ABSTRACT
The genus Rumex (Polygonaceae), comprising about 193 species, is widely distributed across temperate and subtropical regions of Europe, Asia, Africa, and North America, and has long been used in traditional medicine worldwide. These species are rich in diverse phytochemicals, including anthraquinones, flavonoids, tannins, naphthalenes, and stilbenes, which contribute to a broad range of biological activities. This review provides an updated synthesis of current knowledge on the taxonomy, phytochemistry, and pharmacological properties of Rumex species. Particular attention is given to R. dentatus , R. vesicarius and R. crispus , which exhibit antioxidant, anti‐inflammatory, antimicrobial, hepatoprotective, antidiabetic, and anticancer effects in both in vitro and in vivo studies. Clinical and toxicological aspects, including oxalate accumulation and anthraquinone‐associated adverse effects, are also discussed. Major research limitations include the lack of standardized extracts, insufficient clinical evidence, and poor bioavailability of key compounds. Enhancing bioavailability is vital because many bioactive compounds in Rumex are poorly absorbed and rapidly metabolized, which restricts their therapeutic potential. Environmental factors and phenological stages influencing phytochemical expression are highlighted as additional underexplored determinants of bioactivity. By integrating ethnopharmacological knowledge with experimental data, this review identifies future research priorities, including the optimization of formulation strategies, pharmacokinetic evaluation, and clinical validation. Collectively, these efforts may support the development of safe and effective Rumex‐based nutraceuticals and therapeutic products.
Keywords: anticancer, antimicrobial, antioxidant, bioavailability, ethnopharmacology, pharmacological activities, phytochemistry, Rumex species, therapeutic potential, traditional medicine
Rumex dentatus , R. vesicarius , and Emex spinosa are traditionally used for managing gastrointestinal, respiratory, hepatic, and inflammatory disorders. In addition to their ethnomedicinal value, some species are consumed as wild edibles. Pharmacological studies support their antimicrobial, antioxidant, hepatoprotective, cytotoxic, and antidiabetic activities. These effects are linked to bioactive phytochemicals, including anthraquinones, flavonoids, phenolic acids, and fatty acids, underscoring their potential as functional food sources and candidates for evidence‐based phytotherapeutics.

Abbreviations
- ALP
alkaline phosphatase
- ALT
alanine aminotransferase
- AST
aspartate aminotransferase
- Bcl‐2
B‐cell lymphoma 2
- COX
cyclooxygenase
- ERK
extracellular signal‐regulated kinase
- IC₅₀
half‐maximal inhibitory concentration
- IL
interleukin
- JNK
c‐Jun N‐terminal kinase
- MAPK
mitogen‐activated protein kinase
- mTOR
mechanistic target of rapamycin
- NFATc1
nuclear factor of activated T‐cells 1
- NF‐κB
nuclear factor kappa‐light‐chain‐enhancer of activated B cells
- NO
nitric oxide
- PI3K/Akt
phosphoinositide 3‐kinase/Protein kinase B (Akt)
- PPAR
peroxisome proliferator‐activated receptor
- ROS
reactive oxygen species
- TNF‐α
tumor necrosis factor‐alpha
1. Introduction
The family Polygonaceae, known as the Buckwheat family, comprises approximately 61 genera and over 1100 species worldwide, divided into two subfamilies: Polygonoideae, characterized by the presence of an ocrea, and Eriogonoideae, distinguished by its absence (Borsch et al. 2020; Mabberley 1997). In the Egyptian flora, the family Polygonaceae is represented by eight genera and 26 species (El Gazzar et al. 2023). Globally, members of the Polygonaceae family are widespread, with the greatest diversity found in temperate regions of the Northern Hemisphere, particularly in Asia and North America, but also represented in tropical and subtropical regions worldwide (Gillani et al. 2024). The family consists mainly of herbs, shrubs, and small trees, characterized by simple, alternate leaves and a sheathing stipule known as the ocrea, usually found at swollen stem nodes. Inflorescences bear flowers with 4–6 petaloid tepals, 5–9 stamens, and 2–3 carpels in a superior ovary, producing a 3‐angled achene fruit (Ann and Kron 2003). Rumex L., the second largest genus in the family Polygonaceae, comprises approximately 200 species distributed worldwide, with the greatest diversity in temperate regions of the Northern Hemisphere but also represented in tropical and subtropical areas (Jiang et al. 2007). In the Northern Hemisphere, Rumex typically flowers between April and May, with seeds ripening from May to June. However, flowering and fruiting periods vary in the Southern Hemisphere (e.g., South America, southern Africa, Australia). Plants belonging to this genus are generally characterized as perennial herbs. Plants belonging to this genus are generally characterized as perennial herbs (Newman et al. 2000). Rumex species have had a valued place in global folk medicine, and different species present various biologically active compounds that explain their use in medicine and the pharmaceutical industry (Hasan et al. 1995). To date, 268 phytoconstituents have been identified from 29 Rumex species (Li et al. 2022), including anthraquinones, flavonoids, tannins, stilbenes, naphthalenes, diterpene alkaloids, terpenes, and lignans, which exhibit a broad spectrum of biological activities such as anti‐inflammatory, antioxidant, antibacterial, antitumor, and antidiabetic effects (Chelly et al. 2020; Elsayed et al. 2020; Farooq et al. 2020; Froldi et al. 2020; Quradha et al. 2019). Rumex vesicarius L., known as “Bladder Dock,” is locally called “Hammad” in parts of Egypt and the Middle East, and is an annual herb (Rahman et al. 2004). It is widely distributed throughout desert and semi‐desert areas of North Africa, Australia, and Asia (Yahya 1990). Phytochemical researchers showed that R. vesicarius presents numerous bioactive compounds such as quinones, carotenoids, flavonoids, vitamins (especially vitamin C), lipids, carbohydrates, anthraquinones particularly in roots (emodin and chrysophanol), saponins, tannins, phenols, reducing sugars, triterpenoids, and organic acids. R. vesicarius contains several minerals, such as K, Na, Ca, Mg, Fe, Mn, and Cu (Khan et al. 2014). This plant is used in traditional folk medicine to treat asthma, bronchitis, dyspepsia, vomiting, piles, hepatic diseases, constipation, bad digestion, scabies, leucoderma, toothache, appetizer, spleen disorders, pains, heart troubles, flatulence, and as a laxative, stomachic, tonic, diuretic, and analgesic (Batanouny et al. 1999). Several researchers indicated the potential activity of R. vesicarius as antioxidant, membrane stabilizing, hepatoprotective, and antifibrotic effects against CCl4 intoxication in rats (El‐Hawary et al. 2011). Rumex dentatus L., also known as “Dentate Dock”, “Indian Dock”, and “Toothed Dock” (Fatima et al. 2009) is traditionally employed for its bactericidal (Yildirim et al. 2001), anti‐inflammatory (Humeera et al. 2013; Vasas et al. 2015), anti‐dermatitis, astringent, antitumor, diuretic, laxative, cholagogue, and tonic (Litvinenko and Muzychkina 2003). Chemical investigations have shown the presence of anthraquinones in its roots (Liu et al. 1997; Zhu et al. 2006), and various phenolic compounds, flavonoids, and tannins in its aerial parts (Hussain et al. 2010; Zhu et al. 2006). This review provides a comprehensive and updated overview of the botanical characteristics, traditional uses, phytochemical composition, pharmacological properties, clinical relevance, and toxicological data of Rumex species, with particular emphasis on mechanistic insights and research gaps that merit further investigation.
2. Methodology
To conduct this review, a comprehensive and systematic search was performed across several scientific databases, including PubMed/MEDLINE, ScienceDirect, Scopus, Web of Science, and Google Scholar. Search terms were derived from MeSH (Medical Subject Headings) and relevant literature, and included the following keywords: Rumex, Rumex classification, Rumex chemistry, Rumex species, flavonoids, anthraquinones, plant extracts pharmacology, medicinal plants, phytotherapy, phytochemicals, ethnopharmacology, bioavailability, toxicology, antioxidants, antimicrobial agents, anti‐inflammatory agents, antineoplastic agents, phytogenic, hepatoprotective agents, and hypoglycemic agents, plant‐derived. The selection process prioritized studies that provided relevant data on botanical features, phytochemistry, traditional uses, pharmacological properties, and toxicology of Rumex species. Inclusion criteria comprised English‐language publications that presented original research on the chemical composition, bioactivity, clinical relevance, or traditional medicinal application of Rumex species. Studies were excluded if they were not peer‐reviewed, lacked pharmacological focus, included homeopathic formulations or minerals, or were case reports, editorials, or reviews. Duplicate records were eliminated using citation management tools, and plant taxonomy was validated using the World Flora Online database. Chemical structures were verified through PubChem. Full‐text assessments were performed to confirm eligibility, and only studies meeting the predefined criteria were included in the qualitative synthesis (Figure 1).
FIGURE 1.

Summary of search and inclusion criteria used in this review.
3. Taxonomy and Botanical Features
Within the family Polygonaceae, which is divided into subfamilies Polygonoideae and Eriogonoideae, the genus Rumex comprises approximately 200 species that are broadly distributed across the globe, with its greatest diversity in temperate regions. As described by Sanchez and Kron (2008), Eriogonoideae is defined by the presence of involucres (clusters of bracts). The flowers are enclosed, except in the case of Gilmania. This subfamily is distinguished by non‐swollen nodes and the lack of ocrea, although perennial species of Chorizanthe in South America are an exception. Some authors reported four tribes in Polygonoideae named Persicarieae, Rumiceae, Polygoneae, and Muehlenbeckia. Emex Neck. ex Campderá, Oxyria, Hill, Rheum L., and Rumex L., is characterized by a fimbriate or pennicilate stigma (Sanchez et al. 2011) (Figure 2).
FIGURE 2.

Photographs of Rumex dentatus L. (A) leaf, (B) petiole, (C) inflorescences, (D) root, and (E) upper leaves.
3.1. Polygonaceae Juss
In Egyptian flora, there are 28 wild species in 8 genera from Polygonaceae including herbs, lianas, or shrubs, and rarely trees. In comparison, the Flora of North America recognizes approximately 35 genera and 442 species of Polygonaceae, while the Flora of China reports 13 genera (including two endemic) and 238 species (65 endemic) within China (Boulos 1999 & 2005). Stems are occasionally twining or climbing, typically jointed with nodes that are swollen. Leaves are simple and alternate, though they are rarely opposite or whorled. They can be entire or exhibit various lobed or toothed margins. Stipules are fused to form a membranous sheath known as ocrea. The inflorescence can take the form of heads, spikes, or panicles, consisting of cymose clusters, though it may also feature solitary flowers. Bracts, numbering 1–2, are fused into a sheath called ochreole. Pedicels are generally articulate. The flowers of Rumex are actinomorphic, hermaphroditic, or occasionally unisexual, with plants being monoecious or dioecious. The perianth consists of two whorls of sepaloid or colored tepals, numbering 3–6, which may be free or partially fused and are typically persistent. In some cases, the inner whorl becomes accrescent or modified in fruit. The stamens, usually 6–9, sometimes more or fewer, are opposite the tepals and contain 2‐celled anthers. The ovary is superior, 1‐celled and single‐ovuled, with 2–4 styles that are free or partially fused at the base. The fruit is a flat, angular, or winged achene, often enclosed by a membranous or leathery perianth. The endosperm is abundant and mealy, although the embryo is lateral or eccentric, either curved or straight (Boulos 1999 & 2005; Zohary and Feinbrun‐Dothan 1966).
3.2. Rumex dentatus L.
Common names of Rumex dentatus include Devil's thorn, Lesser Jack, Spiny Threecornerjack, and Toothed Dock. In Arabic, it is known by several names, including Dirs al‐Ajooz, Hommeid, Hommal, Khilla, Khella, and Khellala (Takhol 1974). This species is annual, glabrous, with 15–120 cm long or more (Figure 2C). Stems as shown in Figure 2C are erect, either simple or slightly branched. Branches are acute‐divaricated to erect. Basal leaves measure 8–12 cm long, 3 cm broad, ovate‐oblong or oblong‐lanceolate as presented in Figure 2A (Zohary and Feinbrun‐Dothan 1966). Basal leaves are occasionally fiddle‐shaped, with a more or less rounded apex and a base that ranges from truncate to subcordate, often appearing slightly wavy. The petiole is either equal in length to or shorter than the blade. As depicted in Figure 2B, the upper leaves are smaller, short‐petioled, and lanceolate to oblong or elliptical, somewhat acute, minutely crisped, or nearly smooth. The panicles are either branched or nearly simple, consisting of many‐whorled clusters. These whorls are densely flowered, spaced apart, and sometimes merge in the upper section, with nearly all subtended by linear‐lanceolate leaves. The pedicels, jointed near the base, are reflexed and nearly equal in length to the fruiting perianth. Valves measure 3–6 mm in length and 2–7 mm in width, including the teeth. They are 1.5–3 mm long warts, ovate‐lanceolate in shape, somewhat acute, and reticulate, often exhibiting sibilate teeth and occasionally subentire margins. The achene is approximately 2 mm long, trigonous, broadly ovoid, and dark brown (Boulos 1999 & 2005; Zohary and Feinbrun‐Dothan 1966). R. dentatus flowering commences in March and finishes in June. R. dentatus inhabits the banks of rivers and canals, moist waste ground, and roadsides. R. dentatus is widely distributed in the Mediterranean region, Euro‐Siberian, Sino‐Japanese, and also in some tropical regions. In Egypt, this species was recorded in Nile, Oasis, Sinai, and Mediterranean phyto‐geographical regions.
3.3. Rumex vesicarius L.
The common name of R. vesicarius is Ruby dock, and Bladder dock. This plant is known in Arabic names as Hambeit, Hanbeit, Hamaad, Hammaad, Hammaad el‐‘eshb, Hommaad (الحُمَّاض) and Hommeid (الحُمَّيْض) (Takhol 1974). R. vesicarius is an annual plant, green‐glaucescent, glabrous, 10–60 cm long or more as in Figure 3B. Stems as shown in Figure 3B are decumbent ascending, branching from the base rather thick. Leaves as shown in Figure 3C measured 7 cm long, and 1.5–5 cm broad, are fleshy, petiolate, ovate to deltoid or oblong‐triangular, cuneate or truncate, subcordate or sub hastate at the base, more or less obtuse, entire. Flowers as shown in Figure 3B are hermaphrodite or unisexual, racemose, or paniculate. Pedicles are single in axils, often bearing 2 flowers as in Figure 3B, jointed below the middle, elongated, and reflexed in fruit. Valves measured 1–2 cm long and are longer than pedicles. There are membranous, subequal, suborbicular, narrowly sinuate‐cordate at the base and apex, longitudinally folded and hence more or less ovate in outline, concealing the second flower, entire, purplish, and netted‐veined, without marginal vein, 2 of the 3 valves bearing a basal wart. Achene measured 3 mm long or longer, are trigonous, ovoid, acuminate, and brownish; those of the first flowers are somewhat longer than those of the second flowers (Boulos 1999 & 2005; Zohary and Feinbrun‐Dothan 1966). R. vesicarius flowering commences in February and finishes in May. R. vesicarius habitats are desert roads, rocky, and sandy desert wadis. R. vesicarius is widely distributed in North Africa, Southwest Asia (extending to South Greece), eastwards to North India and Afghanistan. In Egypt, the species was recorded in the Mediterranean, Desert, Sinai, Red Sea, and Gebel‐Elba phyto‐geographical regions.
FIGURE 3.

Photographs of Rumex vesicarius L. (A) root, (B) inflorescences, (C) leaf, and (D) upper leaf.
4. Phytochemistry
Several researchers have been attracted to study the chemical composition of many species belonging to the genus Rumex because of their medicinal properties. Numerous classes of phytocompounds appearing in different species belong to the genus Rumex (Shafiq et al. 2017). Methanolic extract of Rumex dentatus has shown the presence of saponins, alkaloids, tannins, and anthraquinones although flavonoids were found in both methanolic and hexane extracts (Fatima et al. 2009). Most of the species under this genus, such as Rumex japonicus Houtt., Rumex patientia L., Rumex crispus L., Rumex hymenosepalus Torr., and Rumex dentatus contain flavonoids, anthraquinones, and triterpenoids (Rao et al. 2011). Moreover, flavonoids were abundant in R. vesicarius although R. dentatus was characterized by the abundance of anthraquinones and chromones (Elfotoh et al. 2013). The phytochemical contents of Rumex species can vary significantly depending on the harvest season, geographical location, soil composition and other environmental conditions, which may influence their pharmacological activity and therapeutic consistency.
4.1. Fatty Acids
Several fatty acids have been identified and isolated from different species of the genus Rumex.
Therefore, one important group of fatty acids includes two types: essential fatty acids, such as linoleic acid, and non‐essential fatty acids, such as saturated acids like palmitic acid and stearic acid, as shown in Figure 4. Rumex induratus Boiss. & Reut. (syn. Rumex scutatus subsp. induratus (Boiss. & Reut.) Malag.) has been reported to contain fatty acids such as linoleic acid, palmitic acid, and stearic acid. In contrast, Rumex abyssinicus Jacq. has been found to contain high levels of oxalic acid, a naturally occurring organic acid known for its antinutritional properties Figure 4. Moreover, Rumex nervosus Vahl was reported to contain organic acids such as citric acid and tartaric acid. The crude extracts of these plants exhibited antibacterial activity, which has been attributed to the presence of oxalic acid, citric acid, and tartaric acid (Shafiq et al. 2017). In addition, volatile compounds derived from fatty acid metabolism, including 4‐hexen‐1‐ol and 2,4‐hexadiene‐1‐ol, were detected in R. induratus (Mabberley 1997). Furthermore, R. dentatus and R. vesicarius have been reported to contain high levels of fatty acids such as palmitic, linoleic, and oleic acids (Fatima et al. 2009).
FIGURE 4.

Structures of the reported acids and fatty acids: Short‐chain organic acids such as oxalic, citric, and tartaric acids contain multiple carboxyl groups and exhibit strong metal‐chelating and acidifying properties, contributing to antimicrobial effects. In contrast, long‐chain saturated fatty acids such as palmitic and stearic acids primarily support membrane structure and have limited direct bioactivity. Among unsaturated fatty acids, linoleic acid—characterized by two double bonds—has demonstrated antioxidant and anti‐inflammatory properties due to its unsaturation and role in lipid signaling.
4.2. Phenolic Acids
Several phenolic acids have been isolated from Rumex species. As shown in Figure 5, benzoic acid has been identified in R. induratus, occurring naturally in free or esterified forms such as methyl or ethyl esters (Cunnane 2003; Taveira et al. 2009). Ferulic acid was found in the leaves of R. induratus (Shafiq et al. 2017), and gallic acid was reported in the roots of R. dentatus (Zhu et al. 2006). The names and chemical structures of these compounds are presented in Figure 5.
FIGURE 5.

Structures of the reported phenolic acids. Hydroxyl groups on the aromatic ring enhance antioxidant activity, with gallic acid (three–OH) being the most potent. Ferulic acid's conjugated side chain improves radical stabilization. 4‐vinylguaiacol's vinyl group contributes to moderate antioxidant and antimicrobial effects. Benzoic acid, with no additional –OH groups, shows weaker antioxidant activity but retains antimicrobial activity.
4.3. Anthraquinones
Research has indicated that anthraquinones show several biological activities. This is the case with the roots of R. crispus , which contain three important anthraquinones: 1,5‐dihydroxy‐3‐methylanthraquinone (ziganein), 1,3,5‐trihydroxy‐6‐hydroxymethylanthraquinone, and 1,5‐dihydroxy‐3‐methoxy‐7‐methylanthraquinone (shiraiarin) (Figure 6). These compounds confirmed the use of the roots of R. crispus to purify the blood in skin complaints, and to cure constipation. In addition, these compounds have purgative, anti‐microorganic, and anti‐inflammatory activities (Shafiq et al. 2017). R. abissinicus and leaves of Rumex nepalensis Spreng. have been indicated to have chrysophanol, which, when given to dysmenorrhea patients, showed antifungal activity (Sharma et al. 2008). The roots of R. japonicus , which presented emodine, were reported for curing acute and chronic cutaneous disease (Başkan et al. 2007; Li et al. 2000). Moreover, many Rumex species, such as Rumex acetosa L., Rumex acetosella L., Rumex confertus Willd., R. crispus , R. hydrolapathum Huds., R. nepalensis , and R. obtusifolius L. have been investigated to identify anthraquinone derivatives such as physcion, physcioni, rhein, and physcion‐8‐O‐β‐D‐glucopyranoside (Figure 6). These compounds are significant secondary metabolites in plants and have been identified and isolated through chromatographic methods. All these compounds of these plants can be used in phytotherapy as they deliver a material called radix lapathi which has laxative properties (Cos et al. 2002). Nepodin, a key antifungal agent, was extracted from the roots of R. nepalensis; although emodin and physcion, isolated from the roots of R. abyssinicus , were noted for their antibacterial activity against a broad range of bacteria (Desta 1995). Furthermore, the leaves, roots, and fruits of R. crispus and R. obtusifolius have been reported to contain sennoside A, a compound widely used as a natural laxative. (Cos et al. 2002). Similarly, 3‐acetyl‐5‐hyroxy‐7‐methoxy‐2‐methyl‐1,4‐naphthaquinone isolated from R. japonicus was used as an antimicrobial agent. The anthraquinone derivative aloe‐emodine acetate serves as a precursor for the synthesis of anthracycline antibiotics and exhibits a variety of biological activities, including cathartic, antimicrobial, antiseptic, antibacterial, anti‐mutagenic, and anti‐leukemic effects. The presence of anthraquinone glycosides such as emodine‐6‐O‐β‐D‐glucopyranoside, chrysophanein, emodine‐8‐O‐β‐D‐glucopyranoside extracted from dried roots of R. patientia has been investigated. These last two are used as purgatives, constipation remedies, depuratives, and tonics in Turkish medicine (Demirezer, Kuruüzüm‐Uz, et al. 2001). Additionally, several anthraquinones have been identified in the extract of R. nepalesis. These compounds reinforce the traditional medicine system by enhancing intestinal peristalsis, promoting gastrointestinal motility, and exhibiting purgative effects. Aloe‐emodine‐8‐O‐β‐D‐glucopyranoside (Figure 6), emodin and nataloin were isolated from Rumex spinosus L.
FIGURE 6.

Structures of the reported anthraquinones identified in Rumex species, including emodin, chrysophanol, physcion, rhein, and aloe‐emodin derivatives.
4.4. Flavonoids
The leaves of various Rumex species are traditionally used in the treatment of diverse skin and systemic conditions, which can be attributed to their rich flavonoid content. Compounds such as hyperin, quercetin, quercitrin, iso‐orientin, avicularin, vitexin, and orientin (Figure 7) are present and have been linked to therapeutic applications against hives, ringworm, boils, jaundice, psoriasis, eczema, scabies, itching, acne, and other skin disorders (Shafiq et al. 2017). Studies on R. japonicus have identified the presence of quercetin, isoquercitrin, quercitrin, as well as astragalin and catechin (Figure 7) (Spedding et al. 1989). These phytochemicals have shown promising antidiabetic effects and are considered potential agents for the management of diabetes and associated metabolic disorders (Shafiq et al. 2017). Notably, both rutin and quercitrin have demonstrated anticancer properties, particularly against breast cancer. R. crispus exhibits strong antioxidant activity attributed to its high flavonoid content. Catechin has been isolated from R. patientia , along with halogenated derivatives such as 6‐chlorocatechin, which possess significant free radical‐scavenging capacity and cytotoxic activity (Middleton Jr. et al. 2000). Furthermore, several flavonoids have been detected in R. nervosus (Burt 2004; Kerem et al. 2006), which is traditionally used for the treatment of acne, wound healing, typhus, eczema, and rabies, and is also valued for its hypoglycemic and ophthalmic antiseptic properties (Lee et al. 2005). A flavonoid glycoside, querciturone, isolated from R. aquaticus (Kim et al. 2010), has been reported to provide protection against gastritis and esophagitis (Clark et al. 2000; Takahashi et al. 2004), although it also exhibits anti‐inflammatory (Hill‐Kapturczak et al. 2001; Zhou et al. 2004), anti‐proliferative (Deng et al. 2004), and anti‐apoptotic effects (Pae et al. 2004). The seeds and leaves of R. crispus also display antioxidant and antimicrobial activity, demonstrated by their ability to reduce 2,2‐diphenyl‐1‐picrylhydrazyl (DPPH) radicals and inhibit microbial growth (Warth 1991). In addition, quercetin has been extracted from the roots of R. dentatus (Zhu et al. 2006). In R. vesicarius , several bioactive compounds have been identified, including orientin (8‐C‐glucosyl‐luteolin), vitexin (8‐C‐glucosyl‐apigenin), naringin, diosmin (7‐O‐rutinosyl‐diosmetin), and catechin (El‐Hawary et al. 2011), all of which contribute to its pharmacological potential.
FIGURE 7.

Structure of the reported flavonoids. The flavonoid core with multiple hydroxyl groups enhances antioxidant and anti‐inflammatory activity. Glycosylation (e.g., rutin, hyperoside, quercitrin) improves solubility but may reduce cell permeability. C2–C3 double bonds and 4‐oxo groups (e.g., in quercetin) favor radical scavenging and enzyme inhibition. Substituent positions influence bioactivity potency.
The diversity of phytochemical constituents across various Rumex species is summarized in Table 1.
TABLE 1.
Phytochemical constituents of Rumex species and their biological activities.
| Phytochemical class | Compounds | Rumex species | Biological activities | References |
|---|---|---|---|---|
| Fatty acids |
Fatty acids
Organic acids
Volatile compounds (fatty acid derivatives)
|
R. induratus R. abyssinicus R. nervosus R. dentatus R. vesicarius |
Antibacterial Metabolic support Essential lipid source |
Shafiq et al. (2017) |
| Phenolic acids | Benzoic acid, ferulic acid, gallic acid |
R. induratus R. dentatus |
Antioxidant Anti‐aging Anticancer Antimicrobial |
Cunnane (2003), Taveira et al. (2009) and Zhu et al. (2006) |
| Anthraquinones | Emodin, chrysophanol, physcion, rhein, nepodin, aloe‐emodin, glycosides (e.g., emodin‐8‐O‐β‐D‐glucoside) |
R. crispus R. japonicus R. abyssinicus R. nepalensis R. acetosa R. obtusifolius R. patientia |
Laxative Antimicrobial Antifungal Anti‐inflammatory Anti‐mutagenic |
Cos et al. (2002), Sharma et al. (2008), Demirezer, Kuruüzüm‐Uz, et al. (2001) and Desta (1995) |
| Flavonoids | Quercetin, catechin, rutin, quercitrin, isoquercitrin, hyperoside, orientin, vitexin, naringin, diosmin |
R. japonicus R. dentatus R. crispus R. vesicarius R. aquaticus R. nervosus R. patientia |
Antioxidant Antidiabetic Anticancer Dermatological applications |
Shafiq et al. (2017), Spedding et al. (1989), Kim et al. (2010) and El‐Hawary et al. (2011) |
| Chromones and triterpenoids |
Chromones
|
R. dentatus R. japonicus R. patentia R. hymenosepalus |
Anti‐inflammatory hepatoprotective Antitumor |
Rao et al. (2011) |
| Other secondary metabolites | Saponins, alkaloids, tannins |
R. dentatus R. japonicus |
Astringent, tonic, antimicrobial, cytotoxic | Shafiq et al. (2017) and Fatima et al. (2009) |
5. Biological Activities: Underlying Mechanisms of Action
Several biologically active compounds identified in various Rumex species have demonstrated hepatoprotective, cytotoxic, antidiabetic, and other pharmacological activities. These findings have encouraged researchers to investigate the potential applications of this genus in medicine and the pharmaceutical industry. The biological activities of Rumex species are largely attributed to their diverse chemical constituents, which exhibit anti‐malarial, anti‐inflammatory, and additional therapeutic properties (Getie et al. 2003), anti‐diarrheal, and anti‐viral activities (Cos et al. 2002) and many other activities. Moreover, numerous studies have demonstrated that the Rumex genus possesses antioxidant, antiviral, antibacterial, antifungal, immunosuppressive, antitumor, and antimutagenic properties (Lee et al. 2005; Shafiq et al. 2017; Yildirim et al. 2001). The biologically active compounds identified in the Rumex genus, such as alkaloids, phenylpropanoids, sulfated compounds, isoprenoids, and peptides, are responsible for different biological activities. In pharmacological studies, extracts or isolated compounds from Rumex species have been tested at doses typically ranging from 25 to 500 mg/kg in animal models and from 1 to 1000 μg/mL in in vitro assays, depending on the extract type, species studied, and biological endpoint. In traditional medicine, decoctions or infusions are often administered orally at approximate doses equivalent to 1–5 g/day of dried plant material, though these practices vary by region and preparation method. However, standardization across studies is often lacking, complicating direct comparison (Prakash Mishra et al. 2018).
5.1. Antibacterial
The antibacterial activity of Rumex species has been investigated using different experimental approaches (such as agar well diffusion and broth dilution methods). Several research groups studied the antibacterial activity of Rumex species against Gram‐positive and Gram‐negative bacteria. Wegiera et al. (2011) tested the effects of Rumex fruit extracts: R. acetosa , R. acetosella , R. confertus , R. crispus , Rumex hydrolapathum Huds., and R. obtusifolius in inhibiting the growth of seven Gram‐positive ( Staphylococcus aureus ATCC 25923, S. aureus 209P—ATCC 6538P, S. epidermidis ATCC 12228, two S. aureus , and two S. epidermidis ) and Gram‐negative ( Escherichia coli O55, E. coli ATCC 3521, Proteus mirabilis , P. vulgaris , Klebsiella pneumoniae , K. oxytoca , and Pseudomonas aeruginosa ) bacteria. Indeed, the extract of four Rumex ( R. crispus , R. hydrolapathum, R. obtusifolius , and R. confertus ) showed a minimum inhibitory concentration (MIC) ranging from 62.5 to 125 μg/mL against Staphylococci, although for Escherichia coli ATCC 3521, Pseudomonas aeruginosa , and Proteus mirabilis , the MIC ranged between 125 and > 500 μg/mL. In addition, R. dentatus root extracts demonstrated antibacterial and antibiofilm activities against clinical isolates of P. aeruginosa , with MIC values ranging from 200 to 1000 μg/mL (Khan et al. 2024). The bioactive components gallic acid and emodin were identified, and in silico studies confirmed their interaction with the LasR protein—a key quorum‐sensing transcriptional regulator in Pseudomonas aeruginosa that controls the expression of numerous virulence factors. Inhibiting LasR disrupts bacterial communication and biofilm formation, thereby supporting the potential use of R. dentatus in treating infections caused by this pathogen (Khan et al. 2024). Similarly, R. vesicarius methanolic extracts exhibited antibacterial and antibiofilm properties, notably inhibiting Bacillus subtilis and P. aeruginosa with MIC values of 250–500 μg/mL (Sulieman et al. 2024). Phytochemical analysis revealed flavonoids, phenolics, tannins, and terpenes as major constituents responsible for these effects (Sulieman et al. 2024). In another study, Hussain et al. (2010) investigated the potential antibacterial effect of methanolic extraction of R. nepalensis roots against six bacterial species. The results showed significant activity against Citrobacter frundii, E. coli , Enterobacter aerogenes , and S. aureus with MICs of 16, 5.0, 25, and 0.156 μg/mL, respectively. Furthermore, the root, leaves, and stem of R. nepalensis have been studied for their antibacterial activity against human pathogenic strains. The results were promoted for leaf extracts. Aqueous leaf extracts of R. nepalensis revealed an inhibition zone of 24 mm against E. coli (Mungole and Chaturvedi 2011). However, the water extract of stems of R. nepalensis exhibited an inhibition zone of 20 mm against S. aureus although the water extract of the R. nepalensis roots showed an inhibition zone of 19.1 mm against Pseudomonas sp. In another study, Yadav et al. (2011) evaluated the effect of methanol, hexane, ethyl acetate, and chloroform of R. nepalensis against S. aureus , Salmonella enterica serovar Typhimurium, E. coli , and Klebsiella pneumoniae . The antibacterial effect of the ethyl acetate fraction revealed a MIC value of 6.3 and 3.1 mg/mL against S. aureus and E. coli , respectively. Moreover, bioassay‐guided fractionation of R. abyssinicus led to the isolation of six antimicrobial anthraquinones, including chrysophanol, emodin, and physcion, with MICs between 8 and 256 μg/mL (Kengne et al. 2021). Naphthalenes such as torachrysone and 2‐methoxy‐stypandrone, isolated from R. japonicus , demonstrated bacteriostatic effects against various Gram‐positive and Gram‐negative bacteria (Nishina et al. 1993). Further research on Rumex species from the Carpathian Basin revealed strong antibacterial activity, especially in the n‐hexane and chloroform fractions of roots from R. acetosa , R. alpinus , R. aquaticus , R. conglomeratus , and R. patientia , with inhibition zones > 15 mm. Among the compounds tested, naphthalenes exhibited greater antimicrobial capacity (e.g., MIC = 48–57.8 μM for Moraxella catarrhalis ) than anthraquinones, flavonoids, stilbenes, and glycerides (Orbán‐Gyapai et al. 2017). Notably, R. crispus and R. sanguineus showed activity against Acinetobacter baumannii , suggesting potential for wound healing applications (MIC = 1.0–4.0 mg/mL) (Aleksic Sabo et al. 2020). In addition, anti‐ Helicobacter pylori activity was observed in extracts of R. acetosa , where six anthraquinones exhibited MIC values between 3.13 and 25 μM (Kang et al. 2023). These compounds, including emodin and chrysophanol glucosides, also showed urease inhibitory effects, suggesting their potential for managing H. pylori ‐related gastric infections (Kang et al. 2023). Furthermore, green synthesis of silver nanoparticles (AgNPs) using Rumex sp. leaf extracts showed significant antimicrobial and antibiofilm activity. The AgNPs reduced biofilm formation in A. baumannii , K. pneumoniae , and E. coli , offering a potential nanotechnology‐based solution against resistant pathogens (Akay et al. 2024). Although antibacterial compounds such as anthraquinones and naphthalenes are effective, high concentrations or prolonged use may disrupt normal microbiota or cause mild gastrointestinal irritation.
5.2. Antifungal
Several works showed the antifungal effect of different extracts from different plant parts of Rumex species. Sharma et al. (2008) studied the effect of root extract of R. nepalensis for their antifungal activity against 12 different fungal pathogens. The results demonstrated that the root ethanolic extract of R. nepalensis showed a significant active result against the fungal pathogens such as Fusarium moniliforme, Fusarium solani, Avicularia versicolor , Pythium sp., Fusarium semitectum, Sporotrichum sp., Rhizopus sp., and Thermomyces sp. According to Wegiera et al. (2011), six species of Rumex fruit extracts ( R. acetosella , R. acetosa , R. crispus , R. confertus , R. obtusifolius , and R. hydrolapathum) showed antifungal activity using an agar dilution method. The extracts from fruits of R. crispus , R. confertus , R. obtusifolius , and R. hydrolapathum inhibited the growth of Candida spp. and Trichophyton mentagrophytes with MIC values from 250 to 500 μg/mL. Also, the anti‐fungal activity of the methanolic extraction of R. nepalensis leaf showed significant activity against Candida albicans (Yadav et al. 2011), although R. nepalensis root extraction showed higher activity against A. niger and moderate activity against A. solani , and A. flavus (Jain and Parkhe 2018). Extracts from R. japonicus leaves exhibited strong in vitro activity against T. mentagrophytes, T. rubrum , and C. albicans (MIC: 1.96–62.5 μg/mL), primarily attributed to anthraquinones such as rhein, emodin, and aloe‐emodin, as well as polyphenols and flavonoids like rutin and quercetin (Xiao et al. 2024). These compounds disrupt fungal spore germination, impair germ tube formation, increase cell membrane permeability, damage biofilms, and downregulate adhesion‐related gene expression. Additional studies confirmed the potent antifungal effects of rhein, emodin, and aloe‐emodin from R. japonicus against dermatophytes, including Microsporum canis, with MICs ranging from 1.9 to 62.5 μg/mL (Sun et al. 2024). A quality evaluation study of R. japonicus highlighted the antifungal activity of emodin‐8‐O‐β‐D‐glucopyranoside and ethyl acetate fractions against C. albicans (Xiao et al. 2022). Furthermore, ethanolic and aqueous extracts of R. nervosus also exhibited antifungal activity (Al‐Garadi et al. 2022). In vitro assays demonstrated complete inhibition of Aspergillus fumigatus and significant suppression of A. niger mycelial growth. Methanolic leaf and root extracts of R. nervosus also showed notable activity against C. albicans , as well as several oral bacterial pathogens, supporting its traditional medicinal use (Al‐Farhan et al. 2022). Some antifungal constituents, especially emodin and related anthraquinones, may cause mucosal irritation or hepatotoxic effects at elevated doses or with systemic use.
5.3. Antiviral
Several species of the Rumex genus have shown notable antiviral properties, particularly against members of the Herpesviridae family. Specifically, 1,4‐naphthoquinones and naphthalene derivatives isolated from R. aquaticus exhibited inhibitory effects on the replication of herpes simplex virus type 2 (HSV‐2) in Vero cell cultures. Among these, musizin demonstrated dose‐dependent antiviral activity, resulting in a 2.00 log₁₀ reduction in viral load at a concentration of 6.25 μM, as determined by conventional virus yield reduction assays and quantitative PCR (Rédei et al. 2019). Similarly, an acetone–water extract prepared from the aerial parts of R. acetosa , rich in oligomeric and polymeric proanthocyanidins as well as flavonoids, demonstrated strong antiviral effects against herpes simplex virus type 1 (HSV‐1). Using plaque reduction and MTT assays on Vero cells, the extract showed complete inhibition of HSV‐1 at concentrations above 1 μg/mL (IC₅₀ = 0.8 ± 0.04 μg/mL), with minimal cytotoxicity (CC₅₀ = 78.6 ± 12.7 μg/mL) (Gescher et al. 2011). Although antiviral polyphenols are generally well tolerated, excessive intake or long‐term use of concentrated extracts may lead to gastrointestinal upset or interference with micronutrient absorption.
5.4. Antioxidant
Gomaa and Saleh (2015) reported significant antioxidant and antiproliferative effects of aqueous extracts from R. vesicarius leaves, flowers, and their combination, with the flower extract showing the highest activity in a dose‐dependent manner. Consistently, wild‐grown R. vesicarius from the mountains of Hail demonstrated significant antioxidant activity at lower concentrations compared to ascorbic acid, butylated hydroxytoluene (BHT), and β‐carotene, confirming its richness in bioactive phytochemicals and essential minerals (Sulieman et al. 2024). A comprehensive metabolic profiling of R. vesicarius identified 60 metabolites, including phenolic acids, flavonoids, terpenes, and organic acids. Molecular docking studies revealed strong binding affinities of predominant metabolites to NADPH oxidase and human peroxiredoxin 5, confirming the stems and roots of R. vesicarius as rich sources of antioxidant compounds (Sweilam et al. 2024). Elzaawely and Tawata (2012) evaluated ethyl acetate fractions of leaves and roots of R. dentatus , which exhibited strong DPPH radical scavenging activity, with IC50 values of 21 and 12 μg/mL, respectively. Similarly, in R. patientia , halogenated flavan‐3‐ol‐6‐chlorocatechin and catechin showed potent DPPH scavenging, whereas other compounds such as physcion and emodin derivatives lacked activity (Demirezer, Kuruüzüm, et al. 2001). Methanolic extracts of Rumex maritimus L. also demonstrated notable antioxidant potential, with an IC50 value of approximately 80 μg/mL compared to 7 μg/mL for ascorbic acid in DPPH assays (Hossain et al. 2015). Antioxidant properties of R. acetosella were confirmed through DPPH, ABTS+, radical scavenging, and phosphomolybdate assays (Özenver et al. 2020). Regarding R. crispus , antioxidant activity was demonstrated across various assays (DPPH, ABTS+, NO, phosphomolybdate, and SPF), supporting its development as an antioxidant, anti‐aging, and dermatological agent (Uzun and Demirezer 2019). Dichloromethane and ethyl acetate fractions of R. crispus roots showed enhanced activity correlated with polyphenol and flavonoid content (Eom et al. 2020). Chrysophanol and physcion extracted from R. crispus roots displayed significant xanthine oxidase inhibitory activity, with IC₅₀ values of 36.4 and 45.0 μg/mL, respectively (Minh et al. 2019). In R. balcanicus Rech.f., fruits exhibited remarkable antioxidant potential, with IC50 values of 4.9 μg/mL (DPPH), 0.8 μg/mL (ABTS), and a FRAP value of 5.9 mmol Fe2+/g. These effects were attributed to high contents of miquelianin and procyanidin B1 (Krgović et al. 2025). Furthermore, isolated compounds from R. hastatus, including chrysophanol, 1,3,7‐trihydroxy‐6‐methylanthraquinone, przewalskinone B, and p‐coumaric acid, demonstrated nitric oxide (NO) radical scavenging activity with IC50 values between 0.39 and 0.47 mM (Shafiq et al. 2020). Finally, silver nanoparticles synthesized using Rumex sp. leaf extracts exhibited antioxidant activity, benefiting from plant‐derived phytochemicals involved in nanoparticle synthesis (Gam et al. 2024). It is important to note that high doses of antioxidant‐rich extracts, particularly those containing anthraquinones and tannins, can cause gastrointestinal discomfort such as cramping or diarrhea.
5.5. Anti‐Inflammatory
Several Rumex species have demonstrated anti‐inflammatory properties in both in vitro and in vivo models. In cellular models, methanolic extracts from the roots and stems of Rumex roseus L. suppressed the expression of interleukin (IL)‐6 and IL‐8 induced by tumor necrosis factor (TNF)‐α in intestinal epithelial cells, suggesting a regulatory effect on inflammatory pathways (Chelly et al. 2021). For R. crispus , an ethyl acetate extract of its roots exhibited marked anti‐inflammatory effects in vitro, including NO inhibition and downregulation of pro‐inflammatory cytokines (Eom et al. 2020). Additionally, R. crispus extracts enhanced the suppression of lipopolysaccharide (LPS)‐induced inflammatory responses in RAW 264.7 murine macrophages compared to unprocessed extracts, reducing nitrite levels and inflammatory cytokine expression, likely through modulation of immune signaling pathways (Pan et al. 2020). Furthermore, a formulation composed of processed R. crispus and Cordyceps militaris, traditionally used in Korean folk medicine, was evaluated for its effects on ovalbumin‐induced allergic rhinitis (AR) in rats. The formulation significantly reduced serum and nasal mucosa levels of immunoglobulin E (IgE), histamine, thymic stromal lymphopoietin, TNF‐α, IL‐1, IL‐4, IL‐5, and IL‐13 (H. Y. Kim et al. 2019). The ethanol extract of R. japonicus roots showed therapeutic promise for atopic dermatitis, significantly alleviating skin inflammation in a Balb/c mouse model (Yang et al. 2019). In vitro studies with HaCaT cells demonstrated that the extract reduced mitogen‐activated protein kinase (MAPK) phosphorylation and modulated nuclear factor (NF)‐κB activation in response to TNF‐α stimulation. Additionally, R. japonicus methanolic extract protected intestinal epithelial integrity in a dextran sulfate sodium (DSS)‐induced colitis model in C57BL/6N mice (H. R. Yang et al. 2019). For R. nepalensis , chloroform and ethyl acetate root extracts significantly reduced ear edema in a 12‐O‐tetradecanoylphorbol‐13‐acetate (TPA)‐induced acute inflammation mouse model (Gautam et al. 2010). Mechanistically, these extracts exhibited strong cyclooxygenase (COX)‐1 and COX‐2 inhibition, attributed to isolated anthraquinones (emodin, endocrocin) and naphthalene derivatives (nepodin). Emodin displayed moderate COX‐2 selectivity, although nepodin showed COX‐1 selectivity and potent antioxidant capacity, suggesting a dual mechanism of COX inhibition and radical scavenging in the anti‐inflammatory effect (Gautam et al. 2010). An aqueous extract of R. patientia roots exhibited anti‐inflammatory effects in vivo, with the higher tested dose (150 mg/kg) reducing carrageenan‐induced paw edema in rats by 41.7%, a greater inhibition than that observed with the reference drug indomethacin (10 mg/kg, 36.6%) (Süleyman et al. 1999). Rumex abyssinicus rhizome extracts demonstrated both wound healing and anti‐inflammatory effects (Mulisa et al. 2015). In vitro, 80% methanol extracts exhibited dose‐dependent anti‐inflammatory activity in the carrageenan‐induced paw edema model. Topical application of extract‐based ointments (5% and 10%) significantly improved wound contraction, epithelialization time, tensile strength, and hydroxyproline content, supporting its traditional use in wound healing (Mulisa et al. 2015). At pharmacological doses, some anti‐inflammatory constituents (e.g., anthraquinones, flavonoids) may induce gastrointestinal irritation or, in rare cases, electrolyte imbalance due to their mild laxative effects.
5.6. Hepatoprotective
The antioxidant capacity of Rumex species has been associated with their hepatoprotective properties (Table 1). Extracts of Rumex tingitanus L. have shown strong radical scavenging activity in vitro, with both the ethyl acetate fraction and the hydroalcoholic extract exhibiting notable antioxidant effects. Additionally, the hydroalcoholic extract demonstrated hepatoprotective activity in a CCl4‐induced liver toxicity model, evidenced by reduced lipid peroxidation (MDA levels) and enhanced antioxidant enzyme activities. These effects were further supported by the isolation of 4′‐p‐acetylcoumaroyl luteolin, a compound with superior DPPH radical scavenging activity compared to BHT (IC₅₀: 11.7 ± 0.2 μg/mL) (Mhalla et al. 2018). In vitro antioxidant analyses of R. abyssinicus rhizome extracts confirmed dose‐dependent radical‐scavenging activity, supporting its traditional use for liver protection (Adamu et al. 2020). Methanolic extracts of R. vesicarius (100 and 200 mg/kg body weight) exhibited hepatoprotective activity in CCl4‐treated rats (Tukappa et al. 2015). In addition, the methanolic extract of R. vesicarius was evaluated for cytotoxicity against HepG2 cell lines, revealing an IC50 value of 563.33 ± 0.8 μg/mL, indicating low cytotoxicity and suggesting its safety for further therapeutic applications. The administration of R. dentatus at doses of 250 and 500 mg/kg significantly mitigated paracetamol‐induced hepatic damage in mice, as evidenced by reductions in serum ALT, AST, ALP, and bilirubin levels, comparable to the standard hepatoprotective agent silymarin. Histopathological analysis corroborated these findings, although phytochemical screening confirmed the presence of active constituents such as quercetin, kaempferol, and myricetin (Saleem et al. 2014). Ethanol and butanol extracts from roots, leaves, and fruits of R. vesicarius significantly reduced hepatic enzyme markers (ALT, AST, ALP) and improved antioxidant status after 4 weeks of treatment at 100 mg/kg, with efficacy comparable to silymarin (50 mg/kg) (El‐Hawary et al. 2011). These effects were attributed to antioxidant mechanisms, membrane stabilization, and antifibrogenic activities (El‐Hawary et al. 2011). R. abyssinicus also demonstrated significant hepatoprotection in pre‐ and post‐treatment CCl4‐induced liver injury models. Oral administration of its extracts at 125, 250, and 500 mg/kg effectively attenuated the elevation of liver enzymes, with histological analysis revealing preserved hepatic structure (Adamu et al. 2020). Despite hepatoprotective potential, chronic or high‐dose use of Rumex extracts may paradoxically induce liver enzyme elevation or hepatotoxicity, especially in formulations lacking standardization (Figure 7).
FIGURE 8.

Anticancer mechanisms of Rumex L. Bcl‐2, B‐cell lymphoma 2; JNK, c‐Jun N‐terminal kinase; mTOR, mechanistic target of rapamycin; PI3K/Akt, phosphoinositide 3‐kinase/protein kinase B (Akt). Symbols: ↑ increase; ↓ decrease.
5.7. Anticancer
MTT assays conducted on HeLa (human cervical carcinoma), A431 (skin epidermoid carcinoma), and MCF7 (human breast adenocarcinoma) cell lines revealed that R. acetosa and Rumex thyrsiflorus Fingerh. possess the ability to suppress tumor cell proliferation (Lajter et al. 2013). In addition, phenolic extracts from the flowers of R. acetosa showed a concentration‐dependent antiproliferative effect on HaCaT cells in vitro. Increasing the extract concentration from 25 to 100 μg/mL progressively reduced cell proliferation (Kucekova et al. 2011).
The methanolic extract of R. vesicarius was evaluated for its hepatoprotective properties in vitro, particularly in a model of CCl4‐induced liver damage, which was evident at doses of 100 and 200 mg/kg body weight. This plant also demonstrated cytotoxic effects against HepG2 cells, a human liver cancer cell line (Tukappa et al. 2015). Furthermore, dichloromethane extracts obtained from R. crispus roots inhibited cell proliferation and triggered apoptosis in HepG2 cells (Eom et al. 2020). 2‐Methoxystypandrone, isolated from R. japonicus , displayed antiproliferative activity against Jurkat cells (Qiu et al. 2020). This effect was associated with a reduction in mitochondrial membrane potential and an increase in mitochondrial reactive oxygen species (Qiu et al. 2020). Different parts of the R. vesicarius plant—including stems, roots, flowers, and leaves—were tested for cytotoxicity using the MTS assay on several human cancer cell lines, including MCF7, Lovo, Caco‐2 (colon carcinoma), and HepG2 (Farooq et al. 2020). Among these, the stems showed the strongest cytotoxic effect in vitro and did not interfere with zebrafish embryonic development. The IC₅₀ values ranged from 33.45 to 62.56 μg/mL. At a dose of 30 μg/mL, the chloroform extract from the stems significantly suppressed angiogenesis in zebrafish embryos, inhibiting ≥ 70% of intersegmental blood vessels and completely blocking the formation of subintestinal vein vessels, indicating a promising antitumor effect (Farooq et al. 2020).
In triple‐negative breast cancer (TNBC) cell lines (MDA‐MB‐231), R. vesicarius extract enhanced the efficacy of sorafenib by downregulating anti‐apoptotic and pro‐survival genes BCl2, mTOR, and JNK, although upregulating the tumor suppressor gene p21 (Ghanem et al. 2023). Molecular docking studies confirmed the high affinity of R. vesicarius bioactive compounds for key targets involved in TNBC progression (Ghanem et al. 2023). In R. dentatus , a 70% methanolic leaf extract demonstrated weak in vitro inhibition of Ehrlich ascites carcinoma cells, although higher concentrations showed variable anticancer activity, likely due to differential chemical compositions depending on environmental growth conditions (Hawas et al. 2011). Notably, synergy studies combining R. dentatus aerial part phenolic aglycones with cisplatin in tongue squamous cell carcinoma (HNO97) cells revealed enhanced inhibition of proliferation, increased apoptosis, cell cycle arrest, and downregulation of autophagy pathways. These effects involved multiple signaling pathways including PI3K‐Akt, microRNAs in cancer, and resistance mechanisms to epidermal growth factor receptor inhibitors (Ragab et al. 2025). Further isolation and characterization of R. dentatus extracts identified fractions with potent cytotoxic activity, notably one fraction with an IC₅₀ of 11.29 μg/mL against colon cancer (HCT‐116) cells, which inhibited cell migration and invasion (Khaliq et al. 2023). Studies with R. obtusifolius have also shown promising results in combination therapies. The extract combined with 5‐fluorouracil (5‐FU) triggered apoptosis in A549 lung adenocarcinoma cells by modulating the PI3K/Akt pathway and reducing pro‐inflammatory markers such as TNF‐α and COX‐2, enhancing caspase‐3 activity (Ginovyan et al. 2024). Additional investigations combining R. obtusifolius extracts with arginase and nitric oxide synthase inhibitors demonstrated immunostimulatory, antiproliferative, antioxidant, anti‐inflammatory, and antiangiogenic effects in breast and colon cancer models (Ginovyan et al. 2023). Rumex japonicus root extracts showed protective effects against colitis‐associated colorectal cancer in mice by reducing inflammation, maintaining intestinal barrier integrity, and modulating cytokine profiles, highlighting its anti‐inflammatory and anticancer potential in gastrointestinal cancers (H. Y. Kim et al. 2022). Figure 7 summarizes the main anticancer mechanisms attributed to different Rumex L. species. These mechanisms include antiproliferative and pro‐apoptotic effects on various human cancer cell lines (e.g., HeLa, HepG2, MCF7, A549, HCT‐116), inhibition of angiogenesis, and enhancement of chemotherapeutic efficacy (e.g., sorafenib, cisplatin, 5‐FU). Key pathways modulated by Rumex extracts or isolated compounds involve PI3K/Akt, mTOR, Bcl‐2, JNK, p21, and microRNAs associated with tumor progression and drug resistance. Overall, the genus Rumex exerts its anticancer effects through multi‐targeted actions including apoptosis induction, suppression of angiogenesis, modulation of immune response, and inhibition of tumor cell migration and invasion. Cytotoxic effects of Rumex‐derived compounds may extend to non‐target cells at high concentrations, and long‐term use may carry genotoxic risks without appropriate dose control and safety profiling.
5.8. Anti‐Diabetic Activity
Chrysophanol and physcion, isolated from the roots of R. crispus , demonstrated inhibitory effects on α‐glucosidase with IC₅₀ values of 20.1 and 18.9 μM, respectively (Minh et al. 2019). The alcoholic extract of R. acetosella exhibited a stronger α‐glucosidase inhibitory activity compared to the positive control acarbose (IC₅₀ = 605 μM), with root and aerial parts showing IC₅₀ values of 12.3 μM and less than 10 μM, respectively (Özenver et al. 2020). Furthermore, the methanolic extract from Rumex lunaria L. leaves showed significant α‐glucosidase inhibition starting at a concentration of 3 μg/mL (Froldi et al. 2020). Similarly, the methanolic extracts of R. crispus flowers and leaves showed potent in vitro α‐glucosidase inhibition with IC₅₀ values of 7.3 and 112.0 μg/mL, respectively, and in vivo antihyperglycemic effects in streptozotocin (STZ)‐induced diabetic rats (Aguila‐Muñoz et al. 2023). The hypoglycemic activity of orally administered ethanol extract from R. obtusifolius seeds was assessed in hyperglycemic rabbits (Aghajanyan et al. 2018). Treatment resulted in a 57.3% reduction in fasting glucose levels, enhanced glucose tolerance, and a 1.5‐fold increase in liver glycogen content compared to controls. Additionally, the extract lowered total cholesterol, low‐density lipoprotein cholesterol, and liver enzyme levels while increasing high‐density lipoprotein cholesterol (Aghajanyan et al. 2018). Phenolic compounds extracted from R. dentatus were found to improve hyperglycemia by regulating carbohydrate metabolism in the liver, reducing oxidative stress, and upregulating PPARγ expression in diabetic rats (Elsayed et al. 2020). In vivo studies further confirmed that polyphenol‐rich extracts from R. dentatus administered orally at doses of 50–200 mg/kg reduce hyperglycemia and insulin resistance while enhancing carbohydrate metabolism in diabetic rats (Y. Yang et al. 2013). The ethanolic extract of R. vesicarius leaves exhibited dose‐dependent α‐glucosidase inhibition (up to 60% inhibition at 9 mg/mL) and significantly decreased blood glucose levels in STZ‐induced diabetic rats at doses of 100, 200, and 400 mg/kg (intraperitoneal administration) (Reddy et al. 2017). Histopathological examination confirmed protection of pancreatic β‐cells from STZ‐induced damage. Additionally, thermal treatment of R. vesicarius extracts maintained phenolic content and enhanced α‐amylase and α‐glucosidase inhibitory activities (Bouafia et al. 2025). Hydromethanolic root extracts of R. abyssinicus showed significant antidiabetic activity in normoglycemic, glucose‐loaded, and STZ‐induced diabetic mouse models, with doses of 100–400 mg/kg reducing blood glucose levels and supporting its traditional use in diabetes treatment (Yrga Adugna et al. 2022). These studies illustrate the principal antidiabetic mechanisms exerted by various Rumex species and their bioactive constituents. These include strong in vitro inhibition of α‐glucosidase and α‐amylase enzymes by extracts or isolated anthraquinones (e.g., chrysophanol and physcion), resulting in delayed carbohydrate digestion and postprandial glucose control. In vivo studies demonstrated significant antihyperglycemic effects, improved glucose tolerance, and enhanced hepatic glycogen storage in diabetic animal models. Some species, such as R. dentatus and R. nervosus, were also shown to reduce oxidative stress and inflammation, thereby contributing to improved insulin sensitivity, protection of pancreatic β‐cells, and prevention of diabetic nephropathy. Figure 8 illustrates the main pharmacological activities of Rumex species along with their associated bioactive compounds and molecular targets. Some Rumex extracts may potentiate the effects of antidiabetic medications, increasing the risk of hypoglycemia. Gastrointestinal effects such as nausea and diarrhea have also been reported at higher doses.
FIGURE 9.

Overview of major pharmacological activities of Rumex species and their associated bioactive compounds. Antibacterial activity includes antibiofilm effects mediated by compounds such as gallic acid, emodin, flavonoids, phenolics, tannins, and terpenes. Antifungal effects are primarily attributed to anthraquinones and involve inhibition of fungal germination, increased membrane permeability, and biofilm disruption. Antiviral activity is linked to proanthocyanidins and involves suppression of viral replication. Hepatoprotective effects are mediated by flavonoids and polyphenols, resulting in reduced hepatotoxicity and improved antioxidant enzyme activity. The anti‐inflammatory activity involves emodin and other constituents that downregulate interleukins, TNF‐α, and COX enzymes. Antioxidant effects, driven by flavonoids and polyphenols, include free radical scavenging and reduced oxidative stress. Antidiabetic activity is supported by chrysophanol, physcion, and phenolics through inhibition of α‐amylase and α‐glucosidase, improved glucose uptake, and modulation of carbohydrate metabolism and PPAR expression. ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; COX, cyclooxygenase; PPAR, peroxisome proliferator‐activated receptor; ROS, reactive oxygen species; TNF‐α, tumor necrosis factor‐alpha.
5.9. Other Pharmacological Properties
Intravenous administration of the methanolic extract from R. acetosa leaves at 50 mg/kg led to a reduction in mean arterial pressure by 27.9% ± 4.6% in normotensive rats, and by 48.4% ± 4.9% in hypertensive rats (Qamar et al. 2018). The antiplatelet properties and cardiovascular protective mechanisms of R. acetosa were also investigated. The extract of R. acetosa inhibited collagen‐induced platelet aggregation by regulating the phosphorylation of signaling pathways such as MAPK, PI3K/Akt, and Src family kinases, and reduced ATP release in a dose‐dependent manner ranging from 25 to 200 μg/mL (Jeong et al. 2020). Chrysophanol, emodin, and physcion from the aqueous extract of R. crispus suppressed osteoclast differentiation induced by RANKL by inhibiting the MAPK/NF‐κB/NFATc1 signaling pathway and increasing inhibitors of NFATc1, thus potentially stimulating osteoblast differentiation via the ERK/Runx2 pathway (Shim et al. 2017). Methanol extracts of R. nervosus demonstrated protective effects against STZ‐induced diabetic nephropathy in rats by activating the Nrf2 signaling pathway, reducing oxidative stress, inflammation markers, and apoptosis in renal tissues, thereby preserving kidney function (AlMousa et al. 2022).
Table 2 summarizes the biological effects of various Rumex species, detailing the extracts tested, experimental models used, key mechanisms of action, and supporting references.
TABLE 2.
Biological effects of Rumex species.
| Species | Extract/compound | Dose/route of administration/concentration | Experimental model | Potential mechanisms | References |
|---|---|---|---|---|---|
| Rumex crispus , R. hydrolapathum, R. obtusifolius , R. confertus | Methanolic extracts/fruits | MIC: 62.5–125 μg/mL (Staphylococci); 125–500 μg/mL (Gram‐negatives) | In vitro antibacterial assays | Antibacterial activity; flavonoids, phenolics, tannins, terpenes identified | Wegiera et al. (2011) |
| Rumex dentatus | Root extract | MIC: 200–1000 μg/mL | In vitro antibacterial and antibiofilm assays | Antibacterial, antibiofilm activity; gallic acid, emodin bioactives; LasR protein interaction | Khan et al. (2024) |
| Rumex vesicarius | Methanolic extract | MIC: 250–500 μg/mL |
In vitro antibacterial and antibiofilm assays |
Antibacterial activity; flavonoids, phenolics, tannins, terpenes | (Sulieman et al. 2024) |
| Rumex japonicus | Leaf extract | MIC: 1.96–62.5 μg/mL | In vitro antifungal assays | Antifungal activity; anthraquinones (rhein, emodin), flavonoids (rutin, quercetin); disrupt membrane, biofilm, spore germination | Xiao et al. (2024) |
| Rumex nervosus | Ethanolic, aqueous extracts/leaves, roots | Not specified | In vitro antifungal assays | Antifungal activity; complete inhibition of Aspergillus fumigatus ; against Candida albicans | Al‐Garadi et al. (2022) |
| Rumex aquaticus | 1,4‐Naphthoquinones, naphthalene derivatives | 6.25 μM musizin |
In vitro antiviral assays (HSV‐2) |
Antiviral activity; viral replication inhibition | (Rédei et al. 2019) |
| Rumex acetosa | Acetone‐water extract/aerial parts | IC₅₀ = 0.8 μg/mL (HSV‐1) | In vitro antiviral assays | Antiviral activity; proanthocyanidins, flavonoids; complete HSV‐1 inhibition | Gescher et al. (2011) |
| Rumex vesicarius | Aqueous extract/leaves, flowers | Dose‐dependent | In vitro antioxidant assays | Antioxidant activity; significant radical scavenging; phenolics, flavonoids identified | Gomaa and Saleh (2015) |
| Rumex patientia | Methanolic extract | Not specified | In vitro antioxidant assays | Antioxidant activity; halogenated flavan‐3‐ol and catechin potent DPPH scavengers | Demirezer, Kuruüzüm, et al. (2001) |
| Rumex abyssinicus | 80% Methanolic extract/rhizome | 125, 250, and 500 mg/kg orally (p.o.) | In vivo—CCl4‐induced hepatotoxicity/mice | ↓ ALT, AST, ALP at 500 mg/kg; histological preservation of hepatic structure | Adamu et al. (2020) |
| Rumex dentatus | Aqueous methanolic extract/leaves | 500 and 250 mg/kg orally (p. o.) | In vivo—paracetamol‐induced hepatotoxicity/mice | ↓ ALT, AST, ALP, bilirubin; histopathological protection | Saleem et al. (2014) |
| Rumex tingitanus | Hydroalcoholic extract | 250 mg/kg intraperitoneally | In vitro (antioxidant assays); in vivo—CCl4‐induced hepatotoxicity rats | ↓ MDA; ↑ antioxidant enzymes; compound 4′‐p‐acetylcoumaroyl luteolin showed strong antioxidant activity; no acute toxicity up to 2000 mg/kg orally; antidepressant effect in mice | Mhalla et al. (2018) |
| Rumex vesicarius | Ethanolic extracts/roots, leaves, fruits | 100 mg/kg orally (p.o.) | In vivo—CCl4‐induced hepatotoxicity/rats | ↑ regeneration of parenchymal cells | El‐Hawary et al. (2011) |
| Rumex vesicarius | Methanolic extract | 100 and 200 mg/kg orally (p.o.) | In vivo—CCl4‐induced hepatotoxicity/rats | ↓ liver damage; IC₅₀ (cytotoxicity in HepG2 cells): 563.33 μg/mL → low cytotoxicity, potential therapeutic use | Tukappa et al. (2015) |
| Rumex acetosa , R. thyrsiflorus | Phenolic extracts | 25–100 μg/mL | In vitro—HeLa, A431, MCF7, HaCaT cells | Antiproliferative activity; concentration‐dependent inhibition | Lajter et al. (2013) and Kucekova et al. (2011) |
| Rumex acetosa | Methanolic extract/leaves | 50 mg/kg intravenous | In vivo—normotensive, hypertensive rats | ↓ mean arterial pressure; cardiovascular effect | Qamar et al. (2018) |
| Rumex acetosa | Extract | 25–200 μg/mL | In vitro antiplatelet assays | Inhibition of platelet aggregation; MAPK, PI3K/Akt, Src regulation | Jeong et al. (2020) |
| Rumex crispus | Aqueous extract | Not specified | In vitro osteoclastogenesis assay | Suppressed osteoclast differentiation; MAPK/NF‐κB/NFATc1 inhibition | Shim et al. (2017) |
| Rumex nervosus | Methanolic extract | Not specified | In vivo—STZ‐induced diabetic nephropathy rats | ↓ oxidative stress; ↓ inflammation; ↓ apoptosis; Nrf2 activation | AlMousa et al. (2022) |
| Rumex abyssinicus | Hydromethanolic extract/root | 100–400 mg/kg orally | In vivo—diabetic mouse models | ↓ blood glucose; improved glucose tolerance | Yrga Adugna et al. (2022) |
| Rumex dentatus | Phenolic compounds | 50–200 mg/kg orally | In vivo—diabetic rats | ↓ hyperglycemia; ↓ oxidative stress; ↑ PPARγ | Elsayed et al. (2020) |
| Rumex obtusifolius | Ethanol extract/seeds | Not specified | In vivo—hyperglycemic rabbits | ↓ fasting glucose by 57.3%; improved lipid profile | Aghajanyan et al. (2018) |
| Rumex lunaria | Methanolic extract/leaves | 3 μM | In vitro α‐glucosidase assay | Significant α‐glucosidase inhibition | Froldi et al. (2020) |
| Rumex crispus | Methanolic extract/flowers, leaves | 7.3 and 112.0 μg/mL (IC₅₀) | In vitro α‐glucosidase assay; in vivo diabetic rats | Potent α‐glucosidase inhibition; antihyperglycemic effect | Aguila‐Muñoz et al. (2023) |
Abbreviations: ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; COX, cyclooxygenase; IC₅₀, half‐maximal inhibitory concentration; MAPK, mitogen‐activated protein kinase; MDA, malondialdehyde; MIC, minimum inhibitory concentration; NF‐κB, nuclear factor kappa B; NFATc1, nuclear factor of activated T‐cells 1; PPARγ, peroxisome proliferator‐activated receptor gamma; STZ, streptozotocin.
6. Clinical Studies
Species of the genus Rumex have been studied in diverse clinical contexts. R. acetosa is included as one of the five herbal components in the fixed combination extract BNO 1016 (Sinupret, Bionorica SE, Germany), developed for the treatment of sinusitis (Jund et al. 2012). Preclinical studies have demonstrated antimicrobial, antiviral, anti‐inflammatory, and secretolytic properties of the formulation. A Phase IIb/III clinical trial identified 160 mg, administered three times daily, as the optimal dose. This dosage was found to be effective and well tolerated over a 15‐day treatment period in patients with acute viral rhinosinusitis.
In dermatological applications, a randomized, double‐blind, placebo‐controlled trial evaluated the efficacy and safety of a 3% Rumex occidentalis S.Watson cream compared to a 4% hydroquinone (HQ) cream in the treatment of melasma among Filipino patients (Mendoza et al. 2014). Over 8 weeks, all treatment groups showed reductions in pigmentation as measured by the Melasma Area Severity Index (MASI) and mexameter readings. By week 8, the R. occidentalis group exhibited comparable or greater improvements than the HQ group, with both treatments rated similarly effective by investigators and participants. The study concluded that R. occidentalis cream is a safe and effective alternative to HQ for skin lightening in melasma management. In the context of oral health, a randomized, placebo‐controlled pilot study assessed the effects of a mouth rinse containing 0.8% proanthocyanidin‐enriched R. acetosa extract in systemically healthy carriers of Porphyromonas gingivalis (Selbach et al. 2023). Although no significant intergroup differences in microbiological or cytological parameters were observed, intragroup analysis showed a significant reduction in the Sulcular Bleeding Index and Approximal Plaque Index at days 7 and 14 in the Rumex group. The mouth rinse was well tolerated, with no serious adverse events reported.
7. Toxicity and Side Effects
Rumex plants contain notable amounts of oxalic acid, which can cause health issues when ingested in large quantities. High dietary oxalate intake is associated with secondary hyperoxaluria and an increased risk of calcium oxalate kidney stones. Oxalates also reduce the absorption of calcium and other minerals by forming insoluble complexes, decreasing their bioavailability. The risk of poisoning can be reduced by avoiding consumption of the cooking water, where oxalate concentration may be higher. Due to these risks, Rumex consumption should be avoided by individuals with kidney stones, gout, arthritis, rheumatism, or hyperacidity. The estimated lethal dose of oxalic acid in adults ranges from 15 to 30 g, with lower doses (< 5 g) sometimes causing fatal outcomes (A. Vasas et al. 2015). A summary of other clinically reported toxicities associated with Rumex species is provided in Table 3.
TABLE 3.
Reported human toxicities and adverse effects associated with Rumex species.
| Rumex species | Toxic components | Reported clinical effects | References |
|---|---|---|---|
| R. crispus (yellow dock, curly dock) | Oxalic acid; anthraquinone glycosides (emodin) | Fatal oxalate poisoning (hypocalcemia, metabolic acidosis, hepatic/renal failure) after large ingestion; also causes nausea, vomiting, diarrhea, electrolyte imbalances (hypokalemia), and rare immune thrombocytopenia from tea use | Reig et al. (1990) |
| R. acetosella (sheep sorrel) | Oxalic acid; anthraquinones (emodin, chrysophanol) | Diarrhea, cramping, and gastroenteritis with overuse; renal impairment and hepatotoxicity in large doses (oxalate nephropathy, elevated liver enzymes); contraindicated in kidney stone sufferers | Vasas et al. (2015) |
| R. acetosa (common sorrel) | Oxalic acid (abundant in leaves) |
Acute kidney injury (oxalate crystal nephropathy) reported in a child after excessive sorrel ingestion; risk of hypocalcemia and multi‐organ oxalosis; Llarge amounts can cause nausea and mucosal irritation |
Selçuk et al. (2015) |
| R. dentatus (toothed dock) | Anthraquinones Tannins |
Severe gastrointestinal distress (dysentery, bloody diarrhea, gastric pain) noted in ethnomedicine reports if used improperly as a purgative Symptoms of colic and dehydration can occur with high doses |
Qazi et al. (2022) |
| R. vesicarius (bladder dock) | Oxalic acid; anthraquinones (less in leaves) |
No acute human poisonings recorded; high oxalate content suggests risk of kidney stone formation if overconsumed Mild laxative effect; large portions may upset stomach or cause hyperacidity |
El‐Hawary et al. (2011) |
| R. obtusifolius (broadleaf dock) | Polyphenols oxalates |
Contact dermatitis from sap (skin rash upon handling leaves); infrequent ingestion can cause oropharyngeal irritation Not recommended as food due to potential skin and GI irritation |
Vasas et al. (2015) |
8. Limitations and Clinical Gaps
Although the genus Rumex exhibits a wide array of promising pharmacological activities, several critical limitations hinder its progression toward clinical application. The current evidence base is dominated by preclinical studies; human clinical trials evaluating safety, efficacy, and dosing remain virtually absent. A further constraint is the considerable heterogeneity in study design, with inconsistent methodologies for extract preparation, characterization, and phytochemical analysis, making cross‐study comparisons difficult. In addition, most studies do not systematically account for variables such as species differences, plant part selection, harvest timing, and environmental influences, all of which can affect phytochemical composition. Pharmacokinetic and bioavailability data for key bioactive compounds are scarce, and most reports do not address issues of solubility, absorption, metabolism, or tissue distribution. Toxicological profiles for many Rumex species, especially under chronic administration, remain incomplete, leaving gaps in our understanding of long‐term safety. Furthermore, herb–drug interaction potential has not been adequately explored, which is an important consideration in populations with polypharmacy. Addressing these limitations will be essential to reliably characterize the therapeutic potential of Rumex species and guide future research toward clinically meaningful outcomes.
9. Conclusion and Future Prospects
Rumex species have long been integral to traditional medicine systems across diverse cultures. Notably, several phytoconstituents, including anthraquinones, flavonoids, tannins, stilbenes, and naphthalenes, have been identified in Rumex species, with a broad spectrum of biological effects such as antioxidant, antimicrobial, antidiabetic, hepatoprotective, and cytotoxic activities. Nevertheless, important gaps remain. First, the phytochemical profiles of many Rumex species remain poorly studied in relation to plant developmental stages. Environmental factors can also significantly influence the yield and composition of secondary metabolites. Second, although in vitro and in vivo studies provide encouraging data on biological activities, there is a notable lack of well‐designed clinical trials and standardized extracts. Third, current knowledge about the pharmacokinetics and bioavailability of key Rumex phytoconstituents is limited, which hinders the translation of promising preclinical findings into effective therapeutic applications. Given that compounds such as emodin, chrysophanol, flavonoids and tannins exhibit poor bioavailability in their native form, future research should prioritize formulation strategies that improve solubility, absorption, and stability. However, such enhancements must be approached cautiously, as increased bioavailability could also elevate the risk of toxicity, particularly for compounds with narrow therapeutic windows. Toxicological evaluations are scarce and scattered, particularly for lesser‐studied species and chronic administration models. Therefore, detailed toxicokinetic investigations and long‐term toxicity assessments are needed. In conclusion, Rumex species represent a rich but underutilized reservoir of bioactive compounds with substantial pharmacological potential. Future research should aim to (i) explore interspecies phytochemical variability in relation to phenology and habitat, (ii) develop standardized bioactive extracts, (iii) conduct pharmacokinetic and clinical studies and (iv) rigorously evaluate toxicological safety. These efforts would pave the way toward the rational development of Rumex‐based therapeutics and functional products, fully integrating traditional knowledge with modern biomedical approaches.
Author Contributions
Aya Khouchlaa: writing – original draft, writing – review and editing, visualization, validation, methodology, investigation, data curation. William N. Setzer: writing – original draft, investigation, data curation, validation, methodology, visualization, writing – review and editing. Daniela Calina: writing – review and editing, writing – original draft, investigation, validation, visualization, methodology, project administration, supervision, data curation. Shahira Mohamed Ezzat: investigation, writing – original draft, writing – review and editing, visualization, validation, methodology, supervision, data curation. Miquel Martorell: writing – original draft, writing – review and editing, visualization, validation, methodology, investigation, data curation, supervision. Javad Sharifi‐Rad: writing – original draft, writing – review and editing, project administration, supervision, data curation, methodology, validation, visualization, investigation, conceptualization. Seham Salaheldin Elhawary: writing – original draft, writing – review and editing, visualization, validation, methodology, investigation, data curation. Balakyz Yeskaliyeva: writing – original draft, writing – review and editing, visualization, validation, methodology, investigation, data curation. Farid Noshey Kirollos: writing – original draft, writing – review and editing, visualization, validation, methodology, investigation, data curation.
Funding
The authors have nothing to report.
Conflicts of Interest
The authors declare no conflicts of interest.
Mohamed Gohar, M. , Ezzat S. M., Yeskaliyeva B., et al. 2025. “ Rumex Species: Phytochemistry, Pharmacology and Nutritional Potential for Food and Health Applications.” Food Science & Nutrition 13, no. 12: e71300. 10.1002/fsn3.71300.
Contributor Information
Daniela Calina, Email: daniela.calina@umfcv.ro.
Javad Sharifi‐Rad, Email: javadsharifirad@uees.edu.ec.
Miquel Martorell, Email: mmartorell@udec.cl.
Data Availability Statement
The authors have nothing to report.
References
- Adamu, B. A. , Emiru Y. K., Sintayehu B., Araya E. M., Periasamy G., and Gebrelibanos Hiben M.. 2020. “In Vivo Hepatoprotective and In Vitro Radical Scavenging Activities of Extracts of Rumex abyssinicus Jacq. Rhizome.” Journal of Experimental Pharmacology 12: 221–231. 10.2147/jep.S258566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aghajanyan, A. , Nikoyan A., and Trchounian A.. 2018. “Biochemical Activity and Hypoglycemic Effects of Rumex obtusifolius L. Seeds Used in Armenian Traditional Medicine.” BioMed Research International 2018: 4526352. 10.1155/2018/4526352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguila‐Muñoz, D. G. , Jiménez‐Montejo F. E., López‐López V. E., et al. 2023. “Evaluation of α‐Glucosidase Inhibition and Antihyperglycemic Activity of Extracts Obtained From Leaves and Flowers of Rumex crispus L.” Molecules 28, no. 15: 5760. 10.3390/molecules28155760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akay, S. , Yüksel G., and Özad Düzgün A.. 2024. “Investigation of Antibiofilm and Antibacterial Properties of Green Synthesized Silver Nanoparticles From Aqueous Extract of Rumex sp.” Applied Biochemistry and Biotechnology 196, no. 2: 1089–1103. 10.1007/s12010-023-04592-w. [DOI] [PubMed] [Google Scholar]
- Aleksic Sabo, V. , Svircev E., Mimica‐Dukic N., Orcic D., Narancic J., and Knezevic P.. 2020. “Anti‐ Acinetobacter baumannii Activity of Rumex crispus L. and Rumex sanguineus L. Extracts.” Asian Pacific Journal of Tropical Biomedicine 10, no. 4: 172–182. [Google Scholar]
- Al‐Farhan, A. H. , Asiri K. A., Raju M., Hichri F., and Abu‐Melha A. S.. 2022. “In Vitro Antimicrobial Activity of Medicinal Plant Rumex nervosus Against Selected Oral Pathogens.” Journal of Pharmacy & Bioallied Sciences 14, no. Suppl 1: S753–s756. 10.4103/jpbs.jpbs_188_22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al‐Garadi, M. A. , Qaid M. M., Alqhtani A. H., Pokoo‐Aikins A., and Al‐Mufarrej S. I.. 2022. “In Vitro Phytochemical Analysis and Antibacterial and Antifungal Efficacy Assessment of Ethanolic and Aqueous Extracts of Rumex nervosus Leaves Against Selected Bacteria and Fungi.” Veterinary World 15, no. 11: 2725–2737. 10.14202/vetworld.2022.2725-2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- AlMousa, L. A. , AlFaris N. A., Alshammari G. M., et al. 2022. “ Rumex nervosus Could Alleviate Streptozotocin‐Induced Diabetic Nephropathy in Rats by Activating Nrf2 Signaling.” Science Progress 105, no. 2: 368504221102751. 10.1177/00368504221102751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ann, S. L. F. , and Kron K. A.. 2003. “rbcL Phylogeny and Character Evolution in Polygonaceae.” Systematic Botany 28, no. 2: 326–332. [Google Scholar]
- Başkan, S. , Daut‐Ozdemir A., Günaydin K., and Erim F. B.. 2007. “Analysis of Anthraquinones in Rumex crispus by Micellar Electrokinetic Chromatography.” Talanta 71, no. 2: 747–750. 10.1016/j.talanta.2006.05.018. [DOI] [PubMed] [Google Scholar]
- Batanouny, K. H. , Aboutabl E., Shabana M., and Soliman F.. 1999. Wild Medicinal Plants in Egypt: An Inventory to Support Conservation and Sustainable Use. Palm Press. [Google Scholar]
- Borsch, T. , Berendsohn W., Dalcin E., et al. 2020. “World Flora Online: Placing Taxonomists at the Heart of a Definitive and Comprehensive Global Resource on the World's Plants.” Taxon 69, no. 6: 1311–1341. 10.1002/tax.12373. [DOI] [Google Scholar]
- Bouafia, A. , Laib I., Laouini S. E., et al. 2025. “Comprehensive Bioactivities and Phytochemical Profiling of Rumex vesicarius: Antioxidant Potential, Anti‐Diabetic Properties, and Anti‐Biofilm Effects Under Thermal Treatment.” Chemistry & Biodiversity 22: e202500329. 10.1002/cbdv.202500329. [DOI] [PubMed] [Google Scholar]
- Boulos, L. 1999. & 2005. Flora of Egypt. Al Hadara Publishing. [Google Scholar]
- Burt, S. 2004. “Essential Oils: Their Antibacterial Properties and Potential Applications in Foods—A Review.” International Journal of Food Microbiology 94, no. 3: 223–253. 10.1016/j.ijfoodmicro.2004.03.022. [DOI] [PubMed] [Google Scholar]
- Chelly, M. , Chelly S., Ben Salah H., et al. 2020. “Characterization, Antioxidant and Protective Effects of Edible Rumex roseus on Erythrocyte Oxidative Damage Induced by Methomyl.” Journal of Food Measurement and Characterization 14, no. 1: 229–243. 10.1007/s11694-019-00285-3. [DOI] [Google Scholar]
- Chelly, M. , Chelly S., Occhiuto C., et al. 2021. “Comparison of Phytochemical Profile and Bioproperties of Methanolic Extracts From Different Parts of Tunisian Rumex roseus .” Chemistry & Biodiversity 18, no. 6: e2100185. 10.1002/cbdv.202100185. [DOI] [PubMed] [Google Scholar]
- Clark, J. E. , Foresti R., Green C. J., and Motterlini R.. 2000. “Dynamics of Haem Oxygenase‐1 Expression and Bilirubin Production in Cellular Protection Against Oxidative Stress.” Biochemical Journal 348, no. Pt 3: 615–619. [PMC free article] [PubMed] [Google Scholar]
- Cos, P. , Hermans N., De B. T., et al. 2002. “Antiviral Activity of Rwandan Medicinal Plants Against Human Immunodeficiency Virus Type‐1 (HIV‐1).” Phytomedicine 9, no. 1: 62–68. 10.1078/0944-7113-00083. [DOI] [PubMed] [Google Scholar]
- Cunnane, S. C. 2003. “Problems With Essential Fatty Acids: Time for a New Paradigm?” Progress in Lipid Research 42, no. 6: 544–568. 10.1016/s0163-7827(03)00038-9. [DOI] [PubMed] [Google Scholar]
- Demirezer, L. O. , Kuruüzüm‐Uz A., Bergere I., Schiewe H. J., and Zeeck A.. 2001. “The Structures of Antioxidant and Cytotoxic Agents From Natural Source: Anthraquinones and Tannins From Roots of Rumex patientia .” Phytochemistry 58, no. 8: 1213–1217. 10.1016/s0031-9422(01)00337-5. [DOI] [PubMed] [Google Scholar]
- Demirezer, O. , Kuruüzüm A., Bergere I., Schiewe H. J., and Zeeck A.. 2001. “Five Naphthalene Glycosides From the Roots of Rumex patientia .” Phytochemistry 56, no. 4: 399–402. 10.1016/s0031-9422(00)00368-x. [DOI] [PubMed] [Google Scholar]
- Deng, Y. M. , Wu B. J., Witting P. K., and Stocker R.. 2004. “Probucol Protects Against Smooth Muscle Cell Proliferation by Upregulating Heme Oxygenase‐1.” Circulation 110, no. 13: 1855–1860. 10.1161/01.Cir.0000142610.10530.25. [DOI] [PubMed] [Google Scholar]
- Desta, B. 1995. “Ethiopian Traditional Herbal Drugs. Part I: Studies on the Toxicity and Therapeutic Activity of Local Taenicidal Medications.” Journal of Ethnopharmacology 45, no. 1: 27–33. 10.1016/0378-8741(94)01191-2. [DOI] [PubMed] [Google Scholar]
- El Gazzar, A. F. , Shalabi L., Eisa A., and Khattab A.. 2023. “Computer‐Generated Keys to the Flora of Egypt. 11. The Polygonaceae.” Taeckholmia 43, no. 1: 68–87. 10.21608/taec.2023.188149.1047. [DOI] [Google Scholar]
- Elfotoh, M. A. , Shams K. A., Anthony K. P., et al. 2013. “Lipophilic Constituents of Rumex vesicarius L. and Rumex dentatus L.” Antioxidants (Basel) 2, no. 3: 167–180. 10.3390/antiox2030167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El‐Hawary, S. A. , Sokkar N. M., Ali Z. Y., and Yehia M. M.. 2011. “A Profile of Bioactive Compounds of Rumex vesicarius L.” Journal of Food Science 76, no. 8: C1195–C1202. 10.1111/j.1750-3841.2011.02370.x. [DOI] [PubMed] [Google Scholar]
- Elsayed, R. H. , Kamel E. M., Mahmoud A. M., et al. 2020. “ Rumex dentatus L. Phenolics Ameliorate Hyperglycemia by Modulating Hepatic Key Enzymes of Carbohydrate Metabolism, Oxidative Stress and PPARγ in Diabetic Rats.” Food and Chemical Toxicology 138: 111202. 10.1016/j.fct.2020.111202. [DOI] [PubMed] [Google Scholar]
- Elzaawely, A. A. , and Tawata S.. 2012. “Antioxidant Capacity and Phenolic Content of Rumex dentatus L. Grown in Egypt.” Journal of Crop Science and Biotechnology 15, no. 1: 59–64. 10.1007/s12892-011-0063-x. [DOI] [Google Scholar]
- Eom, T. , Kim E., and Kim J. S.. 2020. “In Vitro Antioxidant, Antiinflammation, and Anticancer Activities and Anthraquinone Content From Rumex crispus Root Extract and Fractions.” Antioxidants 9, no. 8: 726. 10.3390/antiox9080726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farooq, M. , Abutaha N., Mahboob S., Baabbad A., Almoutiri N. D., and Wadaan M.. 2020. “Investigating the Antiangiogenic Potential of Rumex vesicarius (Humeidh), Anticancer Activity in Cancer Cell Lines and Assessment of Developmental Toxicity in Zebrafish Embryos.” Saudi Journal of Biological Sciences 27, no. 2: 611–622. 10.1016/j.sjbs.2019.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatima, N. , Zia M. A., Riaz‐ur‐Rehman Rizvi Z. F., Ahmad S., Mirza B., and Chaudhary M. F.. 2009. “Biological Activities of Rumex dentatus L: Evaluation of Methanol and Hexane Extracts.” African Journal of Biotechnology 8: 6945–6951. [Google Scholar]
- Froldi, G. , González D. L., Rosteghin F., et al. 2020. “α‐Glucosidase and Glycation Inhibitory Activities of Rumex lunaria Leaf Extract: A Promising Plant Against Hyperglycaemia‐Related Damage.” Natural Product Research 34, no. 23: 3418–3422. 10.1080/14786419.2019.1569655. [DOI] [PubMed] [Google Scholar]
- Gam, H. J. , Injamum‐Ul‐Hoque M., Kang Y., et al. 2024. “Allelopathic Effect of the Methanol Extract of the Weed Species‐Red Sorrel (Rumex acetosella L.) on the Growth, Phytohormone Content and Antioxidant Activity of the Cover Crop – White Clover (Trifolium repens L.).” BMC Plant Biology 24, no. 1: 523. 10.1186/s12870-024-05240-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gautam, R. , Karkhile K. V., Bhutani K. K., and Jachak S. M.. 2010. “Anti‐Inflammatory, Cyclooxygenase (COX)‐2, COX‐1 Inhibitory, and Free Radical Scavenging Effects of Rumex nepalensis .” Planta Medica 76, no. 14: 1564–1569. 10.1055/s-0030-1249779. [DOI] [PubMed] [Google Scholar]
- Gescher, K. , Hensel A., Hafezi W., Derksen A., and Kühn J.. 2011. “Oligomeric Proanthocyanidins From Rumex acetosa L. Inhibit the Attachment of Herpes Simplex Virus Type‐1.” Antiviral Research 89, no. 1: 9–18. 10.1016/j.antiviral.2010.10.007. [DOI] [PubMed] [Google Scholar]
- Getie, M. , Gebre‐Mariam T., Rietz R., et al. 2003. “Evaluation of the Anti‐Microbial and Anti‐Inflammatory Activities of the Medicinal Plants Dodonaea viscosa, Rumex nervosus and Rumex abyssinicus .” Fitoterapia 74, no. 1–2: 139–143. 10.1016/s0367-326x(02)00315-5. [DOI] [PubMed] [Google Scholar]
- Ghanem, A. , Ali M. A., Elkady M. A., et al. 2023. “ Rumex vesicarius L. Boosts the Effectiveness of Sorafenib in Triple‐Negative Breast Cancer by Downregulating BCl2, mTOR, and JNK, and Upregulating p21 Expression.” Pathology, Research and Practice 250: 154807. 10.1016/j.prp.2023.154807. [DOI] [PubMed] [Google Scholar]
- Gillani, S. W. , Ahmad M., Ali M. A., et al. 2024. “Phyto‐Ecological Studies and Distribution Patterns of Subfamily Polygonoideae in Relation to Edaphic Factors Across Diverse Ecological Zones.” Heliyon 10, no. 17: e36571. 10.1016/j.heliyon.2024.e36571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ginovyan, M. , Javrushyan H., Hovhannisyan S., et al. 2024. “5‐Fluorouracil and Rumex obtusifolius Extract Combination Trigger A549 Cancer Cell Apoptosis: Uncovering PI3K/Akt Inhibition by In Vitro and In Silico Approaches.” Scientific Reports 14, no. 1: 14676. 10.1038/s41598-024-65816-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ginovyan, M. , Javrushyan H., Petrosyan G., et al. 2023. “Anti‐Cancer Effect of Rumex obtusifolius in Combination With Arginase/Nitric Oxide Synthase Inhibitors via Downregulation of Oxidative Stress, Inflammation, and Polyamine Synthesis.” International Journal of Biochemistry & Cell Biology 158: 106396. 10.1016/j.biocel.2023.106396. [DOI] [PubMed] [Google Scholar]
- Gomaa, S. , and Saleh N. M.. 2015. “Medicinal Attributes of Rumex vesicarius (Polygonaceae) Growing in Sakaka, Al‐Jouf, Saudi Arabia.” Medical Journal of Cairo University 82: 917–926. [Google Scholar]
- Hasan, A. , Ahmed I., Jay M., and Voirin B.. 1995. “Flavonoid Glycosides and an Anthraquinone From Rumex chalepensis .” Phytochemistry 39, no. 5: 1211–1213. 10.1016/0031-9422(95)00071-e. [DOI] [PubMed] [Google Scholar]
- Hawas, U. W. , Ahmed E. F., Abdelkader A. F., and Taie H. A. A.. 2011. “Biological Activity of Flavonol Glycosides From Rumex dentatus Plant, an Egyptian Xerophyte.” Journal of Medicinal Plants Research 5, no. 17: 4239–4243. [Google Scholar]
- Hill‐Kapturczak, N. , Thamilselvan V., Liu F., Nick H. S., and Agarwal A.. 2001. “Mechanism of Heme Oxygenase‐1 Gene Induction by Curcumin in Human Renal Proximal Tubule Cells.” American Journal of Physiology. Renal Physiology 281, no. 5: F851–F859. 10.1152/ajprenal.2001.281.5.F851. [DOI] [PubMed] [Google Scholar]
- Hossain, M. S. , Rashid A. H. M. A., Rahman M. M., and Sadhu S. K.. 2015. “Antioxidant, Antimicrobial and Antidiarrhoeal Activity of Methanolic Extract of Rumex maritimus L. (Polygonaceae).” Journal of Applied Pharmaceutical Science 5: 56–60. 10.7324/japs.2015.510.S10. [DOI] [Google Scholar]
- Humeera, N. , Kamili A. N., Bandh S. A., Amin S. U., Lone B. A., and Gousia N.. 2013. “Antimicrobial and Antioxidant Activities of Alcoholic Extracts of Rumex dentatus L.” Microbial Pathogenesis 57: 17–20. 10.1016/j.micpath.2013.02.001. [DOI] [PubMed] [Google Scholar]
- Hussain, F. , Ahmad B., Hameed I., Dastagir G., Sanaullah P., and Azam S.. 2010. “Antibacterial, Antifungal and Insecticidal Activities of Some Selected Medicinal Plants of Polygonaceae.” African Journal of Biotechnology 9: 5032–5036. [Google Scholar]
- Jain, P. , and Parkhe G.. 2018. “An Updated Review on Pharmacological Studies of Rumex nepalensi .” Pharma Innovation Jounal 7, no. 212: 175–181. [Google Scholar]
- Jeong, D. , Irfan M., Lee D. H., Hong S. B., Oh J. W., and Rhee M. H.. 2020. “ Rumex acetosa Modulates Platelet Function and Inhibits Thrombus Formation in Rats.” BMC Complementary Medicine 20, no. 1: 98. 10.1186/s12906-020-02889-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang, L. , Zhang S., and Xuan L.. 2007. “Oxanthrone C‐Glycosides and Epoxynaphthoquinol From the Roots of Rumex japonicus .” Phytochemistry 68, no. 19: 2444–2449. 10.1016/j.phytochem.2007.05.032. [DOI] [PubMed] [Google Scholar]
- Jund, R. , Mondigler M., Steindl H., Stammer H., Stierna P., and Bachert C.. 2012. “Clinical Efficacy of a Dry Extract of Five Herbal Drugs in Acute Viral Rhinosinusitis.” Rhinology 50, no. 4: 417–426. 10.4193/Rhino.12.015. [DOI] [PubMed] [Google Scholar]
- Kang, D. M. , Khalil A. A. K., Park W. S., et al. 2023. “Anti‐Helicobacter pylori Activity of Six Major Compounds Isolated From Rumex acetosa .” ACS Omega 8, no. 45: 42548–42554. 10.1021/acsomega.3c05282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kengne, I. C. , Feugap L. D. T., Njouendou A. J., et al. 2021. “Antibacterial, Antifungal and Antioxidant Activities of Whole Plant Chemical Constituents of Rumex abyssinicus .” BMC Complementary Medicine and Therapies 21, no. 1: 164. 10.1186/s12906-021-03325-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kerem, Z. , Bilkis I., Flaishman M. A., and Sivan L.. 2006. “Antioxidant Activity and Inhibition of Alpha‐Glucosidase by Trans‐Resveratrol, Piceid, and a Novel Trans‐Stilbene From the Roots of Israeli Rumex bucephalophorus L.” Journal of Agricultural and Food Chemistry 54, no. 4: 1243–1247. 10.1021/jf052436+. [DOI] [PubMed] [Google Scholar]
- Khaliq, T. , Waseem M. A., Mir S. A., Sultan P., Malik F. A., and Hassan Q. P.. 2023. “Isolation and Characterisation of Pharmaceutically Versatile Molecules From Rumex dentatus and Evaluation of Their Cytotoxic Activity Against Human Cancer Cell Lines.” Natural Product Research 37, no. 5: 857–862. 10.1080/14786419.2022.2092864. [DOI] [PubMed] [Google Scholar]
- Khan, I. , Khan U., Khan W., et al. 2024. “Antibacterial and Antibiofilm Potentials of Rumex dentatus Root Extract Characterized by HPLC‐ESI‐Q‐TOF‐MS.” Saudi Journal of Biological Sciences 31, no. 4: 103962. 10.1016/j.sjbs.2024.103962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khan, T. H. , Ganaie M. A., Siddiqui N. A., Alam A., and Ansari M. N.. 2014. “Antioxidant Potential of Rumex vesicarius L.: In Vitro Approach.” Asian Pacific Journal of Tropical Biomedicine 4, no. 7: 538–544. 10.12980/apjtb.4.2014c1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim, H. Y. , Jee H., Yeom J. H., Jeong H. J., and Kim H. M.. 2019. “The Ameliorative Effect of AST2017‐01 in an Ovalbumin‐Induced Allergic Rhinitis Animal Model.” Inflammation Research 68, no. 5: 387–395. 10.1007/s00011-019-01226-y. [DOI] [PubMed] [Google Scholar]
- Kim, H. Y. , Seo J. E., Lee H., Bae C. H., Ha K. T., and Kim S.. 2022. “ Rumex japonicus Houtt. Extract Suppresses Colitis‐Associated Colorectal Cancer by Regulating Inflammation and Tight‐Junction Integrity in Mice.” Frontiers in Pharmacology 13: 946909. 10.3389/fphar.2022.946909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim, J. S. , Song H. J., Ko S. K., Whang W. K., and Sohn U. D.. 2010. “Quercetin‐3‐O‐Beta‐d‐Glucuronopyranoside (QGC)‐Induced HO‐1 Expression Through ERK and PI3K Activation in Cultured Feline Esophageal Epithelial Cells.” Fitoterapia 81, no. 2: 85–92. 10.1016/j.fitote.2009.08.008. [DOI] [PubMed] [Google Scholar]
- Krgović, N. , Radović Selgrad J., Ilić T., et al. 2025. “Endemic Plant Rumex balcanicus: Phenolic Composition, Antioxidant Activity, Enzyme Inhibitory Potential and Molecular Docking Analysis.” Chemistry & Biodiversity 22, no. 2: e202401488. 10.1002/cbdv.202401488. [DOI] [PubMed] [Google Scholar]
- Kucekova, Z. , Mlcek J., Humpolicek P., Rop O., Valasek P., and Saha P.. 2011. “Phenolic Compounds From Allium schoenoprasum , Tragopogon pratensis and Rumex acetosa and Their Antiproliferative Effects.” Molecules 16, no. 11: 9207–9217. 10.3390/molecules16119207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lajter, I. , Zupkó I., Molnár J., et al. 2013. “Antiproliferative Activity of Polygonaceae Species From the Carpathian Basin Against Human Cancer Cell Lines.” Phytotherapy Research 27, no. 1: 77–85. 10.1002/ptr.4690. [DOI] [PubMed] [Google Scholar]
- Lee, N. J. , Choi J. H., Koo B. S., et al. 2005. “Antimutagenicity and Cytotoxicity of the Constituents From the Aerial Parts of Rumex acetosa .” Biological & Pharmaceutical Bulletin 28, no. 11: 2158–2161. 10.1248/bpb.28.2158. [DOI] [PubMed] [Google Scholar]
- Li, J. J. , Li Y. X., Li N., Zhu H. T., Wang D., and Zhang Y. J.. 2022. “The Genus Rumex (Polygonaceae): An Ethnobotanical, Phytochemical and Pharmacological Review.” Natural Products and Bioprospecting 12, no. 1: 21. 10.1007/s13659-022-00346-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li, Y. P. , Takamiyagi A., Ramzi S. T., and Nonaka S.. 2000. “Inhibitory Effect of Rumex japonicus Houtt on the Porphyrin Photooxidative Reaction.” Journal of Dermatology 27, no. 12: 761–768. 10.1111/j.1346-8138.2000.tb02278.x. [DOI] [PubMed] [Google Scholar]
- Litvinenko, Y. A. , and Muzychkina R. A.. 2003. “Phytochemical Investigation of Biologically Active Substances in Certain Kazakhstan Rumex Species. 1.” Chemistry of Natural Compounds 39, no. 5: 446–449. 10.1023/B:CONC.0000011117.01356.4c. [DOI] [Google Scholar]
- Liu, S. Y. , Sporer F., Wink M., et al. 1997. “Anthraquinones in Rheum Palmatum and Rumex dentatus (Polygonaceae), and Phorbol Esters in Jatropha curcas (Euphorbiaceae) With Molluscicidal Activity Against the Schistosome Vector Snails Oncomelania, Biomphalaria and Bulinus.” Tropical Medicine & International Health 2, no. 2: 179–188. 10.1046/j.1365-3156.1997.d01-242.x. [DOI] [PubMed] [Google Scholar]
- Mabberley, D. J. 1997. The Plant‐Book: A Portable Dictionary of the Vascular Plants. Cambridge University Press. [Google Scholar]
- Mendoza, C. G. , Singzon I. A., and Handog E. B.. 2014. “A Randomized, Double‐Blind, Placebo‐Controlled Clinical Trial on the Efficacy and Safety of 3% Rumex occidentalis Cream Versus 4% Hydroquinone Cream in the Treatment of Melasma Among Filipinos.” International Journal of Dermatology 53, no. 11: 1412–1416. 10.1111/ijd.12690. [DOI] [PubMed] [Google Scholar]
- Mhalla, D. , Zouari Bouassida K., Chawech R., et al. 2018. “Antioxidant, Hepatoprotective, and Antidepression Effects of Rumex tingitanus Extracts and Identification of a Novel Bioactive Compound.” BioMed Research International 2018: 7295848. 10.1155/2018/7295848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Middleton, E., Jr. , Kandaswami C., and Theoharides T. C.. 2000. “The Effects of Plant Flavonoids on Mammalian Cells: Implications for Inflammation, Heart Disease, and Cancer.” Pharmacological Reviews 52, no. 4: 673–751. [PubMed] [Google Scholar]
- Minh, T. N. , Van T. M., Andriana Y., et al. 2019. “Antioxidant, Xanthine Oxidase, α‐Amylase and α‐Glucosidase Inhibitory Activities of Bioactive Compounds From Rumex crispus L. Root.” Molecules 24, no. 21: 3899. 10.3390/molecules24213899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mulisa, E. , Asres K., and Engidawork E.. 2015. “Evaluation of Wound Healing and Anti‐Inflammatory Activity of the Rhizomes of Rumex abyssinicus J. (Polygonaceae) in Mice.” BMC Complementary and Alternative Medicine 15: 341. 10.1186/s12906-015-0878-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mungole, A. , and Chaturvedi A.. 2011. “Determination of Antibacterial Activity of Two Medicinally Important Indian Taxa.” Der Pharma Chemica 3: 83–89. [Google Scholar]
- Newman, D. J. , Cragg G. M., and Snader K. M.. 2000. “The Influence of Natural Products Upon Drug Discovery.” Natural Product Reports 17, no. 3: 215–234. 10.1039/a902202c. [DOI] [PubMed] [Google Scholar]
- Nishina, A. , Kubota K., and Osawa T.. 1993. “Antimicrobial Components, Trachrysone and 2‐Methoxystypandrone, in Rumex japonicus Houtt.” Journal of Agricultural and Food Chemistry 41, no. 10: 1772–1775. 10.1021/jf00034a047. [DOI] [Google Scholar]
- Orbán‐Gyapai, O. , Liktor‐Busa E., Kúsz N., et al. 2017. “Antibacterial Screening of Rumex Species Native to the Carpathian Basin and Bioactivity‐Guided Isolation of Compounds From Rumex aquaticus .” Fitoterapia 118: 101–106. 10.1016/j.fitote.2017.03.009. [DOI] [PubMed] [Google Scholar]
- Özenver, N. , Güvenalp Z., Kuruüzüm‐Uz A., and Demirezer L.. 2020. “Inhibitory Potential on Key Enzymes Relevant to Type II Diabetes Mellitus and Antioxidant Properties of the Various Extracts and Phytochemical Constituents From Rumex acetosella L.” Journal of Food Biochemistry 44, no. 10: e13415. 10.1111/jfbc.13415. [DOI] [PubMed] [Google Scholar]
- Pae, H. O. , Choi B. M., Oh G. S., et al. 2004. “Roles of Heme Oxygenase‐1 in the Antiproliferative and Antiapoptotic Effects of Nitric Oxide on Jurkat T Cells.” Molecular Pharmacology 66, no. 1: 122–128. 10.1124/mol.66.1.122. [DOI] [PubMed] [Google Scholar]
- Pan, Y. , Zhao X., Kim S. H., Kang S. A., Kim Y. G., and Park K. Y.. 2020. “Anti‐Inflammatory Effects of Beopje Curly Dock (Rumex crispus L.) in LPS‐Induced RAW 264.7 Cells and Its Active Compounds.” Journal of Food Biochemistry 44, no. 7: e13291. 10.1111/jfbc.13291. [DOI] [PubMed] [Google Scholar]
- Prakash Mishra, A. , Sharifi‐Rad M., Shariati M. A., et al. 2018. “Bioactive Compounds and Health Benefits of Edible Rumex Species‐A Review.” Cellular and Molecular Biology (Noisy‐le‐Grand, France) 64, no. 8: 27–34. [PubMed] [Google Scholar]
- Qamar, H. M. , Qayyum R., Salma U., Khan S., Khan T., and Shah A. J.. 2018. “Vascular Mechanisms Underlying the Hypotensive Effect of Rumex acetosa .” Pharmaceutical Biology 56, no. 1: 225–234. 10.1080/13880209.2018.1446031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qazi, N. G. , Khan A.‐u., Abbasi S. W., Malik I., and Naeem K.. 2022. “Effect of Rumex dentatus on Gastrointestinal Protection and Toxicology in Rodents via Investigating H+/K+‐ATPase, Calcium Channels, and PDE Mediated Signaling.” Frontiers in Pharmacology 13: 936161. 10.3389/fphar.2022.936161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu, Y. , Li A., Lee J., et al. 2020. “Inhibition of Jurkat T Cell Proliferation by Active Components of Rumex japonicus Roots via Induced Mitochondrial Damage and Apoptosis Promotion.” Journal of Microbiology and Biotechnology 30, no. 12: 1885–1895. 10.4014/jmb.2007.07018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quradha, M. M. , Khan R., Rehman M. U., and Abohajeb A.. 2019. “Chemical Composition and In Vitro Anticancer, Antimicrobial and Antioxidant Activities of Essential Oil and Methanol Extract From Rumex nervosus .” Natural Product Research 33, no. 17: 2554–2559. 10.1080/14786419.2018.1452009. [DOI] [PubMed] [Google Scholar]
- Ragab, A. E. , Al‐Ashmawy G. M., Afify S. R. E., El‐Feky O. A., and Ibrahim A. O.. 2025. “Synergistic Anticancer Effects of Cisplatin and Phenolic Aglycones of the Aerial Part of Rumex dentatus L. in Tongue Squamous Cell Carcinoma: Insights From Network Pharmacology and Biological Verification.” BMC Complementary Medicine and Therapies 25, no. 1: 25. 10.1186/s12906-024-04718-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahman, M. A. , Mossa J. S., Al‐Said M. S., and Al‐Yahya M. A.. 2004. “Medicinal Plant Diversity in the Flora of Saudi Arabia 1: A Report on Seven Plant Families.” Fitoterapia 75, no. 2: 149–161. 10.1016/j.fitote.2003.12.012. [DOI] [PubMed] [Google Scholar]
- Rao, K. , Ch S., Banji D., Sanhya S., and Mahesh V.. 2011. “A Study on the Nutraceuticals From the Genus Rumex .” Hygeia Journal for Drugs and Medicines 3, no. 1: 76–88. [Google Scholar]
- Reddy, N. , Sabbani V., and Choday V.. 2017. “In Vitro and In Vivo Antidiabetic Activity of Rumex vesicarius Leaves Extract in Streptozotocin Induced Diabetic Albino Wister Rats.” Journal of Diabetes & Metabolism 8, no. 6: 745. 10.4172/2155-6156.1000745. [DOI] [Google Scholar]
- Rédei, D. , Kúsz N., Rafai T., et al. 2019. “14‐Noreudesmanes and a Phenylpropane Heterodimer From Sea Buckthorn Berry Inhibit Herpes Simplex Type 2 Virus Replication.” Tetrahedron 75, no. 10: 1364–1370. 10.1016/j.tet.2019.01.050. [DOI] [Google Scholar]
- Reig, R. , Sanz P., Blanche C., Fontarnau R., Dominguez A., and Corbella J.. 1990. “Fatal Poisoning by Rumex crispus (Curled Dock): Pathological Findings and Application of Scanning Electron Microscopy.” Veterinary and Human Toxicology 32, no. 5: 468–470. [PubMed] [Google Scholar]
- Saleem, M. , Ahmed B., Karim M., et al. 2014. “Hepatoprotective Effect of Aqeous Methanolic Extract of Rumex dentatus in Paracetamol Induced Hepatotoxicity in Mice.” Bangladesh Journal of Pharmacology 9, no. 3: 284–289. 10.3329/bjp.v9i3.18874. [DOI] [Google Scholar]
- Sanchez, A. , and Kron K. A.. 2008. “Phylogenetics of Polygonaceae With an Emphasis on the Evolution of Eriogonoideae.” Systematic Botany 33, no. 1: 87–96. 10.1600/036364408783887456. [DOI] [Google Scholar]
- Sanchez, A. , Schuster T. M., Burke J. M., and Kron K. A.. 2011. “Taxonomy of Polygonoideae (Polygonaceae): A New Tribal Classification.” Taxon 60, no. 1: 151–160. 10.1002/tax.601013. [DOI] [Google Scholar]
- Selbach, S. , Klocke A., Peters U., et al. 2023. “Microbiological and Clinical Effects of a Proanthocyanidin‐Enriched Extract From Rumex acetosa in Periodontally Healthy Carriers of Porphyromonas gingivalis: A Randomized Controlled Pilot Study.” Planta Medica 89, no. 11: 1052–1062. 10.1055/a-1728-2249. [DOI] [PubMed] [Google Scholar]
- Selçuk, S. N. , Gülhan B., Düzova A., and Tekşam Ö.. 2015. “Acute Tubulointerstitial Nephritis due to Large Amount of Sorrel ( Rumex acetosa ) Intake.” Clinical Toxicology (Philadelphia, Pa.) 53, no. 5: 497. 10.3109/15563650.2015.1033061. [DOI] [PubMed] [Google Scholar]
- Shafiq, N. , Noreen S., Rafiq N., et al. 2020. “Isolation of Bioactive Compounds From Rumex hastatus Extract and Their Biological Evaluation and Docking Study as Potential Anti‐Oxidant and Anti‐Urease Agents.” Journal of Food Biochemistry 44, no. 8: e13320. 10.1111/jfbc.13320. [DOI] [PubMed] [Google Scholar]
- Shafiq, N. , Saleem M., Kousar S., et al. 2017. “Investigation of Genus Rumex for Their Biologically Active Constituents.” Research Journal of Life Sciences, Bioinformatics, Pharmaceutical and Chemical Sciences 2, no. 6. 10.26479/2017.0206.11. [DOI] [Google Scholar]
- Sharma, R. S. , Mishra V., Singh R., Seth N., and Babu C. R.. 2008. “Antifungal Activity of Some Himalayan Medicinal Plants and Cultivated Ornamental Species.” Fitoterapia 79, no. 7–8: 589–591. 10.1016/j.fitote.2008.06.004. [DOI] [PubMed] [Google Scholar]
- Shim, K. S. , Lee B., and Ma J. Y.. 2017. “Water Extract of Rumex crispus Prevents Bone Loss by Inhibiting Osteoclastogenesis and Inducing Osteoblast Mineralization.” BMC Complementary and Alternative Medicine 17, no. 1: 483. 10.1186/s12906-017-1986-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spedding, G. , Ratty A., and Middleton E. Jr. 1989. “Inhibition of Reverse Transcriptases by Flavonoids.” Antiviral Research 12, no. 2: 99–110. 10.1016/0166-3542(89)90073-9. [DOI] [PubMed] [Google Scholar]
- Süleyman, H. , Demirezer L. O., Kuruüzüm A., et al. 1999. “Antiinflammatory Effect of the Aqueous Extract From Rumex patientia L. Roots.” Journal of Ethnopharmacology 65, no. 2: 141–148. 10.1016/s0378-8741(98)00175-5. [DOI] [PubMed] [Google Scholar]
- Sulieman, A. M. E. , Abdallah E. M., Alanazi N. A., et al. 2024. “Bioactive Profiling of Rumex vesicarius L. From the Hail Region of Saudi Arabia: A Study on Its Phytochemical and Elemental Analysis, Antibiofilm, Antibacterial, Antioxidant Properties, and Molecular Docking Analysis.” Frontiers in Microbiology 15: 1421143. 10.3389/fmicb.2024.1421143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun, H. , Xiao D., Li X., et al. 2024. “Study on the Chemical Composition and Anti‐Fungi Activities of Anthraquinones and Its Glycosides From Rumex Japonicus Houtt.” Journal of Natural Medicines 78, no. 4: 929–951. 10.1007/s11418-024-01834-x. [DOI] [PubMed] [Google Scholar]
- Sweilam, S. H. , Abd El Hafeez M. S., Mansour M. A., and Mekky R. H.. 2024. “Unravelling the Phytochemical Composition and Antioxidant Potential of Different Parts of Rumex vesicarius L.: A RP‐HPLC‐MS‐MS/MS, Chemometrics, and Molecular Docking‐Based Comparative Study.” Plants (Basel) 13, no. 13: 1815. 10.3390/plants13131815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi, T. , Morita K., Akagi R., and Sassa S.. 2004. “Heme Oxygenase‐1: A Novel Therapeutic Target in Oxidative Tissue Injuries.” Current Medicinal Chemistry 11, no. 12: 1545–1561. 10.2174/0929867043365080. [DOI] [PubMed] [Google Scholar]
- Takhol, V. 1974. Students' Flora of Egypt. Cairo University. [Google Scholar]
- Taveira, M. , Guedes de Pinho P., Gonçalves R. F., Andrade P. B., and Valentão P.. 2009. “Determination of Eighty‐One Volatile Organic Compounds in Dietary Rumex induratus Leaves by GC/IT‐MS, Using Different Extractive Techniques.” Microchemical Journal 93, no. 1: 67–72. 10.1016/j.microc.2009.04.008. [DOI] [Google Scholar]
- Tukappa, N. K. A. , Londonkar R. L., Nayaka H. B., and Kumar C. B. S.. 2015. “Cytotoxicity and Hepatoprotective Attributes of Methanolic Extract of Rumex vesicarius L.” Biological Research 48, no. 1: 19. 10.1186/s40659-015-0009-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uzun, M. , and Demirezer L. O.. 2019. “Anti‐Aging Power of Rumex crispus L.: Matrixmetalloproteinases Inhibitor, Sun Protective and Antioxidant.” South African Journal of Botany 124: 364–371. 10.1016/j.sajb.2019.05.028. [DOI] [Google Scholar]
- Vasas, A. , Orbán‐Gyapai O., and Hohmann J.. 2015. “The Genus Rumex: Review of Traditional Uses, Phytochemistry and Pharmacology.” Journal of Ethnopharmacology 175: 198–228. 10.1016/j.jep.2015.09.001. [DOI] [PubMed] [Google Scholar]
- Warth, A. D. 1991. “Mechanism of Action of Benzoic Acid on Zygosaccharomyces bailii: Effects on Glycolytic Metabolite Levels, Energy Production, and Intracellular pH.” Applied and Environmental Microbiology 57, no. 12: 3410–3414. 10.1128/aem.57.12.3410-3414.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegiera, M. , Kosikowska U., Malm A., and Smolarz H.. 2011. “Antimicrobial Activity of the Extracts From Fruits of Rumex L. Species.” Open Life Sciences 6, no. 6: 1036–1043. 10.2478/s11535-011-0066-0. [DOI] [Google Scholar]
- Xiao, D. , Sun H., Li X., et al. 2024. “ Rumex japonicus Houtt. Leaves: The Chemical Composition and Anti‐Fungal Activity.” Journal de Mycologie Médicale 34, no. 4: 101513. 10.1016/j.mycmed.2024.101513. [DOI] [PubMed] [Google Scholar]
- Xiao, D. , Wang J., Zhong Y., et al. 2022. “Study on HPLC Fingerprint, Network Pharmacology, and Antifungal Activity of Rumex japonicus Houtt.” Journal of AOAC International 105, no. 6: 1741–1754. 10.1093/jaoacint/qsac079. [DOI] [PubMed] [Google Scholar]
- Yadav, S. , Kumar S., Jain P., et al. 2011. “Antimicrobial Activity of Different Extracts of Roots of Rumex nepalensis Spreng.” Indian Journal of Natural Products and Resources 2: 155–158. [Google Scholar]
- Yahya, M. A. 1990. Saudi Plants: A Phytochemical and Biological Approach. King Abdulaziz City for Science and Technology. [Google Scholar]
- Yang, H. R. , Lee H., Kim J. H., et al. 2019. “Therapeutic Effect of Rumex japonicus Houtt. On DNCB‐Induced Atopic Dermatitis‐Like Skin Lesions in Balb/c Mice and Human Keratinocyte HaCaT Cells.” Nutrients 11, no. 3: 573. 10.3390/nu11030573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang, Y. , Yan Y. M., Wei W., et al. 2013. “Anthraquinone Derivatives From Rumex Plants and Endophytic Aspergillus fumigatus and Their Effects on Diabetic Nephropathy.” Bioorganic & Medicinal Chemistry Letters 23, no. 13: 3905–3909. 10.1016/j.bmcl.2013.04.059. [DOI] [PubMed] [Google Scholar]
- Yildirim, A. , Mavi A., and Kara A. A.. 2001. “Determination of Antioxidant and Antimicrobial Activities of Rumex crispus L. Extracts.” Journal of Agricultural and Food Chemistry 49, no. 8: 4083–4089. 10.1021/jf0103572. [DOI] [PubMed] [Google Scholar]
- Yrga Adugna, B. , Mequanint Adinew G., Ayalew Getahun K., et al. 2022. “Evaluation of the Antidiabetic Activity of Hydromethanolic Roots Extracts of Rumex abyssinicus Jacq: (Polygonaceae) in Swiss Albino Mice.” Evidence‐Based Complementary and Alternative Medicine 2022: 5193250. 10.1155/2022/5193250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou, H. , Lu F., Latham C., Zander D. S., and Visner G. A.. 2004. “Heme Oxygenase‐1 Expression in Human Lungs With Cystic Fibrosis and Cytoprotective Effects Against Pseudomonas aeruginosa In Vitro.” American Journal of Respiratory and Critical Care Medicine 170, no. 6: 633–640. 10.1164/rccm.200311-1607OC. [DOI] [PubMed] [Google Scholar]
- Zhu, J. J. , Zhang C. F., Zhang M., and Wang Z. T.. 2006. “Studies on Chemical Constituents in Roots of Rumex dentatus .” Zhongguo Zhong Yao Za Zhi 31, no. 20: 1691–1693. [PubMed] [Google Scholar]
- Zohary, M. , and Feinbrun‐Dothan N.. 1966. Flora Palaestina. Israel Academy of Sciences and Humanities. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The authors have nothing to report.
