SC, et al. Anti-Inflammatory and neuroprotective constituents from the peels of Citrus grandis. Molecules. 2017;22(6):967. doi: 10.3390/molecules22060967
95. Lee EJ, Kim JS, Kim HP, Lee J-H, Kang SS. Phenolic constituents from the flower buds of Lonicera japonica and their 5-lipoxygenase inhibitory activities. Food Chem. 2010;120:134-9
96. Lefort EC, Blay J. The dietary flavonoid apigenin enhances the activities of the anti-metastatic protein CD26 on human colon carcinoma cells. Clin Exp Metastasis. 2011;28:337-49
97. Liang Y, Huang Z, Chen H, Zhang T, Ito Y. Preparative isolation and purification of two closely related glycosidic flavonoids from Exocarpium Citri Grandis by high-speed countercurrent chromatography. J Liquid Chromatogr Related Technol. 2007;30:419-30
98. Liao S, Song F, Feng W, Ding X, Yao J, Song H, et al. Rhoifolin ameliorates titanium particle-stimulated osteolysis and attenuates osteoclastogenesis via RANKL-induced NF-κB and MAPK pathways. J Cell Physiol. 2019;234:17600-11. doi: 10.1002/jcp.28384
99. Lin C-C, Yen M-H, Lo T-s, Lin J-M. Evaluation of the hepatoprotective and antioxidant activity of Boehmeria nivea var. nivea and B. nivea var. tenacissima. J Ethnopharmacol. 1998;60:9-17
100. Lin L-d, Qin G-w, Xu R-s. Studies on chemical constituents of Ilex centrochinensis. Acta Botan Sin. 1994;36:393-7
101. Lin Y, Li S, Chen T, Lin Y, Cheng Z, Ni L, et al. Phytochemical compositions and biological activities of the branches and leaves of Ormosia hosiei Hemsl. et Wils. J Pharm Biomed Anal. 2023;226:115238
102. Liu J-q, Ren X-j, Shu, J-c., Zhang R, Zhang W, Pan J-h. Simultaneous determination of five flavonoids in Pteris multifida Poir. By HPLC. Chem Industry Forest Prod. 2013;33:117-20
103. Liu L, Wang Y, Zhang J, Wang S. Advances in the chemical constituents and chemical analysis of Ginkgo biloba leaf extract, and phytopharmaceuticals. J Pharm Biomed Anal. 2021;193:113704
104. Liu L, Xie P, Li W, Wu Y, An W. Augmenter of liver regeneration protects against ethanol-induced acute liver injury by promoting autophagy. Am J Pathol. 2019;189:552-67
105. Liu MH, Liu HB, Su WW, Liu XR, Li JH. Study on impurities in naringin extracted from Citrus grandis 'tomentosa'. Zhong Yao Cai. 2012;35(1):56-61
106. Liu Y, Wang C, Wu J, Tan L, Gao P, Wu S, et al. Study on the comprehensive phytochemicals and the anti-ulcerative colitis effect of Saussurea pulchella. Molecules. 2023;28(4):1526
107. Loftsson T. Physicochemical properties and pharmacokinetics. In: Loftsson T (ed). Essential pharmacokinetics: a primer for pharmaceutical scientists (pp 85-104). Amsterdam: Elsevier, 2015
108. Long K, Du X, Yan Z, Xu J. Rhoifolin provides neuroprotection against spinal cord injury by attenuation of oxidative stress, inflammation and apoptosis in rats. Indian JAnim Res. 2023;57(1):82-9
109. Lopez-Gutierrez N, Romero-Gonzalez R, Plaza-Bolanos P, Vidal JLM, Frenich AG. Identification and quantification of phytochemicals in nutraceutical products from green tea by UHPLC-Orbitrap-MS. Food Chem. 2015;173:607-18
110. Lopez-Gutierrez N, Romero-Gonzalez R, Vidal JLM, Frenich AG. Quality control evaluation of nutraceutical products from Ginkgo biloba using liquid chromatography coupled to high resolution mass spectrometry. J Pharm Biomed Anal. 2016;121:151-60
111. Lou SN, Lai YC, Hsu YS, Ho CT. Phenolic content, antioxidant activity and effective compounds of kumquat extracted by different solvents. Food Chem. 2016;197(Pt A):1-6. doi: 10.1016/j.foodchem.2015.10.096
112. Ma S-G, Wang R-B, Li W-R, Liu Y-B, Qu J, Li Y, et al. New C-2 diastereomers of flavanone glycosides conjugated with 3-hydroxy-3-methylglutaric acid from the pericarp of Citrus grandis (L.) Osbeck. Bioorg Chem. 2018;80:519-24
113. Ma S-G, Yuan S-P, Hou Q, Li Y, Chen X, Yu S-S. Flavonoid glycosides from leaves of Turpinia argute and their anti-inflammatory activity. Zhongguo Zhong Yao Za Zhi. 2013;38:1747-50
114. Ma Y-C, Zhang M, Xu M-T, Feng S-X, Lei M, Yi B. Chemical constituents from Callicarpa nudiflora and the cytotoxic activities. China J Chin Mater Med. 2014;24:3094-101
115. Ma YC, Zhang M, Xu WT, Feng SX, Lei M, Yi B. [Chemical constituents from Callicarpa nudiflora and their cytotoxic activities]. Zhongguo Zhong Yao Za Zhi. 2014;39:3094-101
116. Madrigal-Santillan E, Madrigal-Bujaidar E, Alvarez-Gonzalez I, Sumaya-Martinez MT, Gutierrez-Salinas J, Bautista M, et al. Review of natural products with hepatoprotective effects. World J Gastroenterol. 2014;20:14787-804
117. Mahmood K, Zia KM, Zuber M, Salman M, Anjum MN. Recent developments in curcumin and curcumin based polymeric materials for biomedical applications: A review. Int J Biol Macromol. 2015;81:877-90
118. Mai B, Han L, Zhong J, Shu J, Cao Z, Fang J, et al. Rhoifolin alleviates alcoholic liver disease in vivo and in vitro via inhibition of the TLR4/NF-κB signaling pathway. Front Pharmacol. 2022;13:878898. doi: 10.3389/fphar.2022.878898
119. Maity S, Acharyya A, Chakraborti AS. Flavonoid-based polymeric nanoparticles: A promising approach for cancer and diabetes treatment. Eur Polymer J. 2022;177:111455
120. McNagny KM, Hughes MR, Graves ML, DeBruin EJ, Snyder K, Cipollone J, et al. Podocalyxin in the diagnosis and treatment of cancer. In: Mohan R (ed). Advances in cancer management (pp 155-94). London: IntechOpen, 2012
121. Meliconi R, Pulsatelli L. Are mechanisms of inflammation joint-specific in osteoarthritis? Rheumatology. 2019;58:743-5
122. Mencherini T, Campone L, Piccinelli AL, Mesa MG, Sánchez DM, Aquino RP, et al. HPLC-PDA-MS and NMR characterization of a hydroalcoholic extract of Citrus aurantium L. var. amara peel with antiedematogenic activity. J Agric Food Chem. 2013;61:1686-93. doi: 10.1021/jf302815t
123. Nakajima VM, Macedo GA, Macedo JA. Citrus bioactive phenolics: role in the obesity treatment. LWT Food Sci Technol. 2014;59:1205-12
124. PubChem. Available from: https://pubchem.ncbi.nlm.nih.gov/
125. Negm WA, El-Kadem AH, Elekhnawy E, Attallah NGM, Al-Hamoud GA, El-Masry TA, et al. Wound-healing potential of rhoifolin-rich fraction isolated from sanguisorba officinalis roots supported by enhancing re-epithelization, angiogenesis, anti-inflammatory, and antimicrobial effects. Pharmaceuticals (Basel). 2022;15(2):178. doi: 10.3390/ph15020178
126. Neretina OV, Gromova AS, Lutskii VI. Dactylin and lonicierin from the aerial part of Hedysarum setigerum. Chem Nat Comp. 2005;41:602-3
127. Nijveldt RJ, van Nood E, van Hoorn DEC, Boelens PG, van Norren K, van Leeuwen PAM. Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin Nutr. 2001;74:418-25
128. Occhiuto F, Limardi F. Comparative effects of the flavonoids luteolin, apiin and rhoifolin on experimental pulmonary hypertension in the dog. Phytother Res. 1994;8:153-6
129. Okonwu K, Muonekwu JE. Potentials of underexploited seed of Trichocantes cucumerina Linn. J Appl Sci Environ Manage. 2019;23:791-7
130. Ozkan EE, Ersoy E, Canturk YY, Mataraci K, E., Cinar E, Sahin H, et al. The therapeutic potential of ethnomedicinally important Anatolian Thyme species: a phytochemical and biological assessment. Front Pharmacol. 2022;13:923063
131. Papalia T, Barreca D, Panuccio MR. Assessment of antioxidant and cytoprotective potential of Jatropha (Jatropha curcas) grown in Southern Italy. Int J Mol Sci. 2017;18(3):660. doi: 10.3390/ijms18030660
132. Peng S, Hu C, Liu X, Lei L, He G, Xiong C, et al. Rhoifolin regulates oxidative stress and proinflammatory cytokine levels in Freund’s adjuvant-induced rheumatoid arthritis via inhibition of NF-κB. Braz J Med Biol Res. 2020;53(6):e9489
133. Pernice R, Borriello G, Ferracane R, Borrelli RC, Cennamo F, Ritieni A. Bergamot: a source of natural antioxidants for functionalized fruit juices. Food Chem. 2009;112:545-50
134. Persia FA, Troncoso ME, Rinaldini E, Simirgiotis M, Tapia A, Borquez J, et al. UHPLC-Q/Orbitrap/MS/MS fingerprinting and antitumoral effects of Prosopis strombulifera (LAM.) BENTH. aqueous extract on allograft colorectal and melanoma cancer models. Heliyon. 2020;6(2):e03353
135. Plioukas M, Gabrieli C, Lazari D, Kokkalou E. Phytochemical analysis with the antioxidant and aldose reductase inhibitory capacities of Tephrosia humilis aerial parts' extracts. Nat Prod Res. 2016;30:1366-72. doi: 10.1080/14786419.2015.1057729
136. Popescu M, Terzea DC, Carsote M, Ghenea AE, Costache A, Popescu IAS, et al. COVID-19 infection: from stress-related cortisol levels to adrenal glands infarction. Rom J Morphol Embryol. 2022;63(1):39-48. doi: 10.47162/rjme.63.1.03
137. Prashar Y, Patel NJ. High-performance thin-layer chromatography analysis of gallic acid and other phytoconstituents of methanolic extracts of Myrica nagi fruit. Pharmacogn Res. 2020;12:95-101
138. Puglia C, Lauro MR, Tirendi GG, Fassari GE, Carbone C, Bonina F, et al. Modern drug delivery strategies applied to natural active compounds. Exp Opin Drug Deliv. 2017;14:755-68
139. Qi H, Liu L. Rhoifolin attenuates damage to hippocampal neuronal culture model of acquired epilepsy in vitro by regulating NF-κB/iNOS/COX-2 axis. Qual Assurance Saf Crops Foods. 2022;14:116-23
140. Qiaoyu L, Lingsheng D. Chemical study on the flower of Dolichos lablab L. J China Pharm Univ. 1996;27:205-7
141. Rahmouni F, Badraoui R, Ben-Nasr H, Bardakci F, Elkahoui S, Siddiqui AJ, et al. Pharmacokinetics and therapeutic potential of Teucrium polium against liver damage associated hepatotoxicity and oxidative injury in rats: computational, biochemical and histological studies. Life. 2022;12(7):1092
142. Rajkumar S, Jebanesan A. Bioactivity of flavonoid compounds from Poncirus trifoliata L. (Family: Rutaceae) against the dengue vector, Aedes aegypti L. (Diptera: Culicidae). Parasitol Res. 2008;104:19-25
143. Ramful D, Bahorun T, Bourdon E, Tarnus E, Aruoma OI. Bioactive phenolics and antioxidant propensity of flavedo extracts of Mauritian citrus fruits: Potential prophylactic ingredients for functional foods application. Toxicology. 2010;278:75-87
144. Rao YK, Lee M-J, Chen K, Lee Y-C, Wu W-S, Tzeng Y-M. Insulin-mimetic action of rhoifolin and cosmosiin isolated from Citrus grandis (L.) Osbeck leaves: enhanced adiponectin secretion and insulin receptor phosphorylation in 3T3-L1 cells. Evid Based Complement Alternat Med. 2011;2011:624375
145. Rasouli H, Hosseini-Ghazvini SMB, Khodarahmi R. Therapeutic potentials of the most studied flavonoids: highlighting antibacterial and antidiabetic functionalities. Stud Nat Prod Chem. 2019;60:85-122
146. Refaat J, Desoukey SY, Ramadan MA, Kamel MS. Rhoifolin: A review of sources and biological activities. Int J Pharmacogn. 2015;2:102-9
147. Refaat J, Samy MN, Desoukey SY, Ramadan MA, Sugimoto S, Matsunami K, et al. Chemical constituents from Chorisia chodatii flowers and their biological activities. Med Chem Res. 2015;24:2939-49. doi: 10.1007/s00044-015-1342-8
148. Russo M, Moccia S, Spagnuolo C, Tedesco I, Russo GL. Roles of flavonoids against coronavirus infection. Chem Biol Interact. 2020;328:109211
149. Sawatzky DA, Willoughby DA, Colville-Nash PR, Rossi AG. The involvement of the apoptosis-modulating proteins ERK 1/2, Bcl-xL and Bax in the resolution of acute inflammation in vivo. Am J Pathol. 2006;168:33-41
150. Sawikowska A. Meta-analysis of flavonoids with antiviral potential against coronavirus. Biometr Lett. 2020;57(1):13-22
151. Saxena HO, Mohan B, Kakkar A. Assessment of variation in rhoifolin content in aerial parts of Uraria picta Desv. from different locations of Madhya Pradesh. J Pharm Res. 2016;10:185-90
152. Saxena HO, Soni A, Mohammad N, Kakkar A, Singh N. HPLC analysis of rhoifolin in different plant parts of Uraria picta: a dashmool species. Indian J Trop Biodiv. 2014;22:199-201
153. Sayeed HM, Ahmed AS, Khallaf ISA, Qayed WS, Mohammed AF, Farghaly HSM, et al. Phytochemical investigation, molecular docking studies and DFT calculations on the antidiabetic and cytotoxic activities of Gmelina philippensis. J Ethnopharmacol. 2023;303:115938
154. Senizza B, Rocchetti G, Sinan KI, Zengin G, Mahomoodally MF, Glamocilja J, et al. The phenolic and alkaloid profiles of Solanum erianthum and Solanum torvum modulated their biological properties. Food Biosci. 2021;41:100974
155. Seukep AJ, Zhang Y-L, Xu Y-B, Guo M-Q. In vitro antibacterial and antiproliferative potential of Echinops lanceolatus Mattf. (Asteraceae) and identification of potential bioactive compounds. Pharmaceuticals. 2020;13(4):59
156. Sharifi-Rad J, Zhong J, Ayatollahi SA, Kobarfard F, Faizi M, Khosravi-Dehaghi N, et al. LC-ESI-QTOF-MS/MS characterization of phenolic compounds from Prosopis farcta (Banks & Sol.) J.F.Macbr. and their potential antioxidant activities. Cell Mol Biol. 2021;67 (1):189-200
157. Sharma E, Tewari M, Sati P, Sharma I, Attri DC, Rana S, et al. Serving up health: How phytochemicals transform food into medicine in the battle against cancer. Food Front. 2024;(5):1866-908. doi: 10.1002/fft2.439
158. Shen CY, Jiang JG, Huang CL, Zhu W, Zheng CY. Polyphenols from blossoms of citrus aurantium l. var. amara engl. show significant anti-complement and anti-inflammatory effects. J Agric Food Chem. 2017;65:9061-8. doi: 10.1021/acs.jafc.7b03759
159. Shimokoriyama M. The flavonoid glycosides in the leaves of Poncirus trifoliata. Bot Mag Tokyo. 1966;79:602-7
160. Silva ACO, Santana EF, Saraiva AM, Coutinho FN, Castro RHA, Pisciottano MNC, et al. Which approach is more effective in the selection of plants with antimicrobial activity? Evid Based Complement Alternat Med. 2013;2013:309980
161. Singh N, Baby D, Rajguru JP, Patil PB, Thakkannavar SS, Pujari VB. Inflammation and cancer. Ann Afr Med. 2019;18(3):121-6
162. Singh YD, Jena B, Ningthoujam R, Panda S, Priyadarsini P, Pattayanak S, et al. Potential bioactive molecules from natural products to combat against coronavirus. Adv Tradit Med. 2022;22:259-70
163. Sizemore S, Çiçek M, Sizemore N, Ng KP, Casey G. Podocalyxin increases the aggressive phenotype of breast and prostate cancer cells in vitro through its interaction with ezrin. Cancer Res. 2007;67:6183-91
164. Smith DA. Physicochemical properties in drug metabolism and pharmacokinetics. In: Dr. Han van de Waterbeemd BT, Gerd Folkers (eds). Computer-assisted lead finding and optimization: current tools for medicinal chemistry (pp 265-76). Zürich: Verlag Helvetica Chimica Acta, 1997
165. Son KH, Park JO, Chung KC, Chang HW, Kim HP, Lim JS, et al. Flavonoids from the aerial parts of Lonicera japonica. Arch Pharm Res. 1992;15:365-70
166. Song L, Xiong P, Zhang W, Hu H, Tang S, Jia B, et al. Mechanism of Citri reticulatae pericarpium as an anticancer agent from the perspective of flavonoids: a review. Molecules. 2022;27(17):5622
167. Song Y, Yan S, Zhang H, Liu R, An X, Zhang Q. Ameliorative effect of rhoifolin in cisplatin-induced nephrotoxicity via regulating nuclear factor-κb pathway. Curr Topics Nutraceut Res. 2020;18:266-72
168. Souilah N, Bendif H, Ullah Z, Hamel T, Djarri L, ÖZtürk M, et al. LC-MS/MS simultaneous determination of 37 bioactive compounds in Bunium crassifolium Batt. and its biological activities. J Res Pharm. 2021;25:450-63
169. Souilah N, Ullah Z, Bendif H, Medjroubi K, Hazmoune T, Hamel T, et al. Phenolic compounds from an Algerian endemic species of Hypochaeris laevigata var. hipponensis and investigation of antioxidant activities. Plants (Basel). 2020;9(4):514
170. Sruthi D, Dhanalakshmi M, Rao HCY, Parthasarathy R, Deepanraj SP, Jayabaskaran C. Curative potential of high-value phytochemicals on COVID-19 infection. Biochemistry (Moscow). 2023;88:64-72
171. Sultana B, Yaqoob S, Zafar Z, Bhatti HN. Escalation of liver malfunctioning: a step toward herbal awareness. J Ethnopharmacol. 2018;216:104-19
172. Suryawanshi JAS. An overview of Citrus aurantium used in treatment of various diseases. Afr J Plant Sci. 2011;5:390-5
173. Taheri A, Ganjeali A, Arefi-Oskouie A, Çirak C, Cheniany M. The variability of phenolic constituents and antioxidant properties among wild populations of Ziziphora clinopodioides Lam. Physiol Mol Biol Plants. 2023;29:221-37
174. Tallei TE, Tumilaar SG, Niode NJ, Fatimawali, Kepel BJ, Idroes R, et al. Potential of plant bioactive compounds as SARS-CoV-2 main protease (M(pro)) and Spike (S) glycoprotein inhibitors: a molecular docking study. Scientifica (Cairo). 2020;2020:6307457. doi: 10.1155/2020/6307457
175. Tanwar B, Modgil R. Flavonoids: Dietary occurrence and health benefits. Spatula DD. 2012;2(1):59-68
176. Thorpe TA. History of plant tissue culture. Mol Biotechnol. 2007;37:169-80
177. Tsujimoto T, Yoshitomi T, Maruyama T, Yamamoto Y, Hakamatsuka T, Uchiyama N. High-resolution liquid chromatography−mass spectrometry-based metabolomic discrimination of citrus-type crude drugs and comparison with nuclear magnetic resonance spectroscopy-based metabolomics. J Nat Prod. 2019;82:2116-23
178. Uddin N, Siddiqui BS, Begum S, Bhatti HA, Khan A, Parveen S, et al. Bioactive flavonoids from the leaves of Lawsonia alba (Henna). Phytochem Lett. 2011;4:454-8
179. Uysal S, Zengin G, Mahomoodally MF, Yilmaz MA, Aktumsek A. Chemical profile, antioxidant properties and enzyme inhibitory effects of the root extracts of selected Potentilla species. S Afr J Bot. 2019;120:124-8
180. Vadhana P, Singh BR, Bharadwaj M, Singh SV. Emergence of herbal antimicrobial drug resistance in clinical bacterial isolates. Pharm Anal Acta. 2015;6(10):1-7
181. Vega-Ruiz YC, Hayano-Kanashiro C, Gamez-Meza M, Medina-Juarez LA. Determination of chemical constituents and antioxidant activities of leaves and stems from Jatropha cinerea (Ortega) Müll. Arg. and Jatropha cordata (Ortega) Müll. Arg. Plants (Basel). 2021;10(2):212
182. Visalli G, Ferlazzo N, Cirmi S, Campiglia P, Gangemi S, Di Pietro A, et al. Bergamot juice extract inhibits proliferation by inducing apoptosis in human colon cancer cells. Anticancer Agents Med Chem. 2014;14:1402-13
183. Vo GT, Liu Z, Chou O, Zhong B, Barrow CJ, Dunshea FR, et al. Screening of phenolic compounds in Australian grown grapes and their potential antioxidant activities. Food Bioscience. 2022;47:101644
184. Wang D, Du N, Wen L, Zhu H, Liu F, Wang X, et al. An efficient method for the preparative isolation and purification of flavonoid glycosides and caffeoylquinic acid derivatives from leaves of lonicera japonica thunb. using high speed counter-current chromatography (HSCCC) and prep-hplc guided by DPPH-HPLC Experiments. Molecules. 2017;22(2):229. doi: 10.3390/molecules22020229
185. Wang D, Qi M, Yang Q, Tong R, Wang RB, Bligh SWA, et al. Comprehensive metabolite profiling of Plantaginis Semen using ultra high performance liquid chromatography time-of-flight tandem mass spectrometry coupled with elevated energy technique. J Separation Sci. 2016;39:1842-52
186. Wang Y, Liu Y. Neutrophil-induced liver injury and interactions between neutrophils and liver sinusoidal endothelial cells. Inflammation. 2021;44:1246-62
187. Wang Z-Y, Ning LI, Wang K-J. Chemical composition in fruit peel of Fortunella margarita. J Plant Resourc Environ. 2010;19(1):92-4
188. Wen S, Ma Z, Tripathi AS. Rhoifolin protects cecal ligation and puncture induced sepsis mice model by regulating inflammatory pathway. Microb Pathog. 2023;180:106112
189. WFO, World Flora Online. An online flora of all known plants 2023 [cited 2023]. Available from: https://www.worldfloraonline.org/
190. Xiao F, Deng T, Deng S, Deng C, Zhang, Xuqian LL. Effect of total flavonoids of Exocarpium Citri Grandis on apoptosis of hepatocytes in rats with alcoholic liver injury. J Guangdong Pharm Univ. 2012;28:316-8
191. Xie L, Fu Q, Shi S, Li JH, Zhou X. Rapid and comprehensive profiling of alpha-glucosidase inhibitors in Buddleja Flos by ultrafiltration HPLC-QTOF-MS/MS with diagnostic ions filtering strategy. Food Chem. 2021;344:128651
192. Xiong L, Lu H, Hu Y, Wang W, Liu R, Wan X, et al. In vitro anti-motile effects of rhoifolin, a flavonoid extracted from Callicarpa nudiflora on breast cancer cells via downregulating Podocalyxin-Ezrin interaction during Epithelial Mesenchymal Transition. Phytomedicine. 2021;93:153486
193. Yadav AK, Yadav D, Shanker K, Verma RK, Saxena AK, Gupta MM. Flavone glycoside based validated RP-LC method for quality evolution of Prishniparni (Uraria picta). Chromatographia. 2009;69:653-8
194. Yan J, Ni B, Sheng G, Zhang Y, Xiao Y, Ma Y, et al. Rhoifolin ameliorates osteoarthritis via regulating autophagy. Front Pharmacol. 2021;12:661072
195. Yang L, Zhang J, Zheng S, Hou A, Wang S, Yu H, et al. The phytochemistry, pharmacology and traditional medicinal use of Glechomae Herba - a systematic review. RSC Adv. 2021;11:19221-37
196. Yantih N, Syabillawati U, Mulatsari E, Sumaryono W. In silico screening of Ziziphus spina-christi (L.) Desf. And Strychnos ligustrine compounds as a protease inhibitor of SARS-COV-2. J Exp Biol Agric Sci. 2021;9:S208-S14
197. Ye J, Su J, Chen K, Liu H, Yang X, He Y, et al. Comparative investigation on chemical constituents of flower bud, stem and leaf of Lonicera japonica Thunb. by HPLC-DAD-ESI-MS/MSn and GC-MS. J Anal Chem. 2014;69:777-84
198. Yıldız Turgut D, Çınar O, Seçmen T. Determination of functional properties of kumquat (Fortunella margarita Swing.) powders obtained by different methods. GIDA J Food. 2019;44:605-17
199. Zengin G, Mahomoodally MF, Sinan KI, Ak G, Etienne OK, Sharmeen JB, et al. Chemical composition and biological properties of two Jatropha species: different parts and different extraction methods. Antioxidants (Basel). 2021;10(5):792
200. Zhang C, Yin Z, Ye W, Guan Y, Guo L, Zhang J, et al. [Chemical constituents from stems of Lonicera japonica]. Zhongguo Zhong Yao Za Zhi. 2009;34:3051-3
201. Zhang J, Yang J, Duan J, Liang Z, Zhang L, Huo Y, et al. Quantitative and qualitative analysis of flavonoids in leaves of Adinandra nitida by high performance liquid chromatography with UV and electrospray ionization tandem mass spectrometry detection. Anal Chim Acta. 2005;532:97-104
202. Zhang L, Zengin G, Mahomoodally MF, Yıldıztugay E, Jugreet S, Simal-Gandara J, et al. Untargeted phenolic profiling and functional insights of the aerial parts and bulbs of Drimia maritima (L.) Stearn. Plants (Basel). 2022;11(5):600
203. Zhang S-P, Gu F-L, He S-Z, Wang Q-H. Separation and identification of the active ingredients in Piper nigrum L. leaves and analysis of their antioxidant activity. Mod Food Sci Technol. 2015;31(2):63-9
204. Zhang Y. Flavonoids in grapefruit and commercial grapefruit juices: concentration, distribution and potential health benefits. Proc Fla State Hort Soc. 2007;120:288-94
205. Zheng B, Zheng Y, Zhang N, Zhang Y, Zheng B. Rhoifolin from Plumula nelumbinis exhibits anti-cancer effects in pancreatic cancer via AKT/JNK signaling pathways. Sci Rep. 2022;12:5654
206. Zheng X, Zheng Y, He Y, Zhang J, Peng W, Su W. Microbial metabolism of naringin and the impact on antioxidant capacity. Nutrients. 2022;14(18):3765. doi: 10.3390/nu14183765
207. Zhou C, Chen J, Zhang H, Zhang S, Zhang Y, Liu K, et al. Investigation of the chemical profile and anti-inflammatory mechanisms of flavonoids from Artemisia vestita Wall. ex Besser via targeted metabolomics, zebrafish model, and networl pharmacology. J Ethnopharmacol. 2023;302:115932
208. Zhou Y, Cao Y, Li JH, Agar OT, Barrow C, Dunshea F, et al. Screening and characterization of phenolic compounds by LC-ESI-QTOF-MS/MS and their antioxidant potentials in papaya fruit and their by-products activities. Food Biosci. 2023;52:102480
 
 

Figure 1: Graphical abstract: Mechanism of rhoifolin's anti-metastatic and pro-apoptotic effects on cancer cells. This diagram illustrates the anti-metastatic and pro-apoptotic effects of rhoifolin on cancer cells. The left side of the image shows the inhibition (↓) of key signaling molecules such as ezrin, PODXL, NHERF-1/2, Cdc42, MAPK, PI3K, Rac1, and RhoA, which are involved in cell motility and migration. This inhibition prevents the migration of cancer cells from the primary tumor site and their entry into the bloodstream, effectively reducing metastasis and invasion. The right side of the image depicts the chemical structure of rhoifolin and its role in promoting apoptosis (↑Apoptosis) by modulating various molecular pathways, including the upregulation (↑) of JNK and p-JNK, and the downregulation (↓) of pAkt, TGF-β2, and SMAD2, leading to increased cancer cell death.

Abbreviations: Cdc42, cell division control protein 42 homolog; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; NHERF, Na+/H+ exchanger regulatory factor; PI3K, phosphoinositide 3-kinase; PODXL, podocalyxin-like protein; Rac1, Ras-related C3 botulinum toxin substrate 1; RhoA, Ras homolog family member A; SMAD2, mothers against decapentaplegic homolog 2; TGF-β2, transforming growth factor-beta 2)

Figure 2: Chemical structure of Rhf-G

Figure 3: Synthetic route for 1a (acacetin) from 3a (naringin) via compounds 1b-1g. Chemical structures for these compounds were also given (adapted from Hanamura et al., 2016)

Figure 4: Chemical structures of apiin and vitexin

Figure 5: Chemical structures of the compounds mentioned in Ma et al. (2014a)

Figure 6: Chemical structure of melitidin, bergamjuicin, and naringin

Figure 7: Chemical structure of hinokinin

 

Table 1: Plant species that are reported to contain rhoifolin

Table 2: Chemical structure, nomenclature, UV absorbance and NMR spectral data of rhoifolin

Table 3: Summary of in vitro anticancer activities of rhoifolin

Table 4: ORAC activity of SPFEs and targeted knockout collections of one or more isolated flavonoids and the activity loss rate values gained according to the knockouts. Values with different letters in each column indicate significant differences (p < 0.05). The mg DW in the unit of ORAC refers to the weight of SPFEs (adapted from Deng et al., 2022).

Table 5: Antioxidant activity values of crude extracts and the isolated compounds from L. japonica leaves in DPPH assay (adapted from Wang et al., 2017)

Table 6: Other pharmacological properties of rhoifolin

[*] Corresponding Author:

Jesús Herrera-Bravo, Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, eMail: jesusherrerabr@santotomas.cl