Skip to main content
Advances in Pharmacological and Pharmaceutical Sciences logoLink to Advances in Pharmacological and Pharmaceutical Sciences
. 2025 Feb 17;2025:8276090. doi: 10.1155/adpp/8276090

Phytochemical Composition of Ziziphus lotus (L.) Lam and Its Impact on the Metabolic Syndrome: A Review

Chaimae Alla 1, Amanat Ali 1, Afaf Mehiou 1, Youssra Salhi 1, Nourelhouda Bouanani 1, Abdelkhaleq Legssyer 1, Abderrahim Ziyyat 1,
PMCID: PMC11873318  PMID: 40035065

Abstract

The long-term pathological state known as metabolic syndrome is characterized by hypertension, insulin resistance diabetes, abdominal obesity, and hyperlipidemia. Seeking healthcare strategies with fewer side effects, such as herbal remedies, is preferable in terms of mitigating the negative consequences of synthetic medications. Ziziphus lotus (L.) (Rhamnaceae) or wild jujube, commonly known as “Sedra,” is one of the best choices as it contains a variety of phytochemicals and biologically active compounds. Several flavonoids and stilbenes have been recognized as the primary bioactive components in wild jujube, including rutin, hyperin, isoquercitrin, and resveratrol. These polyphenols are pharmacologically active and have broad-spectrum beneficial effects for reducing the risk factors associated with metabolic syndrome. They exhibit antioxidant and anti-inflammatory properties, regulate lipid metabolism, and possess antiobesity, antihypertensive, and antidiabetic characteristics. However, there are certain limitations to their therapeutic application, such as low bioavailability. Various strategies have been proposed to enhance their pharmacokinetic profile and therapeutic potential for future use. The main goal of this review is to explore the underlying mechanisms related to the therapeutic effects of wild jujube and its active compounds in the treatment and prevention of metabolic syndrome.

Keywords: diabetes, hypertension, metabolic syndrome, obesity and dyslipidemia, polyphenols, Ziziphus lotus (L.)

1. Introduction

The World Health Organization defines metabolic syndrome as a chronic pathological state. It features abdominal obesity, insulin resistance diabetes, hypertension, and hyperlipidemia [1]. The pathogenesis of metabolic syndrome results from the interplay of biological mechanisms such as low-grade chronic inflammation, oxidative stress, and hormonal imbalances. A key feature of the syndrome is insulin resistance. It is due to impaired signaling pathways in adipose tissue, skeletal muscle, and the liver. This predisposes the individual to hyperglycemia and dyslipidemia [2, 3]. Additionally, oxidative stress and inflammation may further drive metabolic syndrome by promoting endothelial dysfunction and thereby increasing cardiovascular risks [4].

The likelihood of metabolic syndrome increases due to several risk factors, such as an unhealthy diet, sedentarism, and environmental variables. Consequently, the management of metabolic syndrome needs to be multifactorial, rather than solely relying on clinical therapy at the time of diagnosis. These measures include adherence to a healthy lifestyle through balanced dietary habits, regular physical activity, and stress management [5]. Functional foods such as fiber, polyphenols, and omega-3 fatty acids have also showed preventive effects [6].

In recent years, the anticipated harmful side effects of synthetic medicines have drawn increasing attention to harness the potential of medicinal plants and natural phytochemicals in restoring metabolic balance [7]. Medicinal plants contain bioactive compounds such as flavonoids, anthocyanins, and phenolic acids that have showed potential against the symptoms of metabolic syndrome through mechanisms involving the reduction of oxidative stress, increase in insulin sensitivity, and modulation of inflammatory pathways and lipids metabolism. Flavonoids constitute a significant proportion of these bioactive chemicals [8]. Concurrently, plant extracts, foods rich in polyphenols, and pure polyphenols are being researched for their potential to prevent and/or treat disorders linked to metabolic syndrome [8]. For instance, 6-gingerol, a phenolic compound extracted from ginger, has been reported to effectively regulate AMP-activated protein kinase (AMPK) to enhance insulin sensitivity and modulate peroxisome proliferator–activated receptors (PPARs) involved in lipid metabolism [9]. Wild jujube (Ziziphus lotus [L.] [Z. lotus], Rhamnaceae) is one of the most popular herbal remedies used in folk medicine to cure and prevent a wide range of illnesses, including the risk factors of metabolic syndrome [10, 11]. Numerous phytochemicals and biologically active compounds have been identified in Z. lotus [12]. Among them, flavonoid and nonflavonoid polyphenols such as rutin, isoquercitrin, hyperin, and resveratrol are considered the primary bioactive components in wild jujube [1315]. They have demonstrated a wide range of pharmacological advantages, such as improved lipid profiles [16], increased blood glucose tolerance, and better regulation of inflammatory markers [17]. Likewise, Moringa oleifera, a therapeutic plant widely investigated for its medicinal effects, has also showed benefits in the prevention of blood glucose spikes, blood pressure, hyperlipidemia, and obesity. Several molecular mechanisms of these actions have been elucidated, including AMPK activation, β-hydroxy-β-methylglutaryl-coenzyme A (HMG-CoA) inhibition, α-glucosidase inhibition, and glucose transporter type 4 (GLUT4) expression augmentation. These findings underscore the broader potential of medicinal plants to affect multiple pathways associated with metabolic syndrome [18].

This review encompasses the underlying mechanisms of action and focuses on the recent research regarding the therapeutic effects of wild jujube and its active constituents in the management and prevention of metabolic syndrome.

2. Methodology

A comprehensive literature review was undertaken using search terms such as “medicinal uses,” “phytochemistry,” “metabolic syndrome,” “diabetes,” “obesity,” “dyslipidemia,” and “hypertension” coupled with the core search keyword “Z. lotus.” All the citable data were gathered from various online databases such as Google Scholar, Science Direct, PubMed, Web of Science, and Scopus. Only articles published in English were included. Specific inclusion and exclusion criteria guided the selection of studies.

  • The inclusion criteria include the following:
    • - The literature documenting the traditional medicinal applications, therapeutic properties, or ethnopharmacological value of Z. lotus
    • - The research detecting, purifying, or evaluating phytochemical components in Z. lotus.
    • - The research on Z. lotus and its bioactive components
    • - The research focusing on the components of metabolic syndrome, including diabetes, obesity, dyslipidemia, and hypertension
    • - Peer-reviewed articles: both experimental studies (in vitro, animal, and clinical) and pertinent review articles.
  • The exclusion criteria include the following:
    • - Studies on other species of Ziziphus without relevant data on Z. lotus
    • - Articles unrelated to metabolic syndrome or lacking experimental evidence
    • - Non–peer-reviewed publications

The following data were extracted: All reported medicinal uses of Z. lotus in Africa and the Middle East; the phytochemical data, including compounds found in Z. lotus, the type of extract (i.e., aqueous, methanolic, and ethanolic) and the region where the specimens were collected; study design (animal model, human clinical trial, or in vitro), the dose of Z. lotus or its bioactive compounds, the bioactive compounds investigated, specific outcomes (such as blood glucose reduction, lipid profile improvements, or anti-inflammatory effects), and proposed mechanism of action with an emphasis on molecular targets.

The integrated molecular drawing platform ChemDraw 15.0 (PerkinElmer, Waltham, MA, USA) was used to sketch the chemical structures.

3. Botanical Description of Z. lotus (L)

Z. lotus is a thorny and frutescent shrub, reaching up to 2-3 m in height m in height (Figure 1(a)). The stems are glabrous and branching, with long woody shoots that sprout in short green branches. These branches begin as herbaceous and then become woody and bare when their leaves fall off. The young branches are yellowish-green, longitudinally striated, and have internodes of about 1.5–3 cm. They are characterized by a zigzag branching pattern, resembling a sympodial structure (Figure 1(b)). The leaves measure 1-2 cm long and 7–9 mm wide. They are simple, ovate-oblong, obtuse, alternate, crenulated, glabrous, and shortly petiolate, with two prickly and pointed stipules, out of which one is shorter and curved (Figure 1(b)). The leaves have a shiny and bright green lamina, with a three-nerved stretch from the base to the upper margin. They are covered with a thin cuticle, possess an astringent flavor, and are odorless. These leaves fall in the autumn and re-emerge in spring [1922].

Figure 1.

Figure 1

The whole plant (a); zigzag branches, leaves, and flowers (b); fruits in the tree (c); and isolated fruit and seeds (d) of Z. lotus (L.). Photos (a), (b), and (c) were taken by Pr Elachouri Mostafa in the region of Abbou Lakhal near Figuig City (Morocco) and (d) by Alla Chaimae.

The inflorescence of the Z. lotus consists of a fascicle of 6–11 flowers; however, in most cases, only one fruit matures per inflorescence. The flowers are tiny and have a glabrous appearance throughout. The corolla is alternipetalous and yellowish, comprising five small petals. The calyx starts out green and gradually changes to yellow. The androecium's stamens are epipetalous, each with two styles, intrastaminal and joined by a disc. They also exhibit extensive dehiscence. The flowers are polygamous and mostly pentamerous (Figure 1(b)). The plant has an inferior ovary with two or three carpels that split at the stigma level. It contains two anatropous and integument ovules, distinguishing the gynoecium. The fruit ranges from yellow-orange to red in color (Figure 1(c)). At the ripened stage, it resembles an edible brown globular drupe with a diameter of 1–1.5 cm and can contain two round seeds (Figure 1(d)) [20, 22, 23].

4. Ecogeographical Distribution of Z. lotus (L.)

Z. lotus has a wide geographical and ecological range, highlighting its adaptability to diverse environmental conditions. It can be found in humid, subhumid, arid, semiarid, and Saharan climates [24], although it is more commonly present in dry and semiarid areas [25, 26]. This plant species thrives better in deep sandy soils [27, 28]. Z. lotus has a broad distribution in the Mediterranean region, especially in northern African countries such as Morocco, Algeria, Tunisia, Libya, and Egypt, as well as in some of northern European countries such as Spain, Cyprus, Sicily, and Greece [29]. Additionally, it has re-emerged in Yemen, on the island of Socotra, and across the Middle East in Palestine, Syria, and Türkiye [20, 24, 30, 31]. It is also grown in several Asian countries such as Iran, China, and South Korea [32].

5. Ethnomedicinal Uses of Z. lotus (L.)

Z. lotus (L.), commonly known as “Sedra,” is an important medicinal plant native to the arid Mediterranean regions [33]. It is used for the prevention and treatment of various ailments [12].

Based on the ethnobotanical surveys of medicinal plants conducted in Africa and the Middle East, 71 studies citing Z. lotus are reviewed and summarized in Table 1 and Figure 2. The countries included are Morocco, Algeria, Cameroon, Nigeria, Mauritania, Mauritius, Jordan, and Palestine.

Table 1.

Ethnomedicinal usage of different parts of Z. lotus (L.) in various countries.

Country Plant part used Medicinal uses References
Morocco Whole plant Smallpox, measles, boils, sores, abscesses, digestive and respiratory problems, circulatory and urinary complaints, pyelonephritis, diabetes, symptoms of COVID-19, urine retention, diuretic, renal, colic, kidney stones, urogenital, dermatological, cardiovascular diseases, rheumatism, typhoid infection, ear disorders, and skin problems [3445]
Stems Snake bites [34]
Leaves Haircare, renal problems, gastric lavage, liver diseases, helminthiasis, colds, heart diseases, diabetes, urolithiasis, hypertension, hair loss, abscesses, sedative, diarrhea, cardiac failure, gastralgia, boils, soars, urinary infections, stomach pain, kidney stones, pneumonia, ulcers, and anorexia [10, 11, 34, 4663]
Roots Diabetes, stomach pains, diarrhea, kidney stones, jaundice, pulmonary and heart diseases, urolithiasis, intestinal pain, cough, bronchitis, and pyelonephritis [46, 48, 54, 6468]
Fruits Renal problems, cardiac ailments, pulmonary infection, hemostatic, colic, throat pains, pectoral pain, irritation, diabetes, hepatitis, immune system, depression, urolithiasis, kidney stones, stomach pain, fever, vomiting, renal pain, cystitis, pyelonephritis, cough, dyspnea, ulcers, diarrhea, and anorexia [11, 37, 4648, 50, 51, 53, 56, 6062, 65, 67, 6971]
Seeds Kidney problems, digestion problems, intestinal comfort, bloating, and diabetes [7274]
Bark Constipation, tachycardia, hemorrhoids, liver edema, stomachache, and pyelonephritis [67, 75].

Algeria Whole plant Colorectal lung and breast cancers, bladder inflammation, prostate kidney and lung infections, jaundice, irritation, and lung affections [7679]
Leaves Eczema, cicatrization, inflammations, skin problems, breast cancer, eye problems, fever, stomach acidity, hypertension, heartburn, constipation, catarrhal, diabetes, and urinary infections [8089]
Roots Diabetes, toothache, gastrointestinal and liver disorders, inflammations, irritation and pectoral pain, pulmonary affections, jaundice, and stomachache [68, 9094]
Fruits Diabetes, respiratory problems, sedative, pectoral pain, emollient, defensive hedge, urine retention, inflammations, hypertension, and stomach acidity [83, 89, 95]

Nigeria Whole plant Cancer [96]

Mauritania Leaves Fever, diabetes, skin infections, toothache, hypertension, abdominal pain, and kidney problems [97]

Cameroun Whole plant Cough, tired, and food poisoning [98]

Mauritius Fruits Asthma, ulcers, and hemorrhoids [99]

Palestine Leaves Diarrhea [100]

Jordan Fruits Cough and measles [101]

Figure 2.

Figure 2

Number of citations for ailments treated with Z. lotus (L.) in ethnobotanical studies.

Figure 2 displays the number of citations from ethnobotanical studies investigating the use of Z. lotus to treat various ailments. It highlights which health conditions have the most extensive medicinal use of Z. lotus.

After analyzing the selected studies, it has been indicated that Z. lotus is extensively used in the treatment and control of diverse medical disorders such as gastrointestinal afflictions, diabetes, dermatological infections, renal dysfunctions, and respiratory ailments [10, 36, 37, 46, 100]. The indigenous medicinal applications of different parts of the plant are listed in Table 1. The leaves and fruits are the most used parts (35% and 25%, respectively), followed by whole plant (19%), roots (15%), barks (3%), and stems (3%) (Figure 3). Specifically, leaves are typically applied to treat conditions such as hypertension, diabetes, gastrointestinal problems, urinary tract issues, and skin infections [10, 4663], whereas fruits are employed to relieve respiratory conditions, skin ailments, and digestive infections [83, 89, 95].

Figure 3.

Figure 3

Percentage of plant parts used for ethnomedicinal purposes.

6. Phytochemistry of Z. lotus (L.)

6.1. Flavonoids

Z. lotus contains various parts, including leaves, branches, seeds, and root bark, that are major sources of flavonoid compounds contributing to its medicinal properties. A summary of these flavonoids is presented in Table 2.

Table 2.

Major bioactive compounds identified in different parts of Z. lotus (L.).

Major compounds Chemical constituents Plant part References
Flavonoids (flavonols, dihydrochalcones, flavones, and flavanones) Myricetin rutinoside Leaf and branch [13, 14, 102105]
Glycoside quercetin
Quercetin 3-O-rutinoside (rutin)
Quercetin-3-β-D-glucoside (isoquercitrin)
Quercetin-3-galactoside (hyperoside or hyperin)
Quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside)-7-O-rhamnoside
Quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside)
Quercetin-3-O-(2,6-di-O-rhamnosyl glucoside)-7-O-glucuronide
Glycoside kaempferol:
Kaempferol-3-O-(2,6-di-O-rhamnosyl-glucoside)
Kaempferol-O-hexoside
Kaempferol-3-O-rutinoside (nicotiflorin)
Kaempferol-3-O-(6-O-rhamnosyl-glucoside)
Phloretin-di-C-hexoside Leaf, root bark
Apigenin
Apigenin-O-hexoside-O-deoxyhexoside
Catechin
Epicatechin
Epicatechin gallate
Naringin
Isorhamnetin Fruit [14, 15, 105107]
Rutin
Hyperoside
Apigenin
Luteolin
Luteolin-7-O-glucoside
Apigenin 7-O-glucoside
Quercetin 3-O-rhamnoside-7-O-glucoside
Diosmin
Cirsiliol
Catechin
Phloretin-3′,5′-di-glucoside
Naringenin
Eriodictyol
Eriodictyol glycoside derivatives: Fruit [14, 15, 105107]
Eriodictyol-O-hexoside
Eriodictyol-O-pentoside
Eriodictyol-O-deoxyhexoside
Quercetin-3-O-galactoside Arial part [108, 109]
Quercetin-3-O-rhamnoside
Rutin
Chamaejasmin Seeds [110]
Glucoliquiritin apioside 5,6,7,8,3′,4′,5′-heptamethoxy flavanone robustaside D
Kaempferol-3-O-robinobioside
Gallocatechin
Luteolin 7-(6‴′-acetylallosyl-(1- > 3)-glucosyl-(1- > 2)-glucoside apigenin 7-methyl ether 5-(6″-malonylglucoside)

Stilbenes Resveratrol Fruit [14]

Phenolic acids p-Hydroxybenzoic acid Fruit [15, 106, 111, 112]
p-coumaric acid
Benzoic acid
Sinapic acid
p-coumaroyl glucose
Cinnamic acid derivatives
Galloyl shikimic acid
Trans-ferulic acid
Rosmarinic acid
Syringic acid
Protocatechuic acid
Gallic acid
Quinic acid
Caffeic acid derivatives
Gentisic acid Leaf [13, 14, 113]
Fumaric acid
Gallic acid
Chlorogenic acid
Caffeic acid
Vanillic acid
p-coumaric acid Leaf [13, 14, 113]
Benzoic acid
P-hydroxybenzoic acid

Alkaloids (cyclopeptide alkaloids) Lotusine A Root bark [114116]
Lotusine B
Lotusine C
Lotusine D
Lotusine E
Lotusine F
Lotusine G
Lotusanine A Aerial part [117]
Lotusanine B
Sanjoinenine
Sanjoinine F
Frangufoline

Saponins (dammarane type) Jujuboside A Root bark [118, 119]
Jujuboside C
Lotoside I
Lotoside II
Jujuboside B Leaf [120]
Jujubasaponine IV
Jujubogenin glycosides

Several reports have identified that Z. lotus contains substantial amounts of flavonoids, with primary emphasis on flavonols represented by kaempferol, quercetin, and myricetin. Flavonol glycosides have been ascertained as the main compounds identified in leaves and branch extracts (both aqueous and hydroethanolic). These include myricetin rutinoside, glycoside quercetin derivatives such as quercetin 3-O-rutinoside (rutin), quercetin-3-β-D-glucoside (isoquercitrin), quercetin-3-galactoside (hyperoside or hyperin), quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside)-7-O-rhamnoside, quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside), and quercetin-3-O-(2,6-di-O-rhamnosyl-glucoside)-7-O-glucuronide. Similarly, the glycoside kaempferol derivatives, including kaempferol-3-O-(2,6-di-O-rhamnosyl-glucoside), kaempferol-O-hexoside, kaempferol-3-O-rutinoside (nicotiflorin), and kaempferol-3-O-(6-O-rhamnosyl-glucoside). On the other hand, dihydrochalcones (phloretin: phloretin-di-C-hexoside), flavones (apigenin) and its glycoside derivatives (apigenin-O-hexoside-O-deoxyhexoside), flavan-3-ols (catechin, epicatechin, and epicatechin gallate), and naringin have also been found in leaves and root bark extracts, both in aqueous and hydroethanolic form from plants originating in Algeria and Morocco [13, 14, 102105]. The biochemical profile of fruit's aqueous and hydroethanolic extracts from Tunisia showed the presence of several flavonoids, for instance, flavonols: resveratrol, isorhamnetin, quercetin 3-O-rutinoside (rutin), and hyperoside; flavones: luteolin-7-O-glucoside, apigenin 7-O-glucoside, apigenin, luteolin, diosmin, and cirsiliol; 3 proanthocyanidins (catechin); 1 chalcone (phloretin-3′,5′-di-glucoside); and flavanones: naringenin, eriodictyol moiety, and its glucoside derivatives, notably eriodictyol glycoside derivatives: eriodictyol-O-hexoside, eriodictyol-O-pentoside, and eriodictyol-O-deoxyhexoside [14, 15, 105107]. On the other hand, the hydromethanolic extracts from the Moroccan plant included more quercetin 3-O-rhamnoside-7-O-glucoside [111]. In the flavonoids profile of aerial parts from Tunisia, quercetin-3-O-galactoside, quercetin-3-O-rhamnoside, and rutin were detected to be the most prevalent components in acetonic and hydroethanolic extracts [108, 109]. Furthermore, certain flavonoids were characterized in the ethanolic extract of seeds from Algeria. These compounds are referred to as biflavonoids, which include chamaejasmin; flavanones such as glucoliquiritin apioside, 5,6,7,8,3′,4′,5′-heptamethoxy flavanone, and robustaside D as an isoflavanone; and flavones: luteolin 7-(6′‴-acetylallosyl-(1- > 3)-glucosyl-(1- > 2)-glucoside, and apigenin 7-methyl ether 5-(6′-malonylglucoside). Kaempferol-3-O-robinobioside and gallocatechin were the only flavonols and flavan-3-ols identified, respectively [110].

6.2. Phenolic Acids

The identification of various bioactive compounds in different extracts of Z. lotus highlights its medicinal potential, particularly in the treatment of a wide range of ailments. The analysis of ethanolic and hydroethanolic fruit extracts from Tunisia, Morocco, and Algeria showed higher concentrations of phenolic acids compared to the methanolic extracts, such as p-hydroxybenzoic acid, p-coumaric acid, benzoic acid, sinapic acid, p-coumaroyl glucose, cinnamic acid derivatives, galloyl shikimic acid, trans-ferulic acid, rosmarinic acid, syringic acid, protocatechuic acid, gallic acid, quinic acid, and caffeic acid derivatives [15, 106, 111, 112]. In addition, two primary phenolic acids have also been revealed in the methanolic leaf extract from Algeria and Morocco Z. lotus, including fumaric acid and gentisic acid, whereas gallic acid, chlorogenic acid, caffeic acid, vanillic acid, p-coumaric acid, benzoic acid, and p-hydroxybenzoic acid have all been found to be present in the water extract [13, 14, 113].

In addition to the aforementioned nonflavonoid polyphenols, the fruits from Morocco also possess another prominent compound “resveratrol,” which belongs to the stilbene category [14]. A summary of the identified phenolic acids is provided in Table 2.

6.3. Alkaloids

Alkaloids are well known for their therapeutic potential in managing various aspects including obesity, insulin resistance, and hypertension [121]. Several cyclopeptide alkaloids were recovered from Z. lotus root bark of Tunisian provenance, such as lotusine A, B, C, D, E, F, and G [114116]. The other cyclopeptide alkaloids that have been identified in aerial parts were named lotusanine A and lotusanine B. These were isolated along with sanjoinenine, sanjoinine F, and frangufoline [117]. The alkaloid content in Tunisian Z. lotus was found to be lower than that of polyphenol compounds [109]. A summary of these alkaloids, along with their presence in different plant parts, is provided in Table 2.

6.4. Saponins

Saponins have showed promise in modulating lipid metabolism and improving insulin sensitivity [122]. It is worth mentioning that all parts of Z. lotus contain saponins, but higher contents were detected in the leaves (340 mg/100 g) [120]. Indeed, according to Renault et al. [119] and Maciuk et al. [118], numerous saponins belonging to the dammarane class have been identified in leaves, including jujuboside B, jujuba saponin IV, and three jujubogenin glycosides. Additionally, the root bark yielded four saponins of the same class, specifically jujuboside A, jujuboside C, lotoside I, and lotoside II [118, 119]. The summary of these saponins across the plant's parts can be found in Table 2.

6.5. Other Bioactive Compounds

In addition to alkaloids and saponins, Z. lotus contains a variety of bioactive compounds such as condensed tannins, secoiridoids, fatty acids, and vitamins, which have potential health benefits [25]. Z. lotus from Algeria and Morocco also contains condensed tannins, which were found to be present in the highest concentrations in the aqueous extract and the lowest in the petroleum ether extract [104, 123]. Higher levels of condensed tannins were observed in leaves [107, 124, 125]. Secoiridoids were first found in branches and then in leaves, with oleoside being the most abundant compound in branches and oleuropein in leaves from Algeria [105]. The richest sources of fatty acids in the fruit were in its aqueous and methanolic extracts, as well as in the seed oil. The principal substances among them included oleic acid, linoleic acid, and elaidic acid [125131]. Different parts of Z. lotus have also been reported to contain some vitamins such as C, A, and E; carotenoids; β-tocopherol; and δ-tocopherol [127, 128, 132].

6.6. Volatile Compounds

According to our knowledge, there is limited research on the essential oil analysis of Z. lotus. Bekkar et al. were the first to focus on the essential oils of Algerian Z. lotus leaves and reported the presence of abundant amounts of phthalate esters and terpenes. Diisooctyl phthalate was identified as the main compound (89.85%), followed by linalool (2.14%). Minor quantities of other bioactive components have also been identified in this plant, for example, thymol (0.56%), p-cymene (0.35%), caryophyllene (0.26%), 2-undecanone (0.20%), ɤ-terpinene (0.16%), linalyl acetate (0.14%), and methyl eugenol + 97 ION (0.13%). Due to its dominance, this essential oil has been categorized as a diisooctyl phthalate chemotype [133]. During the analysis of fruit essential oils from Algerian Z. lotus, some hydrocarbons were also identified, such as heptacosane (3.7%) and nonacosane (3.7%). They also detected sesquiterpenes such as α- and β-eudesmol [130].

A comprehensive study conducted by Letaief et al. in Tunisia unveiled the presence of various compounds in the essential oil extracted from the aerial parts and fruits of the plant. The analysis revealed the presence of significant amounts of sesquiterpenes in the aerial parts, including hexahydro-farnesyl acetone (23.2%), E-nerolidol (5.9%), α-calacorene (3%), and D-nerolidol (1.9%). Additionally, apocarotenoid derivatives such as geranylacetone (12.5%), trans-β-ionone (6%), 2-undecanone (2.9%), and damascenone (1.3%) were also identified. The essential oil extracted from the fruits predominantly contained an oxygenated sesquiterpene along with 2-pentadecanone (16.9%) [134].

7. Data Related to the Safety of Z. lotus (L.)

There is a common fallacy that herbal medicines are safe and do not cause any adverse reactions. No reliable scientific data are available about their safety, as some of them can result in potential adverse effects leading to injuries and life-threatening conditions [135]. Acute toxicity tests were performed on rats to examine the impact of a water extract of Tunisian Z. lotus root bark extracts on mice. The results indicated that intraperitoneal administration of various dosages (50–1000 mg/kg body weight [BW]) produced variable results. No mortality was seen at 50 mg/kg BW, whereas a dose level of 1000 mg/kg BW provoked 100% mortality in mice. The median lethal dose (LD50) value was determined at 400 mg/kg BW [136]. Similarly, the results from another study showed that the oral administration of different doses of aqueous extracts of Moroccan Z. lotus fruit (200–2000 mg/kg BW) did not result in any mortality or behavioral changes in the rats [137]. On the contrary, no morbidity or distinctive clinical signs were recorded during the observation period after the administration of a single dose of 5000 mg/kg BW of the seed oil in mice [138]. For Algerian Z. lotus, the leaf essential oil given at a dose of 5000 mg/kg BW did not produce any visible toxic effects; therefore, LD50 seemed to be greater than 5000 mg/kg BW [133]. On the other hand, it has been demonstrated that the aqueous extract obtained from the leaves, fed at 2000 mg/kg BW as a single dose, did neither cause any mortality nor apparent toxicity symptoms in rats. It was concluded that the LD50 value appeared to be more than 2000 mg/kg BW [139].

8. Z. lotus (L.) Extracts and the Prevention of Metabolic Syndrome

According to the National Cholesterol Education Program's (NCEP ATP III) definition, metabolic syndrome is suspected if three or more of the following five criteria are met: blood pressure greater than 130/85 mmHg, fasting triglyceride (TG) level greater than 40 mg/dL, fasting high-density lipoprotein (HDL-C) cholesterol level less than 40 mg/dL (for men) or 50 mg/dL (for women), fasting blood sugar level greater than 100 mg/dL, and waist circumference exceeding 102 cm for men or 88 cm for women [140]. The development of metabolic syndrome is influenced by many factors, including lifestyle, insulin resistance, genetic predisposition, being overweight or obese, and the natural process of aging. Many risk factors associated with metabolic syndrome may not display symptoms or signs, except for having a waist circumference [141].

The study investigating the antidiabetic and antihyperlipidemic activities of Z. lotus has showed that the aqueous extract from both fruits and leaves exhibited the greatest in vitro inhibitory impact against α-glucosidase (half-maximal inhibitory concentration (IC50): 8.66–27.95 μg/mL) and α-amylase (IC50: 20.40–31.91 μg/mL) compared to standard acarbose [14]. Furthermore, the aqueous extract of the fruit sustained average blood glucose levels, leading to a substantial attenuation in TG and total cholesterol (TC) and causing a rise in HDL-C in mice receiving a high-fat diet compared to those on a normal diet [142]. At dosages of 200 and 400 mg/kg BW, this extract reduced the atherogenic index and the TC/HDL-C ratio, effectively preventing atherosclerosis, which is directly linked to cardiovascular disease [143, 144]. The aqueous extract also significantly lowered the plasma levels of proinflammatory markers, including interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), as well as monocyte chemoattractant protein-1 (MCP-1) mRNA expression, and produced higher levels of anti-inflammatory cytokine IL-10 in hepatic and adipose tissues. These changes have shown to affect insulin signaling during obesity [145]. In addition to improving glucose tolerance and reducing insulin resistance in the homeostatic model, Z. lotus fruit powder improved the levels of circulating glucose and insulin in HFD-induced obesity in C57BL/6j mice. The expression of hepatic glucose-6-phosphatase (G-6-Pase) mRNA was downregulated, inhibiting the liver's ability to produce glucose. As evidenced by a decline in fatty acid synthase (FAS) and acetyl-CoA carboxylase 1 (ACC1) mRNA expression, Z. lotus fruit was found to lower lipogenesis [146]. Besides raising the PPARγ and carnitine palmitoyltransferase 1 alpha mRNA expression, which were lowered in obese mice, Z. lotus fruit appeared to increase the use of fatty acids as an energy source [147].

According to a study conducted by Benammar et al. in 2014, it was observed that the aqueous extracts obtained from the roots and leaves exhibited a substantial decline in blood glucose levels in streptozotocin (STZ)-induced type 2 diabetic rats. These extracts were also shown to expedite the recovery of glucose levels during the glucose tolerance test. Furthermore, as demonstrated by the measures of kinetics of red blood lysis and oxygen radical absorbance capacity, it was noticed that these extracts increased the antioxidant status in diabetic rats. Catalase and glutathione peroxidase activities were greatly enhanced, while glutathione reductase activity was restored [148].

9. The Bioactive Compounds of Z. lotus (L.) and Their Beneficial Effects on Metabolic Syndrome

Bioactive compounds are naturally occurring substances in foods that positively affect health. These substances can lower the chances of developing metabolic syndrome by positively affecting BW, blood pressure, blood glucose control, endothelial function, lipid profile, inflammation, and oxidative stress [149]. Z. lotus contains elements such as polyphenols, which actively participate in preventing the onset of metabolic syndrome. They exhibit anti-inflammatory and antioxidative properties and manage the oxidative stress linked to metabolic syndrome [25, 150]. The research indicates that the compounds found in Z. lotus can improve insulin sensitivity and regulate glucose metabolism [150, 151]. They also contribute to reducing adiposity by inhibiting adipogenesis which is crucial for addressing obesity [152]. The targeted effects of compounds derived from Z. lotus demonstrate their potential in preventing and managing the health conditions associated with metabolic syndrome [25]. Table 3 summarizes the main mechanisms.

Table 3.

Summary of the key mechanisms targeted by Z. lotus (L.) bioactive compounds.

Bioactive compound Mechanisms of action Impact on metabolism Impact on vascular function
Rutin - Inhibition of ACE
- Activation of insulin signaling (IR kinase activity, GLUT4 translocation)
- Inhibition of α-amylase and α-glucosidase
- Reduces the expression of G-6-Pase
- Decreases HMG-CoA
- Inhibits NOD-like receptor pyrin domain inflammasome pathways
- Reduces TNF-α and IFNγ
- Suppresses proinflammatory markers (TNF-α, MCP-1, IFNγ, IL-6, IL-2, and NF-κB)
- Suppressing the expression of NADPH oxidase 4.
- Reduces blood glucose levels
- Improves insulin sensitivity
- Reduces dyslipidemia (TC, TG, and LDL-C)
- Improved LPL and LCAT
- Reduces FBG, HbA1c
- Endothelium-dependent vasorelaxation through NO-guanylyl cyclase pathway, ATP-sensitive K+ channel activation
- Recovering NO generation
- Improves vascular reactivity and baroreflex sensitivity

Hyperin - Inhibition of α-glucosidase
- Inhibits α-amylase
- Upregulating the expression of PPARγ
- Inhibits NF-κB
- Reduces blood glucose levels and plasma lipids (TC, TG, and LDL-C)
- Improves insulin sensitivity
- Suppresses inflammation
-Endothelium-dependent and independent vasodilation through KCa channels and NO
- Increased nitric oxide bioavailability
- Suppression of chymase (Ang-II-forming enzyme)
- Reduces peripheral vascular resistance and coronary vessel medial thickness

Isoquercitrin - Inhibition of α-amylase
- Modulation of AMP-activated protein kinase (AMPK)
- Regulation of insulin signaling genes (IR, IRS-1, IRS-2, and PKB)
- Inhibition of Na+/K+-ATPase activity
- Attenuates NF-κB and TNF-α expression
- Improves blood glucose control
- Enhances insulin secretion
- Reduces dyslipidemia
- Inhibits adipogenesis.
- Suppression of oxidative stress, inflammation, and mitochondrial dysfunction
- Endothelium-dependent vasodilation
- Increase in nitric oxide and hydrogen sulfide in endothelial cells
- Bioavailability enhancement of bradykinin and prostaglandin I2
- Reduction in vascular resistance
- Protects endothelial cells by upregulating antiapoptotic proteins (Bcl-2) and downregulating proapoptotic proteins (p53, Bax, and caspase 3)
- Exhibits potent diuretic effects by increasing Na+ excretion while maintaining K+ balance

Resveratrol - Activation of SIRT1 and AMPK
- Modulation of PPARγ and inflammatory pathways
- Inhibition of α-glucosidase and lipid metabolism enzymes
- Improves FBG and HbA1c and insulin sensitivity
- Reduces glucose and lipid levels
- Decreases adipogenesis
- Endothelial NO generation via eNOS activation
- Vasodilation through improved NO bioavailability
- Suppression of Gi protein signaling in vascular smooth muscle

9.1. Flavonoids and Nonflavonoid Polyphenols

Many flavonoids and nonflavonoid polyphenols demonstrate favorable effects in mitigating the risk factors associated with metabolic syndrome [153155]. In this respect, our attention has been devoted to three flavonoids and one nonflavonoid (stilbene) polyphenol, the most abundant chemical substances found in the plant. In particular, the prominent polyphenols encompass rutin, hyperin, isoquercitrin, and resveratrol (Figure 4) [1315], which are effective in improving conditions such as obesity, diabetes, hypertension, and dyslipidemia (Figure 5).

Figure 4.

Figure 4

Chemical structures of Z. lotus (L.) main flavonoids and stilbene.

Figure 5.

Figure 5

Schematic diagram depicting the various mechanisms employed by bioactive compounds of Z. lotus (L.) in managing the metabolic syndrome. Increase (Inline graphic) or decrease (Inline graphic) after bioactive compounds supplementation.

9.1.1. Effects on Diabetes

Rutin, isoquercitrin, hyperin, and resveratrol are the bioactive compounds that have been mostly studied for their potential to reduce the risk of diabetes.

9.1.1.1. Lowering Blood Glucose Levels

Rutin at doses of 50 or 100 mg/kg BW has shown to effectively lower the fasting blood glucose (FBG) and glycated hemoglobin (HbA1c) levels in STZ-induced type 2 diabetic rat models [156158]. Isoquercitrin is also stated to reduce FBG levels in STZ-induced type 2 diabetic mice [159]. The expression of glucose transporter Type 2 was also found to be dramatically upregulated by isoquercitrin. In addition, the key glucose-regulating metabolic enzymes, such as glucose-6 phosphate dehydrogenase and phosphoenolpyruvate carboxykinase, were also well controlled [160]. Hyperin significantly reduces glycemia levels, protects the pancreatic beta-cells, and enhances insulin secretion in diabetic mice caused by an HFD/alloxan [161, 162]. Hyperin has showed to play a significant role in controlling blood glucose levels by improving the functions of pancreatic islets in rats. At doses of 25 and 50 mg/kg BW in STZ-induced type 2 diabetic rats, it significantly reduced the blood glucose levels after 120 min by increasing glycolysis and decreasing gluconeogenesis. These effects are comparable to the standard medication glibenclamide used for treating type 2 diabetes [163]. Resveratrol dramatically alleviates hyperglycemia and has a beneficial impact on insulin sensitivity in diabetes animal models generated by STZ or alloxan injection [164].

9.1.1.2. Improving the Insulin Sensitivity

Through stimulating the insulin receptor (IR) kinase activity, rutin can act as a glycemic control agent by activating the insulin signaling pathway, thereby increasing GLUT4 translocation and glucose uptake [165]. It also enhances the expression of PPARγ, improves glucose uptake by adipose tissues and skeletal muscles, and reduces insulin resistance [166]. Moreover, rutin can boost the insulin release triggered by hyperglycemia in rat beta-cells while preserving their capacity to sense glucose under high glucose conditions [167]. Rutin has also been reported to reduce insulin resistance by enhancing glucose uptake in insulin-resistant cells in FL83B hepatocytes exposed to high glucose levels and improving insulin signaling via phosphorylated protein kinase C activity suppression [168]. Isoquercitrin positively impacts insulin sensitivity by targeting dipeptidyl peptidase IV, with an IC50 value of 44.93 μg/mL [159]. Furthermore, in the HFD mice model, quercetin and its glycosides such as isoquercitrin averted insulin resistance by improving GLUT4 translocation in skeletal muscles via modulating AMPK-driven pathways [169]. The research conducted by Jayachandran et al. [160] showed that when STZ-induced diabetic rats were supplemented with isoquercitrin; it regulated the expression of genes involved in insulin signaling, such as insulin, IR, IRS-1, IRS-2, and protein kinase B (PKB). In human liver cancer cell line (HepG2) cells resistant to insulin, hyperin enhances the expression of the PPARγ protein [170]. Resveratrol has shown to improve FBG, insulin sensitivity, HbA1c, and homeostasis model assessment of insulin resistance values in participants with type 2 diabetes mellitus and obesity [171173]. It has also shown to upregulate the expression of activated Sirtuin 1 (SIRT1) and AMPK in skeletal muscles [174]. It also stimulates insulin production and improves skeletal muscles glucose absorption via the phosphoinositide 3-kinase—PKB signaling pathway. Likewise, it leads to a rise in the expression of GLUT4 in skeletal muscles and a decline in phosphoenolpyruvate carboxy kinase in the liver [175].

9.1.1.3. Inhibition of Enzymes Associated With Type 2 Diabetes

Rutin exhibits a potent inhibitory impact on the main enzymes associated with type 2 diabetes (α-amylase and α-glucosidase), with IC50 values of 26.27 and 22.61 μg/mL, respectively [176]. Isoquercitrin potently inhibits the α-glucosidase, with an IC50 of 85.82 μg/mL [177]. Hyperin shows a significant α-amylase inhibition effect, with an IC50 value of 491 μg/mL [178]. Resveratrol inhibits the α-glucosidase activity, and its ability is 2.5 times greater than that of acarbose (IC50 = 91 μg/mL) [179].

9.1.1.4. Reducing Oxidative Stress and Inflammation

Rutin reduces G-6-Pase, a crucial gluconeogenic enzyme, in the liver and kidney of diabetic rats by 31% and 37%, respectively [157, 180]. The protective effects of hyperin are associated with the recovery of intracellular redox equilibrium and mitochondrial function via the suppression of mitogen-activated protein kinase signaling, caspase-dependent apoptosis, calcium release, and thioredoxin-interacting protein expression [162]. Hyperin eliminates hyperglycemia-induced vascular inflammation by suppressing monocyte adhesion, cell adhesion molecule expression, reactive oxygen species formation, and nuclear factor (NF-κB) activation [181]. Resveratrol at 5 mg/kg BW can avert the hyperglycemia-induced oxidative stress by boosting oxidative markers such as the lipid peroxidation index and nitrite/nitrate content, and by decreasing the glutathione ratio. It also boosts the enzymic antioxidants such as superoxide dismutase in hepatic tissues. The levels of hepatic proinflammatory cytokines TNF-α and IL-6 are substantially reduced [182, 183]. Resveratrol pretreatment has showed to impede the caspase-3 and poly(ADP-ribose) polymerase (PARP) activation by STZ injection, thus preventing pancreatic beta-cell apoptosis [184].

9.1.2. Effects on Obesity

9.1.2.1. Inflammation

Rutin has been suggested to reduce the TNF-α and interferon-gamma (IFNγ) expression, thereby shielding the liver from excess fat-induced inflammation [185]. Additionally, rutin has been reported to suppress the expression of proinflammatory markers, such as TNF-α, MCP-1, IFNγ, IL-6, IL-2, NF-κB, myeloid differentiation primary response gene 88, and lipopolysaccharide-binding protein in adipose tissues [186, 187]. Isoquercitrin, on the other hand, modulated the elevated proinflammatory secretion induced by obesity in HFD mice, by affecting adipokine expression (decreased leptin and increased adiponectin levels) [188]. It also influences adipokine expression and attenuates NF-κB and TNF-α expression in the adipose tissue and liver in HFD + STZ rats [189]. Adipocytes pretreated with resveratrol demonstrated an anti-inflammatory action, as evidenced by limiting the release of TNF-α and IL-6 and the activation of inflammatory proteins, such as extracellular receptor–activated kinase (ERK) and NF-κB [190]. Furthermore, inflammatory-related adipokines such as leptin, IL-6, IL-8, and MCP-1 were all downregulated by resveratrol in human adipose tissues [191, 192].

9.1.2.2. Impact on Metabolism and Energy Expenditure

Rutin has the potential to ameliorate obesity by promoting the formation of brown-like adipocytes in subcutaneous adipose tissues and by raising brown adipose tissue (BAT) activity. It also inhibits the liver's ability to produce fatty acids by repressing Srebp1c and Cd36 transcription, while enhancing the expression of PPAR gamma coactivator 1 (Pgc1) and deiodinase iodothyronine type 2 (Dio2), two genes involved in energy expenditure, in BAT [193]. Both genetically obese (Db/Db) and diet-induced obese mice showed significant reductions in adiposity, increased energy expenditure, and improved glucose homeostasis after rutin treatment. In obesity mouse models, rutin-induced brown adipocyte (beige) formation [193]. Isoquercitrin prevented weight gain and mesenteric adipose tissue fat deposition. In addition, it stimulated lipogenesis while suppressing lipolysis in white adipose tissues and liver [194]. It also inhibited the formation of cellular lipid droplets by activating AMP kinase, a crucial cellular energy sensor, and its dependent signaling pathways. These pathways improved the expression levels of adiponectin receptor 1 while reducing the activity of sterol regulatory element-binding transcription factor 1 (SREBP-1) and the expression of lipogenic genes/FAS signals [194, 195]. Hyperin possesses the ability to prevent the Swiss 3T3 cell line, preadipocyte clone 1 (3T3-L1) preadipocytes from maturing into fully developed adipocytes. By decreasing PPAR, CCAAT/enhancer-binding proteins (C/EBP), and SREBP-1c gene expression, as well as that of lipoprotein lipase (LPL), hyperin also decreased the intracellular accumulation of TG in mature adipocytes. Even at modest doses, it has showed to effectively prevent adipogenesis [196]. Through cyclin-dependent kinase 6 (CDK6) and transcription factor EB (TFEB), (CDK6-TFEB) signaling, hyperin at 80 mg/kg BW has shown to resist obesity by stimulating the uncoupling protein 1 (UCP1)-dependent transition of white to beige fat, aiding in lipophagy, a particular form of autophagy that helps in breaking down lipid droplets, and enhancing lipolysis [197]. Resveratrol promotes the expression of SIRT1, a key target of cellular energy metabolism and mitochondrial homeostasis, resulting in a dose-dependent reduction in fat formation within cells. It can also lower the expression of adipogenesis-related genes and proteins, including PPAR, C/EBP, and SREBP-1c [198, 199]. The lipolytic action of human and rat adipocytes was boosted by resveratrol through β-adrenergic stimulation and the subsequent rise in cyclic adenosine monophosphate levels [200, 201]. However, by blocking the effects of insulin and insulin-like lipogenic agents, resveratrol can slow down the transfer of glucose into the adipocytes' fat stores [202].

9.1.2.3. Fat Storage and Lipid Metabolism

Rutin treatment significantly decreased lipid droplet development in 3T3-L1 cells, promoted the expression of thermogenic markers, and lowered the expression of adipogenic genes [203]. Through inhibiting Wnt/catenin signaling, isoquercitrin restrains the 3T3-L1 cells from differentiating into adipocytes [204]. The antiobesity effects of resveratrol hint at the nonexpression of adipogenesis or lipogenesis genes such as PPAR-γ, ACC1, and FAS in visceral adipose tissues. Resveratrol has also showed to suppress the expression of LPL and stearoyl-CoA desaturase 1 fatty acid biosynthesis genes in the liver. However, as resveratrol regulates the gut microbiota, it may enhance the fasting-induced adipose factor (FIAF) gene's expression through the FIAF signaling pathway in the intestine [205].

9.1.3. Effects on Dyslipidemia

9.1.3.1. Lipid and Lipoprotein Modulation

Rutin supplementation was found to successfully lower the serum lipid levels in HFD and STZ-elicited diabetic rats and in hypertriacylglyceremic rats fed a high-fat and high-fructose diet including TC, TG, and low-density lipoprotein cholesterol (LDL) while achieving a considerable boost in HDL [206208]. Isoquercitrin has also showed to reduce the plasma concentrations of C-peptide, TG, and TC at a dose of 200 mg/kg BW in KK-Ay mice, a model of spontaneous diabetes [209]. Additionally, plasma TG and TC levels were markedly lowered in obese mice produced by HFD after receiving isoquercitrin at 5 mg/kg BW [210]. Furthermore, TC, TG, LDL-C, VLDL-C, free fatty acids, and phospholipid levels were reduced following supplementation, whereas HDL-C levels increased [211, 212]. Administration of hyperin (200 mg/kg BW) in diabetic mice models produced by HFD or alloxan or STZ resulted in a significant reduction in dyslipidemia markers, including TG, TC, and LDL-C levels, whereas HDL-C levels were elevated [161, 163]. It has showed that hyperin significantly enhanced TG metabolism and reduced TG in insulin-resistant HepG2 cells at a modest concentration of 0.75 μg/mL [170]. Resveratrol significantly and dose-dependently alleviated the blood TC, LDL-C, TG levels, and the arteriosclerosis index in male Sprague–Dawley rats fed a hyperlipidemic diet for 4 weeks. Although HDL-C levels tended to rise, the hepatic concentrations of TC and TG were lowered [213]. Similar results were observed in hyperlipidemic mice treated for 6 weeks [214] and in obese rats fed for 8 weeks [215]. In individuals with dyslipidemia and type 2 diabetes, oral supplementation with resveratrol (100–250 mg/d) has showed to effectively improve glycemic control and can lower the HbA1c, TC, and TG concentrations [216].

9.1.3.2. Regulation of Enzyme Activities

In diabetic rat models, rutin improved the LPL and lecithin cholesterol acyltransferase activities in plasma while showing a decreasing trend in (HMG Co-A) reductase activity [208]. Isoquercitrin treatment (40 mg/kg BW) considerably raised the levels of fat-metabolizing enzymes, such as LPL and lecithin cholesterol acyltransferase, while significantly lowering HMG Co-A levels [211, 212]. Hyperin showed 58.9% inhibition of HMG Co-A reductase [217]. Resveratrol has showed to downregulate the liver enzyme HMG-CoA and impact the expression of cholesterol metabolism-related enzymes [218, 219], with a simultaneous increase in the production and release of bile acids due to improvement of hepatic cholesterol 7a-hydroxylase expression [220].

9.1.3.3. Impact on Gene Expression and Fatty Acid Metabolism

According to Park et al., rutin supplementation at 1000 mg/kg BW enhanced the fecal sterol excretion, thereby lessening dietary cholesterol absorption [221]. Due to its ability to control the expression of genes (in particular the cyclooxygenase 1.3 (cox-1.3), S-trimethylglycine dehydrogenase-3 (stdh-3), and fatty acid transporter 7 (fat-7)), and by extending the production of the related unsaturated fatty acid, rutin diminished fat accumulation in Caenorhabditis elegans [222]. Isoquercitrin is attributed to the effective control of mRNA expression of AMPK-α and ACC [211, 212]. Additionally, via an AMPK-dependent mechanism, resveratrol boosts the LDL receptor expression on hepatocytes and hepatic uptake of LDL [223, 224].

9.1.4. Effects on Hypertension

9.1.4.1. Inhibition of Angiotensin-Converting Enzyme (ACE)

Rutin supplementation has been reported to improve hypertension in rats fed a high-carbohydrate HFD as well as in Nω-nitro-L-arginine methyl ester (L-NAME)-induced hypertensive rats [225, 226]. Additionally, normotensive rats showed improvements at 1–3 mg/kg BW [227]. The antihypertensive impact of rutin is exerted through its capacity to block the ACE, with an inhibitory potency that reached 87% at 151.12 μg/mL with an IC50 value of 19.34 μg/mL [225, 228]. Groups of normotensive and spontaneously hypertensive rats (SHR) receiving intravenous doses of isoquercitrin (0.5–4 mg/kg BW) exhibited dose-dependent hypotension that relied on the inhibition of ACE activity (55% at 4 mg/kg BW), leading to an inhibition in angiotensin II (Ang-II) production in the serum [229]. ACE showed to be significantly inhibited by hyperin, with an IC50 value of 54.40 μg/mL [230].

9.1.4.2. Promotion of Diuresis

Gasparotto Junior et al. explored the mechanisms underlying the hypotensive impact of isoquercitrin in SHR. They observed that the diuretic effect of isoquercitrin was dose-dependent (5–10 mg/kg BW in SHR) without any toxic symptoms. They reported that this effect is connected to its diuretic activity, by increasing the excretion of Na+ while leaving K+ levels unchanged in the urine [231]. In another study, the same authors discovered that the mechanism by which isoquercitrin (10 mg/kg; p.o.) increases diuresis is mostly connected to a higher bioavailability of bradykinin, prostaglandin I2, and nitric oxide, as well as the inhibitory effect on Na+/K+-ATPase [232]. The treatment with hyperin (6 mg/kg BW) lowered the high blood pressure in L-NAME-induced hypertensive rats. It has proposed that it was achieved by enhancing the diuretic and renal nitric oxide synthase (NOS) activities with a simultaneous reduction in the peripheral resistance through a decrease in the medial layer thickness of coronary vessels [233].

9.1.4.3. Activation of the Nitric Oxide–Guanylyl Cyclase Pathway

In isolated rat aorta rings, rutin exhibited an endothelium-dependent vasorelaxant effect in a dose-dependent manner via the nitric oxide–guanylyl cyclase pathway, a prostaglandin-mediated mechanism through the activation of ATP-sensitive potassium channels [234]. Resistance arteries in the rat mesenteric vascular bed respond to isoquercitrin at concentrations of 64.44, 139.32, and 464.4 μg/mL by inducing both endothelium-dependent and endothelium-independent vasodilation, with a peak relaxation of 57.8% observed at 464.4 μg/mL. This relaxation was triggered by nonselective KCa (calcium-activated) channel activation, nitric oxide, and the production of other endothelium mediators mainly at high levels of isoquercitrin [235]. In isolated rat aorta, hyperin demonstrated endothelium-dependent vasodilatory action with an EC50 of 91.3 μg/mL. This effect may be due to the promotion of endogenous hydrogen sulfide in arterial endotheliocytes and the opening of KCa channels [236, 237]. The possible mechanisms of resveratrol's antihypertensive action have also been demonstrated, including endothelial NO generation dependent on calcium-eNOS activation [238]. In addition, the improved bioavailability of NO was correlated with vasodilatory activities brought about by resveratrol in SHRs and Ang-II mice [239]. The endothelial NO bioactivity was boosted by resveratrol via the stimulation of eNOS gene transcription and stability [240]. The suppression of elevated Giα protein levels and the upstream signaling molecules promoting Gi protein overexpression in vascular smooth muscle cells are thought to be responsible for the attenuation of hypertension by resveratrol [241].

In addition to these effects, in high-salt-induced hypertensive mice, hyperin caused a drop in blood pressure by suppressing chymase, an independent Ang-II-forming enzyme [242]. In a clinical investigation, distal segments of internal thoracic arteries from patients undergoing coronary revascularization were treated with isoquercitrin at 28 μg/mL. This treatment produced a potent vasorelaxant effect, reaching 53.29%, that was mediated by activating the COX pathway [243]. In human umbilical vein endothelial cells under high glucose stress, the proapoptotic proteins p53, Bax, and cleaved caspase-3 expression was considerably downregulated by isoquercitrin treatment, whereas the antiapoptotic protein B-cell lymphoma 2 was raised in a dose-dependent manner [244]. Rutin also reestablished vascular reactivity and baroreflex sensitivity in hypertensive rats [245]. Furthermore, under high glucose conditions, rutin restored the acetylcholine-mediated endothelium function in aortic tissues by substantially recovering NO generation and suppressing the expression of nicotinamide adenine dinucleotide phosphate oxidase (NADPH) 4 and nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing three inflammasome pathways, both in vitro and in vivo [246]. A clinical trial carried out by Sattanathan et al. [247] involving patients with type 2 diabetes mellitus showed that rutin decreased both systolic and diastolic blood pressure.

One possible explanation could be that rutin, isoquercitrin, hyperin, and resveratrol could cooperate synergistically with each other, leading to better insulin sensitivity, less oxidative stress, and/or better lipid metabolism. As a result of their complementary antioxidant, anti-inflammatory, and vasodilatory properties, they may modulate important enzymes of glucose and lipid metabolism and NO synthesis more effectively, resulting in a greater therapeutic benefit for metabolic syndrome and cardiovascular risk factor management.

9.2. The Biosynthetic Pathways

Rutin, isoquercitrin, hyperin, and resveratrol share several common steps in their biosynthetic pathways, primarily originating in the phenylpropanoid pathway, starting from phenylalanine. Phenylalanine is first converted to trans-cinnamic acid via the action of phenylalanine ammonia-lyase. This intermediate is then hydroxylated by cinnamate-4-hydroxylase to generate p-coumaric acid, which is, in turn, activated by 4-coumarate-CoA ligase to yield 4-coumaroyl-CoA. The chalcone synthase enzyme catalyzes the condensation of 4-coumaroyl-CoA into chalcone, which is then converted to naringenin by chalcone isomerase. Flavanone-3-hydroxylase catalyzes the hydroxylation of the common precursor naringenin to dihydrokaempferol, which can then be converted into quercetin via flavonol synthase. Quercetin is glycosylated in the biosynthesis of rutin and isoquercitrin: Quercetin is rhamnosylated and glucosylated in rutin, while quercetin is glycosylated at the 3-O position in isoquercitrin by UDP-glucose: flavonoid 3-O-glucosyltransferase (UGT) [248, 249]. In turn, hyperin is an O-glycoside of quercetin-bearing a galactopyranoside moiety, formed by the action of sugar transferases, acting through with specificity in favor of galactose [250]. Conversely, resveratrol is produced via a branch of the phenylpropanoid pathway, where stilbene synthase catalyzes the condensation between p-coumaroyl-CoA and three malonyl-CoA molecules leading to the formation of resveratrol [251].

9.3. Pharmacokinetics: Bioavailability, Metabolism, and Absorption

Rutin has been studied for its bioavailability and metabolism. The research indicates that rutin supplementation can elevate plasma flavonoids without any adverse effects on liver function [252]. Rutin is metabolized by the gut microbiota and absorbed as conjugated metabolites, making it bioavailable [253]. In rats, rutin is absorbed more slowly than its aglycone, quercetin, due to cecal microflora hydrolysis [254]. Studies have identified quercetin sulfates and glucuronides as the major metabolites of both rutin and quercetin in rats, influencing their bioavailability [255]. In rabbits, the metabolic fate of rutin and quercetin has been linked to the urinary excretion of its degradation products, including 3,4‐dihydroxyphenylacetic acid [256]. Several studies have therefore explored the methods to improve the bioavailability of rutin. These studies collectively suggest promising strategies to improve its dissolution rate, permeability, and bioavailability, such as the use of nanoemulsions [257], encapsulation [258], and chitosan nanoparticles [259]. For example, complexation with 2-hydroxypropyl-β-cyclodextrin increased rutin's solubility, dissolution rate, and oral bioavailability [260].

The absolute bioavailability of hyperin is approximately 26%, indicating a moderate absorption rate in the body [261]. Hyperin has showed to undergo rapid metabolism in rats, leading to the formation of 33 metabolites identified in the plasma, urine, and feces, with quercetin and dihydroquercetin being the most prominent ones [262]. Studies suggest that hyperin exhibits bimodal absorption after oral administration in rats, possibly owing to hepatoenteric circulation or absorption by different parts of the intestine [263]. Methylation, sulfation, and glucuronidation are the major metabolic pathways of hyperin in vivo [264]. Human intestinal bacteria can further metabolize hyperin, producing metabolites such as benzoic acid, quercetin, and 3,4-dihydroxyphenylacetic acid [265]. Pharmacokinetic studies recommend its intraperitoneal or intravenous administration for improved efficacy due to low oral bioavailability [263].

Isoquercitrin has been found to have higher bioavailability than quercetin [266]. Pure isoquercitrin can be obtained on a large scale through enzymatic rutin hydrolysis with α-l-rhamnosidase. Although small amounts of intact isoquercitrin can be detected in plasma and tissues after oral administration, it is extensively metabolized in the intestine and liver [267]. Its metabolites were identified as quercetin, acetylated isoquercitrin, dehydroxylated isoquercitrin, hydroxylated quercetin, and hydroxymethylated quercetin [268]. Enzymatically modified isoquercitrin is more easily absorbed compared to other quercetin glycosides or aglycones after oral administration in rats. This enhanced absorption contributes to its bioavailability and therapeutic efficacy [269]. Isoquercitrin undergoes biotransformation in the intestine and liver, involving deglycosylation followed by forming conjugated and methylated derivatives of quercetin or degradation to phenolic acids and carbon compounds [267]. The bioavailability of isoquercitrin can be further enhanced using dietary isoquercitrin-γ-cyclodextrin molecular inclusion complex, as has been demonstrated in both rats and humans [270].

The oral bioavailability of resveratrol is almost zero because of its rapid and extensive metabolism, leading to the formation of various metabolites [271]. The research on absorption, metabolism, and bioavailability of resveratrol has been conducted using different models such as in vitro, ex vivo, and in vivo studies. These studies have showed that resveratrol is absorbed and metabolized in the body. Approximately, 75% of this polyphenol is excreted via various pathways after ingestion [272]. Resveratrol metabolism involves the formation of glucuronide and sulfate conjugates during absorption, which decreases the levels of free trans-resveratrol circulating in the body [273]. Additionally, in the rat small intestine, resveratrol is absorbed and metabolized through its conversion to resveratrol glucuronide [274]. Despite its low bioavailability, resveratrol is well-tolerated and has no marked toxicity in humans [275]. The bioavailability of resveratrol can be improved by its dimethyl ether analog, pterostilbene, which has greater bioavailability as shown by increased total plasma levels [276]. Other methods, such as nanoencapsulation and coadministration with piperine, have also showed promise for enhancing resveratrol's bioavailability [277].

9.4. Toxicity and Side Effects

Rutin, isoquercitrin, hyperin, and resveratrol are generally considered safe in lower doses but may have a different safety profile depending on dosage and exposure duration. Rutin is relatively nontoxic at lower doses, as demonstrated by acute toxicity studies in mice that yielded an LD50 of approximately 1510 mg/kg in male animals and 1490 mg/kg in female animals [278]. In rats, the administration of dietary concentrations up to 1% (542.4 mg/kg/day for males and 674 mg/kg/day for females) over 52 weeks did not produce any adverse effects [278]. On the other hand, large doses of rutin were shown to reduce erythrocytic parameters and attenuate weight gain in males [279]. Isoquercitrin is safe in humans, with an acceptable daily intake of 5.4 mg/kg/day, although higher doses induce benign chromaturia in rats [279]. Another similar compound with higher bioavailability, the enzymatically modified isoquercitrin, is also considered a safe food additive [280]. In rats, the no-observed adverse effect level is known to be 300 mg/kg/day, while doses of resveratrol higher than 3000 mg/kg/day will induce renal toxicity and other adverse effects [281]. In humans, up to 1000 mg/day is well tolerated, but gastrointestinal symptoms have been reported when the dose exceeds 2500–5000 mg [282]. Likewise, hyperin at high dosage (1000 mg/kg/day) can cause renal toxicity that results from its accumulation in the kidneys, but this damage can be reversed after stopping it [283]. In contrast, chronic toxicity studies with lower doses of hyperin (30–175 mg/kg/day) in rats reported no significant adverse effects on behavior, food intake, or weight gain [284].

10. Conclusion and Perspectives

In conclusion, Z. lotus contains bioactive compounds such as rutin, isoquercitrin, hyperin, and resveratrol, which exhibit remarkable potential in alleviating metabolic syndrome and its associated conditions—obesity, diabetes, hypertension, and dyslipidemia. The therapeutic effects are mediated through several critical mechanisms. Z. lotus acts as a regulatory agent for obesity by regulating lipid metabolism through the activation of PPARs and promoting lipid oxidation through AMPK activation. It enhances insulin sensitivity by upregulation GLUT4 expression and decreases postprandial hyperglycemia by suppressing α-glucosidase activity in diabetes. In hypertension, it restores endothelial function through the decrease of oxidative stress and an increase in NO bioavailability, consequently causing vasodilation. In terms of dyslipidemia, it reduces TC and TG levels through the inhibition of important enzymes such as HMG-CoA reductase as well as the modification of lipid metabolism pathways.

Moreover, the plant's antioxidant properties counteract reactive oxygen species, and its anti-inflammatory properties inhibit proinflammatory cytokines, which play a pivotal role in the prevention of the development of cardiovascular and metabolic complications. This holistic targeting of various pathways associated with metabolic syndrome and its associated diseases positions Z. lotus as a potential source of novel therapeutic agents that may have clinical relevance. These results are consistent with the traditional uses of Z. lotus in regions of the Middle East and Africa as a remedy for managing symptoms of diabetes, hypertension, and metabolic disorders, substantiating the therapeutic power of this plant.

However, while these traditional uses form a vast base, their effectiveness and safety need further experimental and clinical studies to validate them. Although toxicity studies report an overall good safety profile for Z. lotus, these data are predominantly focused on acute toxicity tested in animal models, making the long-term safety and human relevance uncertain. Furthermore, this also underlines the ongoing key limitation of a dearth of adequately powered clinical trials, yielding limited data in terms of standardized dosages, formulations, and more varied patient cohorts. The challenges still lie in discovering more effective strategies to improve bioavailability. These gaps need to be overcome in future research, which should ideally include well-designed in vivo “proof-of-concept” experiments, elucidating pharmacological effects, pharmacokinetics, pharmacodynamics, and any potential interactions in a system biology context. For dosages and formulations to be equivalent, standardization is the key; this will lead to consistent efficacy and safety, allowing for clinical effects to be optimized across patient populations. This review has raised several important issues that should be discussed in the future to promote their application and development.

Acknowledgments

We express our utmost gratitude to the CNRST, the Ministry of Higher Education, the National Agency of Aromatic and Medicinal Plants of Taounate, and the Presidency of the Mohammed First University (Project 2VPMA 2020/5) for their financial support (Project 2VPMA 2020/5). We also thank the Faculty of Sciences of Oujda for the logistical and financial support. At last, we thank Pr. Elachouri Mostafa from the biology department (Faculty of Sciences of Oujda) for allowing us to use three photos of Z. lotus that he took in the region of Abbou Lakhal near Figuig City (Morocco).

Nomenclature

ACE:

Angiotensin-converting enzyme

AMPK:

AMP-activated protein kinase

BAT:

Brown adipose tissue

GLUT4:

Glucose transporter 4

HDL:

High-density lipoprotein

HFD:

High-fat diet

HMG Co-A:

Hydroxy-methylglutaryl-CoA reductase

IC50:

Half-maximal inhibitory concentration

IFNγ:

Interferon gamma

LDL:

Low-density lipoprotein

NOS:

Nitric oxide synthase

SIRT1:

Activated sirtuin 1

PPARs:

Peroxisome proliferator–activated receptors

SREBP-1:

Sterol regulatory element-binding transcription Factor 1

TC:

Total cholesterol

TG:

Triglycerides

TNF-α:

Tumor necrosis factor

WAT:

White adipose tissue

STZ:

Streptozotocin

FBG:

Fasting blood glucose

HbA1c:

Glycated hemoglobin

PPARγ:

Peroxisome proliferator–activated receptor γ

IR:

Insulin receptor

PKB:

Protein kinase B

HepG2:

Human liver cancer cell line

G-6-Pase:

Glucose-6-phosphatase

NF-κB:

Nuclear factor

FAS:

Fatty acid synthase

C/EBP:

CCAAT/enhancer-binding proteins

ACC1:

Acetyl-CoA carboxylase 1

FIAF:

Fasting-induced adipose factor

L-NAME:

Nω-nitro-L-arginine methyl ester

SHR:

Spontaneously hypertensive rats

Ang-II:

Angiotensin II

3T3-L1:

Swiss 3T3 cell line, preadipocyte Clone 1

LD50:

The median lethal dose

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Author Contributions

C.A. did the literature search and wrote the review. A.A. and A.Z. revised and edited the contents, improved for English language, restructured, and completed the article with other data from the literature, in particular, with photos and medicinal uses. A.M., Y.S., N.B., and A.L. contributed to the literature search. C.A., A.A., and A.Z. have contributed equally to this work.

Funding

This research was supported by the CNRST, the Ministry of Higher Education, the National Agency of Aromatic and Medicinal Plants of Taounate, and the Presidency of the Mohammed First University (Project 2VPMA 2020/5).

References

  • 1.Saklayen M. G. The Global Epidemic of the Metabolic Syndrome. Current Hypertension Reports . 2018;20(2):p. 12. doi: 10.1007/s11906-018-0812-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Petersen M. C., Shulman G. I. Mechanisms of Insulin Action and Insulin Resistance. Physiological Reviews . 2018;98(4):2133–2223. doi: 10.1152/physrev.00063.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hurza V., Vatashchuk M., Bayliak M. M. Pathogenesis and Biomarkers of Metabolic Syndrome. Journal of Vasyl Stefanyk Precarpathian National University . 2022;8(4):7–19. doi: 10.15330/jpnu.8.4.7-19. [DOI] [Google Scholar]
  • 4.Incalza M. A., D’Oria R., Natalicchio A., Perrini S., Laviola L., Giorgino F. Oxidative Stress and Reactive Oxygen Species in Endothelial Dysfunction Associated With Cardiovascular and Metabolic Diseases. Vascular Pharmacology . 2018;100:1–19. doi: 10.1016/j.vph.2017.05.005. [DOI] [PubMed] [Google Scholar]
  • 5.Martemucci G., Khalil M., Di Luca A., Abdallah H., D’Alessandro A. G. Comprehensive Strategies for Metabolic Syndrome: How Nutrition, Dietary Polyphenols, Physical Activity, and Lifestyle Modifications Address Diabesity, Cardiovascular Diseases, and Neurodegenerative Conditions. Metabolites . 2024;14(6):p. 327. doi: 10.3390/metabo14060327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Shenoy A., Buttar H. S., Dicholkar P., Kaur G., Chintamaneni M. Functional Foods and Nutraceuticals in Metabolic and Non-Communicable Diseases . Elsevier; 2022. Role of Nutraceuticals, Functional Foods, and Spices in the Management of Metabolic Syndrome and Related Disorders. [Google Scholar]
  • 7.Obika P., Beamon J., Ali S., et al. Herbal Medicines for the Treatment of Metabolic Syndrome, Herbal Medicines . Elsevier; 2022. [Google Scholar]
  • 8.Wang S., Du Q., Meng X., Zhang Y. Natural Polyphenols: A Potential Prevention and Treatment Strategy for Metabolic Syndrome. Food & Function . 2022;13(19):9734–9753. doi: 10.1039/d2fo01552h. [DOI] [PubMed] [Google Scholar]
  • 9.Alipour A., Baradaran Rahimi V., Askari V. R. Promising Influences of Gingerols Against Metabolic Syndrome: A Mechanistic Review. BioFactors . 2022;48(5):993–1004. doi: 10.1002/biof.1892. [DOI] [PubMed] [Google Scholar]
  • 10.Barkaoui M., Katiri A., Boubaker H., Msanda F. Ethnobotanical Survey of Medicinal Plants Used in the Traditional Treatment of Diabetes in Chtouka Ait Baha and Tiznit (Western Anti-Atlas), Morocco. Journal of Ethnopharmacology . 2017;198:338–350. doi: 10.1016/j.jep.2017.01.023. [DOI] [PubMed] [Google Scholar]
  • 11.Idm’hand E., Msanda F., Cherifi K. Ethnobotanical Study and Biodiversity of Medicinal Plants Used in the Tarfaya Province, Morocco. Acta Ecologica Sinica . 2020;40(2):134–144. [Google Scholar]
  • 12.Bencheikh N., Radi F. Z., Fakchich J., et al. Ethnobotanical, Phytochemical, Toxicological, and Pharmacological Properties of Ziziphus lotus (L.) Lam.: A Comprehensive Review. Pharmaceuticals . 2023;16(4):p. 575. doi: 10.3390/ph16040575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Benslama A., Harrar A., Gul F., Demirtas I. Phenolic Compounds, Antioxidant and Antibacterial Activities of Zizyphus lotus L. Leaves Extracts. The Natural Products Journal . 2017;7(4):316–322. doi: 10.2174/2210315507666170530090957. [DOI] [Google Scholar]
  • 14.Marmouzi I., Kharbach M., El Jemli M., et al. Antidiabetic, Dermatoprotective, Antioxidant and Chemical Functionalities in Zizyphus lotus Leaves and Fruits. Industrial Crops and Products . 2019;132:134–139. doi: 10.1016/j.indcrop.2019.02.007. [DOI] [Google Scholar]
  • 15.Najjaa H., Abdelkarim B. A., Doria E., et al. Phenolic Composition of Some Tunisian Medicinal Plants Associated With Anti-Proliferative Effect on Human Breast Cancer MCF-7 Cells. The EuroBiotech Journal . 2020;4(2):104–112. doi: 10.2478/ebtj-2020-0012. [DOI] [Google Scholar]
  • 16.Rufino A. T., Costa V. M., Carvalho F., Fernandes E. Flavonoids as Antiobesity Agents: A Review. Medicinal Research Reviews . 2021;41(1):556–585. doi: 10.1002/med.21740. [DOI] [PubMed] [Google Scholar]
  • 17.Neri-Numa I. A., Cazarin C. B. B., Ruiz A. L. T. G., Paulino B. N., Molina G., Pastore G. M. Targeting Flavonoids on Modulation of Metabolic Syndrome. Journal of Functional Foods . 2020;73:p. 104132. doi: 10.1016/j.jff.2020.104132. [DOI] [Google Scholar]
  • 18.Masoumvand M., Ramezani E., Rahimi V. B., Askari V. R. Promising Influences of Moringa oleifera in Functional Foods Against Metabolic Syndrome: A Comprehensive and Mechanistic Review. Endocrine, Metabolic & Immune Disorders-Drug Targets . 2024;24(12):1355–1370. doi: 10.2174/0118715303269893231207071440. [DOI] [PubMed] [Google Scholar]
  • 19.Amara M., Benabdeli K. A Geobotanical and Phenological Study of Zizyphus lotus in the Naama Region (South-Western of Algeria) Journal of Applied Environmental and Biological Sciences . 2017;7(11):1–8. [Google Scholar]
  • 20.Ghedira K. Zizyphus lotus (L.) Desf.(Rhamnaceae): Wild Jujube. Phytothérapie . 2013;11(3):149–153. doi: 10.1007/s10298-013-0776-8. [DOI] [Google Scholar]
  • 21.Quézel P., Santa S. Volume II . Paris, France: CNRS Editions; 1962. New Flora of Algeria and the Southern Desert Regions. [Google Scholar]
  • 22.Tardío J., Sánchez-Mata M. D. C., Morales R., et al. Ethnobotanical and Food Composition Monographs of Selected Mediterranean Wild Edible Plants. In: Sánchez-Mata M. D. C., Tardío J., editors. Mediterranean Wild Edible Plants: Ethnobotany and Food Composition Tables . New York: Springer; 2016. pp. 273–470. [Google Scholar]
  • 23.Nasri-Ayachi M. B., Nabli M. A. Floral Biology Study of Ziziphus lotus L . Leuven, Belgium: International Society for Horticultural Science (ISHS); 2009. [Google Scholar]
  • 24.Quézel P., Santa S. Volume II . Paris, France: CNRS Editions; 1963. New Flora of Algeria and the Southern Desert Regions. [Google Scholar]
  • 25.Abdoul-Azize S. Potential Benefits of Jujube (Zizyphus lotus L.) Bioactive Compounds for Nutrition and Health. Journal of Nutrition and Metabolism . 2016;2016:1–13. doi: 10.1155/2016/2867470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Boussaid M., Taïbi K., Ait Abderrahim L., Ennajah A. Genetic Diversity of Ziziphus lotus Natural Populations From Algeria Based on Fruit Morphological Markers. Arid Land Research and Management . 2018;32(2):184–197. doi: 10.1080/15324982.2018.1424742. [DOI] [Google Scholar]
  • 27.de Cortes Sánchez-Mata M., Tardío J. Mediterranean Wild Edible Plants: Ethnobotany and Food Composition Tables . Springer; 2016. [Google Scholar]
  • 28.Tengberg A., Chen D. A Comparative Analysis of Nebkhas in Central Tunisia and Northern Burkina Faso. Geomorphology . 1998;22(2):181–192. doi: 10.1016/s0169-555x(97)00068-8. [DOI] [Google Scholar]
  • 29.Rsaissi N., Kamili E., Bencharki B., Hillali L., Bouhache M. Antimicrobial Activity of Fruits Extracts of the Wild Jujube Ziziphus lotus (L.) Desf. International Journal of Scientific Engineering and Research . 2013;4(9):1521–1528. [Google Scholar]
  • 30.Bakhtaoui F.-Z., Lakmichi H., Megraud F., Chait A., A Gadhi C. E. Gastro-Protective, Anti-Helicobacter Pylori and, Antioxidant Properties of Moroccan Zizyphus lotus L. Journal of Applied Pharmaceutical Science . 2014;4(10):81–87. doi: 10.7324/japs.2014.401015. [DOI] [Google Scholar]
  • 31.Torres‐García M. T., Salinas‐Bonillo M. J., Gázquez‐Sánchez F., Fernández‐Cortés Á., Querejeta J. I., Cabello J. Squandering Water in Drylands: The Water‐Use Strategy of the Phreatophyte Ziziphus lotus in a Groundwater‐Dependent Ecosystem. American Journal of Botany . 2021;108(2):236–248. doi: 10.1002/ajb2.1606. [DOI] [PubMed] [Google Scholar]
  • 32.Adeli M., Samavati V. Studies on the Steady Shear Flow Behavior and Chemical Properties of Water-Soluble Polysaccharide From Ziziphus lotus Fruit. International Journal of Biological Macromolecules . 2015;72:580–587. doi: 10.1016/j.ijbiomac.2014.08.047. [DOI] [PubMed] [Google Scholar]
  • 33.Hmamouchi M. Pharmacology, Toxicology, Lexicons . 2nd 2002. Medicinal and Aromatic Plants of Morocco: Traditional Uses, Market, Biology, Ecology, Chemistry. [Google Scholar]
  • 34.Bellakhdar J. Traditional Moroccan Pharmacopoeia . IBIS press; 1997. [Google Scholar]
  • 35.Bellakhdar J. Traditional Medicine and West Saharan Toxicology: Contribution to the Study of Moroccan Pharmacopoeia . Rabat, Morocco: Éditions Techniques Nord-Africaines; 1978. [Google Scholar]
  • 36.Bencheikh N., Elbouzidi A., Kharchoufa L., et al. Inventory of Medicinal Plants Used Traditionally to Manage Kidney Diseases in North-Eastern Morocco: Ethnobotanical Fieldwork and Pharmacological Evidence. Plants . 2021;10(9):p. 1966. doi: 10.3390/plants10091966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Benkhnigue O., Akka F. B., Salhi S., Fadli M., Douira A., Zidane L. Catalogue of Medicinal Plants Used in the Treatment of Diabetes in the Al Haouz-Rhamna Region (Morocco) Journal of Animal and Plant Sciences . 2014;23(1):3539–3568. [Google Scholar]
  • 38.Chaachouay N., Douira A., Zidane L. COVID-19, Prevention and Treatment With Herbal Medicine in the Herbal Markets of Salé Prefecture, North-Western Morocco. European Journal of Integrative Medicine . 2021;42:p. 101285. doi: 10.1016/j.eujim.2021.101285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Daoudi A., Bammou M., Zarkani S., Slimani I., Ibijbijen J., Nassiri L. Étude Ethnobotanique de la Flore Médicinale Dans la Commune Rurale d’Aguelmouss Province de Khénifra (Maroc) Phytothérapie . 2016;14(4):220–228. doi: 10.1007/s10298-015-0953-z. [DOI] [Google Scholar]
  • 40.Eddouks M., Ajebli M., Hebi M. Ethnopharmacological Survey of Medicinal Plants Used in Daraa-Tafilalet Region (Province of Errachidia), Morocco. Journal of Ethnopharmacology . 2017;198:516–530. doi: 10.1016/j.jep.2016.12.017. [DOI] [PubMed] [Google Scholar]
  • 41.El Alami A., Farouk L., Chait A. Ethnobotanical Study of Spontaneous Medicinal Plants Growing on the Northern Slope of the Azilal Atlas (Morocco) Algerian Journal of Natural Products . 2016;4(2):271–282. [Google Scholar]
  • 42.El Amri J., El Badaoui K., Zair T., Bouharb H., Chakir S., Alaoui T. E. M. Ethnobotanical Study of Medicinal Plants in the Region El Hajeb (Central Morocco) Journal of Research in Biology . 2015;4(8):1568–1580. [Google Scholar]
  • 43.Fatiha B. A., Souad S., Ouafae B., Jamila D., Allal D., Lahcen Z. Ethnobotanical Study of Medicinal Plants Used in the Region of Middle Oum Rbia (Morocco) Plant Archives . 2019;19(2):2005–2017. [Google Scholar]
  • 44.Jamila F., Mostafa E. Ethnobotanical Survey of Medicinal Plants Used by People in Oriental Morocco to Manage Various Ailments. Journal of Ethnopharmacology . 2014;154(1):76–87. doi: 10.1016/j.jep.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 45.Ouhaddou H., Boubaker H., Msanda F., El Mousadik A. An Ethnobotanical Study of Medicinal Plants of the Agadir Ida Ou Tanane Province (Southwest Morocco) Journal of Applied Biosciences . 2015;84(1):7707–7722. doi: 10.4314/jab.v84i1.5. [DOI] [Google Scholar]
  • 46.Abouri M., El Mousadik A., Msanda F., Boubaker H., Saadi B., Cherifi K. An Ethnobotanical Survey of Medicinal Plants Used in the Tata Province, Morocco. International Journal of Medicinal Plants Research . 2012;1(7):99–123. [Google Scholar]
  • 47.Belhaj s., Zidane L. Ethnobotanical and Ethnopharmacological Study of Medicinal Plants Used for the Treatment of Diseases of the Digestive Tract in the High Atlas Central of Morocco (North Africa) Journal of Analytical Sciences and Applied Biotechnology . 2021;3(1):7–14. [Google Scholar]
  • 48.Bouayyadi L., El Hafian M., Zidane L. Étude Floristique et Ethnobotanique de la Flore Médicinale Dans la Région du Gharb, Maroc. Journal of Applied Biosciences . 2015;93(1):8770–8788. doi: 10.4314/jab.v93i1.10. [DOI] [Google Scholar]
  • 49.El Azzouzi F., Zidane L. La Flore Médicinale Traditionnelle de la Région de Béni- Mellal (Maroc) Journal of Applied Biosciences . 2015;91(1):8493–8502. doi: 10.4314/jab.v91i1.8. [DOI] [Google Scholar]
  • 50.El Hachlafi N., Chebat A., Soulaymani Bencheikh R., Fikri-Benbrahim K. Ethnopharmacological Study of Medicinal Plants Used for Chronic Diseases Treatment in Rabat-Sale-Kenitra Region (Morocco) Ethnobotany Research and Applications . 2020;20:1–23. doi: 10.32859/era.20.2.1-23. [DOI] [Google Scholar]
  • 51.El Hafian M., Benlandini N., Elyacoubi H., Zidane L., Rochdi A. Étude Floristique et Ethnobotanique Des Plantes Médicinales Utilisées au Niveau de la Préfecture d’Agadir-Ida-Outanane (Maroc) Journal of Applied Biosciences . 2014;81(1):7198–7213. doi: 10.4314/jab.v81i1.8. [DOI] [Google Scholar]
  • 52.El Khomsi M., Dandani Y., Chaachouay N., Hmouni D. Ethnobotanical Study of Plants Used for Medicinal, Cosmetic, and Food Purposes in the Region of Moulay Yacoub, Northeast of Morocco. Journal of Pharmacy & Pharmacognosy Research . 2022;10(1):13–29. doi: 10.56499/jppres21.1084_10.1.13. [DOI] [Google Scholar]
  • 53.El Mansouri L., Ennabili A., Bousta D. Socioeconomic Interest and Valorization of Medicinal Plants From the Rissani Oasis (SE of Morocco) Boletin Latinoamericano y del Caribe de Plantas Medicinales y Aromaticas . 2011;10(1):30–45. [Google Scholar]
  • 54.El-Hilaly J., Hmammouchi M., Lyoussi B. Ethnobotanical Studies and Economic Evaluation of Medicinal Plants in Taounate Province (Northern Morocco) Journal of Ethnopharmacology . 2003;86(2-3):149–158. doi: 10.1016/s0378-8741(03)00012-6. [DOI] [PubMed] [Google Scholar]
  • 55.Idm’hand E., Msanda F., Cherifi K. Ethnobotanical Study of Medicinal Plants Used in the Treatment of Urinary Lithiasis in the Tarfaya Province (Morocco) International Journal of Innovation and Applied Studies . 2019;26(3):711–719. [Google Scholar]
  • 56.Jouad H., Haloui M., Rhiouani H., El Hilaly J., Eddouks M. Ethnobotanical Survey of Medicinal Plants Used for the Treatment of Diabetes, Cardiac and Renal Diseases in the North Centre Region of Morocco (Fez–Boulemane) Journal of Ethnopharmacology . 2001;77(2-3):175–182. doi: 10.1016/s0378-8741(01)00289-6. [DOI] [PubMed] [Google Scholar]
  • 57.Lahsissene H., Kahouadji A., Hseini S. Catalogue of Medicinal Plants Used in the Zaër Region (Western Morocco), Lejeunia. Revue de Botanique . 2009 [Google Scholar]
  • 58.Mrabti H. N., Jaradat N., Kachmar M. R., et al. Integrative Herbal Treatments of Diabetes in Beni Mellal Region of Morocco. Journal of Integrative Medicine . 2019;17(2):93–99. doi: 10.1016/j.joim.2019.01.001. [DOI] [PubMed] [Google Scholar]
  • 59.Naceiri Mrabti H., Bouyahya A., Naceiri Mrabti N., Jaradat N., Doudach L., Faouzi M. E. A. Ethnobotanical Survey of Medicinal Plants Used by Traditional Healers to Treat Diabetes in the Taza Region of Morocco. Evidence-Based Complementary and Alternative Medicine . 2021;2021:p. 5515634. doi: 10.1155/2021/5515634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Najem M., Ibijbijen J., Nassiri L. Ethnobotanical Treatment of Respiratory Diseases in the Central Middle Atlas (Morocco): Qualitative and Quantitative Approach. European Journal of Integrative Medicine . 2021;46:p. 101358. doi: 10.1016/j.eujim.2021.101358. [DOI] [Google Scholar]
  • 61.Salhi S., Fadli M., Zidane L., Douira A. Floristic and Ethnobotanical Studies of Medicinal Plants in the City of Kénitra (Morocco) Lazaroa . 2010;31:133–146. [Google Scholar]
  • 62.Tahraoui A., El-Hilaly J., Israili Z. H., Lyoussi B. Ethnopharmacological Survey of Plants Used in the Traditional Treatment of Hypertension and Diabetes in South-Eastern Morocco (Errachidia Province) Journal of Ethnopharmacology . 2007;110(1):105–117. doi: 10.1016/j.jep.2006.09.011. [DOI] [PubMed] [Google Scholar]
  • 63.Ziyyat A., Legssyer A., Mekhfi H., Dassouli A., Serhrouchni M., Benjelloun W. Phytotherapy of Hypertension and Diabetes in Oriental Morocco. Journal of Ethnopharmacology . 1997;58(1):45–54. doi: 10.1016/s0378-8741(97)00077-9. [DOI] [PubMed] [Google Scholar]
  • 64.Katiri A., Barkaoui M., Msanda F., Boubaker H. Ethnobotanical Survey of Medicinal Plants Used for the Treatment of Diabetes in the Tizin’Test Region (Taroudant Province, Morocco) Journal of Pharmacognosy and Natural Products . 2017;3(1):2472–0992. doi: 10.4172/2472-0992.1000130. [DOI] [Google Scholar]
  • 65.Boufous H., Marhoume F., Chait A., Bagri A. Ethnopharmacological Survey of Medicinal Plants With Hallucinogenic Effect and Plants Used Against Pain, Inflammatory Diseases, Diabetes and Urinary Lithiasis in Zagora “Morocco”. Journal of Intercultural Ethnopharmacology . 2017;6(4):342–350. doi: 10.5455/jice.20170721062527. [DOI] [Google Scholar]
  • 66.Benali T., Khabbach A., Ennabili A., Hammani K. Ethnopharmacological Prospecting of Medicinal Plants From the Province of Guercif (NE of Morocco) Moroccan Journal of Biology . 2017;14:1–14. [Google Scholar]
  • 67.Ghourri M., Zidane L., Douira A. Phytotherapy and Urinary Infections (Pyelonephritis and Cystitis) in the Moroccan Sahara (Tan-Tan) Journal of Animal and Plant Sciences . 2014;20(3):3171–3193. [Google Scholar]
  • 68.Senouci F., Ababou A., Chouieb M. Ethnobotanical Survey of the Medicinal Plants Used in the Southern Mediterranean. Case Study: The Region of Bissa (Northeastern Dahra Mountains, Algeria) Pharmacognosy Journal . 2019;11(4):647–659. doi: 10.5530/pj.2019.11.103. [DOI] [Google Scholar]
  • 69.Jaadan H., Akodad M., Moumen A., et al. Ethnobotanical Survey of Medicinal Plants Growing in the Region of “Oulad Daoud Zkhanine” (Nador Province), in Northeastern Morocco. Ethnobotany Research and Applications . 2020;19:1–12. doi: 10.32859/era.19.39.1-12. [DOI] [Google Scholar]
  • 70.Merzouki A., Ed-derfoufi F., Molero Mesa J. Contribution to the Knowledge of Rifian Traditional Medicine. II: Folk Medicine in Ksar Lakbir District (NW Morocco) Fitoterapia . 2000;71(3):278–307. doi: 10.1016/s0367-326x(00)00139-8. [DOI] [PubMed] [Google Scholar]
  • 71.Youbi A. E. H. E., Ouahidi I., Mansouri L. E., Daoudi A., Bousta D. Ethnopharmacological Survey of Plants Used for Immunological Diseases in Four Regions of Morocco. European Journal of Medicinal Plants . 2016;13(1):1–24. doi: 10.9734/ejmp/2016/12946. [DOI] [Google Scholar]
  • 72.Es-Safi I., Mechchate H., Amaghnouje A., et al. Medicinal Plants Used to Treat Acute Digestive System Problems in the Region of Fez-Meknes in Morocco: An Ethnopharmacological Survey. Ethnobotany Research and Applications . 2020;20:1–14. doi: 10.32859/era.20.25.1-14. [DOI] [Google Scholar]
  • 73.Mechchate H., Es-safi I., Jawhari F. Z., Bari A., Grafov A., Bousta D. Ethnobotanical Survey About the Management of Diabetes With Medicinal Plants Used by Diabetic Patients in Region of FezMeknes, Morocco. Ethnobotany Research and Applications . 2020;19(12):1–28. doi: 10.32859/era.19.12.1-28. [DOI] [Google Scholar]
  • 74.Ouhaddou H. O. H., Alaoui A., Sezgin A. Ethnobotanical Survey of Medicinal Plants Used for Treating Diabetes in Agadir Ida Outanane Region, Southwestern Morocco. Arabian Journal of Medicinal and Aromatic Plants . 2020;6(2):72–86. [Google Scholar]
  • 75.HassaniM E., Douiri E., Bammi J., Zidane L., Badoc A., Douira A. Medicinal Plants of the Middle Moulouya (Northeastern Morocco) Ethnopharmacologia . 2013;50(39):39–53. [Google Scholar]
  • 76.Bendif H., Harir M., Yahiaoui M., et al. Ethnobotanical Survey of Herbal Remedies Traditionally Used in El Hammadia (Southern Region of the Province of Bordj Bou Arreridj, Algeria) Algerian Journal of Biosciences . 2021;2(1):6–15. doi: 10.57056/ajb.v2i1.34. [DOI] [Google Scholar]
  • 77.Miara M. D., Hammou M. A., Aoul S. H. Phytotherapy and Taxonomy of Spontaneous Medicinal Plants in the Tiaret Region (Algeria) Phytothérapie . 2013;11(4):206–218. doi: 10.1007/s10298-013-0789-3. [DOI] [Google Scholar]
  • 78.Taïbi K., Aït Abderrahim L., Boussaid M., et al. Unraveling the Ethnopharmacological Potential of Medicinal Plants Used in Algerian Traditional Medicine for Urinary Diseases. European Journal of Integrative Medicine . 2021;44:p. 101339. doi: 10.1016/j.eujim.2021.101339. [DOI] [Google Scholar]
  • 79.Taïbi K., Abderrahim L. A., Ferhat K., et al. Ethnopharmacological Study of Natural Products Used for Traditional Cancer Therapy in Algeria. Saudi Pharmaceutical Journal . 2020;28(11):1451–1465. doi: 10.1016/j.jsps.2020.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Amel B. Traditional Treatment of High Blood Pressure and Diabetes in Souk Ahras District. Journal of Pharmacognosy and Phytotherapy . 2013;5(1):12–20. [Google Scholar]
  • 81.Boudjelal A., Henchiri C., Sari M., et al. Herbalists and Wild Medicinal Plants in M’Sila (North Algeria): An Ethnopharmacology Survey. Journal of Ethnopharmacology . 2013;148(2):395–402. doi: 10.1016/j.jep.2013.03.082. [DOI] [PubMed] [Google Scholar]
  • 82.Bouhaous L., Miara M. D., Bendif H., Souilah N. Medicinal Plants Used by Patients to Fight Cancer in Northwestern Algeria. Bulletin du Cancer . 2022;109(3):296–306. doi: 10.1016/j.bulcan.2021.09.017. [DOI] [PubMed] [Google Scholar]
  • 83.Chermat S., Gharzouli R. Ethnobotanical Study of Medicinal Flora in the North East of Algeria-An Empirical Knowledge in Djebel Zdimm (Setif) Journal of Materials Science and Engineering . 2015;5(2):50–59. doi: 10.17265/2161-6213/2015.1-2.007. [DOI] [Google Scholar]
  • 84.Chohra D., Ferchichi L. Ethnobotanical Study of Belezma National Park (BNP) Plants in Batna: East of Algeria. Acta Scientifica Naturalis . 2019;6(2):40–54. doi: 10.2478/asn-2019-0017. [DOI] [Google Scholar]
  • 85.Mechaala S., Bouatrous Y., Adouane S. Traditional Knowledge and Diversity of Wild Medicinal Plants in El Kantara’s Area (Algerian Sahara Gate): An Ethnobotany Survey. Acta Ecologica Sinica . 2022;42(1):33–45. doi: 10.1016/j.chnaes.2021.01.007. [DOI] [Google Scholar]
  • 86.Sari M., Hendel N., Boudjelal A., Sarri D. Inventory of Medicinal Plants Used for Traditional Treatment of Eczema in the Region of Hodna (M’sila-Algeria) Global Journal of Research on Medicinal Plants & Indigenous Medicine . 2012;1(4):97–100. [Google Scholar]
  • 87.Sari M., Hendel N., Sarri D., Boudjelal A., Benkhaled A. Ethnobotanical Study of Medicinal Flora Used by the People of the Forest of El Haourane-M’Sila-(Algeria) Journal of Ecotourism . 2013;9(2):21–25. [Google Scholar]
  • 88.Sarri M., Boudjelal A., Hendel N., Sarri D., Benkhaled A. Flora and Ethnobotany of Medicinal Plants in the Southeast of the Capital of Hodna (Algeria) Arabian Journal of Medicinal and Aromatic Plants . 2015;1(1):24–30. [Google Scholar]
  • 89.Telli A., Esnault M.-A., Ould El Hadj Khelil A. An Ethnopharmacological Survey of Plants Used in Traditional Diabetes Treatment in South-Eastern Algeria (Ouargla Province) Journal of Arid Environments . 2016;127:82–92. doi: 10.1016/j.jaridenv.2015.11.005. [DOI] [Google Scholar]
  • 90.Benarba B. Medicinal Plants Used by Traditional Healers From South-West Algeria: An Ethnobotanical Study. Journal of Intercultural Ethnopharmacology . 2016;5(4):320–330. doi: 10.5455/jice.20160814115725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Benderradji L., Rebbas K., Ghadbane M., Bounar R., Brini F., Bouzerzour H. Ethnobotanical Study of Medicinal Plants in Djebel Messaad Region (M’sila, Algeria) Global Journal of Research on Medicinal Plants & Indigenous Medicine . 2014;3(12):445–459. [Google Scholar]
  • 92.Bendif H., Miara M., Harir M., et al. Ethnobotany of Medicinal Plants of El Mansourah (West of Bordj Bou Arreridj, Algeria) Journal of Soil and Plant Biology . 2018;1:24–39. [Google Scholar]
  • 93.Miara M. D., Bendif H., Rebbas K., Rabah B., Hammou M. A., Maggi F. Medicinal Plants and Their Traditional Uses in the Highland Region of Bordj Bou Arreridj (Northeast Algeria) Journal of Herbal Medicine . 2019;16:p. 100262. doi: 10.1016/j.hermed.2019.100262. [DOI] [Google Scholar]
  • 94.Zatout F., Benarba B., Bouazza A., Babali B., Bey N. N., Morsli A. Ethnobotanical Investigation on Medicinal Plants Used by Local Populations in Tlemcen National Park (Extreme North West Algeria) Mediterranean Botany . 2021;15(30):p. 12. [Google Scholar]
  • 95.Lakhdari W., Dehliz A., Acheuk F., et al. Ethnobotanical Study of Some Plants Used in Traditional Medicine in the Region of Oued Righ (Algerian Sahara) The Journal of Medicinal Plants Studies . 2016;4(2):204–211. [Google Scholar]
  • 96.Abubakar I. B., Ukwuani-Kwaja A. N., Garba A. D., et al. Ethnobotanical Study of Medicinal Plants Used for Cancer Treatment in Kebbi State, North-West Nigeria. Acta Ecologica Sinica . 2020;40(4):306–314. doi: 10.1016/j.chnaes.2020.02.007. [DOI] [Google Scholar]
  • 97.Yebouk C., Redouan F. Z., Benítez G., et al. Ethnobotanical Study of Medicinal Plants in the Adrar Province, Mauritania. Journal of Ethnopharmacology . 2020;246:112217–217. doi: 10.1016/j.jep.2019.112217. [DOI] [PubMed] [Google Scholar]
  • 98.Ngene J., Ngoule C., Kidik C. P., Ottou P. M., Dibong S., Mpondo E. M. Importance Dans la Pharmacopée Traditionnelle des Plantes à Flavonoïdes Vendues Dans les Marchés de Douala est (Cameroun) Journal of Applied Biosciences . 2015;88:8194–8210. [Google Scholar]
  • 99.Mahomoodally M. F., Muthoorah L. D. An Ethnopharmacological Survey of Natural Remedies Used by the Chinese Community in Mauritius. Asian Pacific Journal of Tropical Biomedicine . 2014;4:S387–S399. doi: 10.12980/apjtb.4.2014c775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Jaradat N. A., Ayesh O. I., Anderson C. Ethnopharmacological Survey About Medicinal Plants Utilized by Herbalists and Traditional Practitioner Healers for Treatments of Diarrhea in the West Bank/Palestine. Journal of Ethnopharmacology . 2016;182:57–66. doi: 10.1016/j.jep.2016.02.013. [DOI] [PubMed] [Google Scholar]
  • 101.Oran S., Al-Eisawi D. Ethnobotanical Survey of the Medicinal Plants in the Central Mountains (North-South) in Jordan. Journal of Biodiversity and Environmental Sciences . 2015;6(3):381–400. [Google Scholar]
  • 102.Aya K., M’hamed T. Chemical Compounds, Antioxidant Activity, and In Vitro and In Silico Litholytic Effects of Zizyphus lotus Extracts. Journal of Basic and Clinical Physiology and Pharmacology . 2020;31(3):p. 20190091. doi: 10.1515/jbcpp-2019-0091. [DOI] [PubMed] [Google Scholar]
  • 103.Maciuk A., Ghedira K., Thepenier P., Lavaud C., Zeches-Hanrot M. A New Flavonol Glycoside From Leaves of Zizyphus lotus. Die Pharmazie . 2003;58(2):158–159. doi: 10.1002/chin.200323205. [DOI] [PubMed] [Google Scholar]
  • 104.Ouldchikh S., TirTouil A., Meddah B. Zizyphus lotus (L.) Extracts as Prebiotics in the Aggregation and Adhesion of Probiotic and Inhibition of Pathologic Bacteria From Patients With Colorectal Cancer. International Journal of Food Studies . 2020;9(1):160–177. doi: 10.7455/ijfs/9.1.2020.a3. [DOI] [Google Scholar]
  • 105.Rached W., Barros L., Ziani B. E., et al. HPLC-DAD-ESI-MS/MS Screening of Phytochemical Compounds and the Bioactive Properties of Different Plant Parts of Zizyphus lotus (L.) Desf. Food & Function . 2019;10(9):5898–5909. doi: 10.1039/c9fo01423c. [DOI] [PubMed] [Google Scholar]
  • 106.Mkadmini Hammi K., Jellouli Ennigrou D., Majdoub H., Ksouri R. Recovery of Phenolic Compounds and Carbohydrates From Hydro-Ethanolic Extract of Zizyphus lotus Fruit Using Ultrafiltration Process. International Journal of Food Engineering . 2017;13(12):p. 20170343. doi: 10.1515/ijfe-2017-0343. [DOI] [Google Scholar]
  • 107.Yahia Y., Benabderrahim M. A., Tlili N., Bagues M., Nagaz K. Bioactive Compounds, Antioxidant and Antimicrobial Activities of Extracts From Different Plant Parts of Two Ziziphus Mill. Species. PLoS One . 2020;15(5):p. e0232599. doi: 10.1371/journal.pone.0232599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Benabderrahim M. A., Yahia Y., Bettaieb I., Elfalleh W., Nagaz K. Antioxidant Activity and Phenolic Profile of a Collection of Medicinal Plants From Tunisian Arid and Saharan Regions. Industrial Crops and Products . 2019;138:p. 111427. doi: 10.1016/j.indcrop.2019.05.076. [DOI] [Google Scholar]
  • 109.Tlili H., Hanen N., Ben Arfa A., et al. Biochemical Profile and In Vitro Biological Activities of Extracts From Seven Folk Medicinal Plants Growing Wild in Southern Tunisia. PLoS One . 2019;14(9):p. e0213049. doi: 10.1371/journal.pone.0213049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Berkani F., Dahmoune F., Serralheiro M. L., et al. New Bioactive Constituents Characterized by LC–MS/MS in Optimized Microwave Extract of Jujube Seeds (Zizyphus lotus L.) Journal of Food Measurement and Characterization . 2021;15(4):3216–3233. doi: 10.1007/s11694-021-00903-z. [DOI] [Google Scholar]
  • 111.Cadi H. E., Bouzidi H. E., Selama G., et al. Physico-Chemical and Phytochemical Characterization of Moroccan Wild Jujube “Zizyphus lotus (L.)” Fruit Crude Extract and Fractions. Molecules . 2020;25(22):p. 5237. doi: 10.3390/molecules25225237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Abcha I., Ben Haj Said L., Salmieri S., Criado P., Neffati M., Lacroix M. Optimization of Extraction Parameters, Characterization and Assessment of Bioactive Properties of Ziziphus lotus Fruit Pulp for Nutraceutical Potential. European Food Research and Technology . 2021;247(9):2193–2209. doi: 10.1007/s00217-021-03779-x. [DOI] [Google Scholar]
  • 113.Bekkar N. E. H., Meddah B., Cakmak Y. S., Keskin B. Phenolic Composition, Antioxidant and Antimicrobial Activities of Zizyphus lotus L. and Ruta chalepensis L. Growing in Mascara (Western Algeria) Journal of Microbiology, Biotechnology and Food Sciences . 2021;10(5):p. e3004. doi: 10.15414/jmbfs.3004. [DOI] [Google Scholar]
  • 114.Ghedira K., Chemli R., Caron C., Nuzilard J.-M., Zeches M., Le Men-Olivier L. Four Cyclopeptide Alkaloids From Zizyphus lotus. Phytochemistry . 1995;38(3):767–772. doi: 10.1016/0031-9422(94)00669-k. [DOI] [Google Scholar]
  • 115.Ghedira K., Chemli R., Richard B., Nuzillard J.-M., Zeches M., Men-Olivier L. L. Two Cyclopeptide Alkaloids From Zizyphus lotus. Phytochemistry . 1993;32(6):1591–1594. doi: 10.1016/0031-9422(93)85186-u. [DOI] [Google Scholar]
  • 116.Le Crouéour G., Thépenier P., Richard B., Petermann C., Ghédira K., Zèches-Hanrot M. Lotusine G: A New Cyclopeptide Alkaloid From Zizyphus lotus. Fitoterapia . 2002;73(1):63–68. doi: 10.1016/s0367-326x(01)00363-x. [DOI] [PubMed] [Google Scholar]
  • 117.Zarga M. A., Sabri S., Al-Aboudi A., Ajaz M. S., Sultana N., Atta-ur-Rahman New Cyclopeptide Alkaloids From Zizyphus lotus. Journal of Natural Products . 1995;58(4):504–511. doi: 10.1021/np50118a004. [DOI] [PubMed] [Google Scholar]
  • 118.Maciuk A., Lavaud C., Thépenier P., Jacquier M.-J., Ghédira K., Zèches-Hanrot M. Four New Dammarane Saponins From Zizyphus lotus. Journal of Natural Products . 2004;67(10):1639–1643. doi: 10.1021/np0499362. [DOI] [PubMed] [Google Scholar]
  • 119.Renault J.-H., Ghedira K., Thepenier P., Lavaud C., Zeches-Hanrot M., Le Men-Olivier L. Dammarane Saponins From Zizyphus lotus. Phytochemistry . 1997;44(7):1321–1327. doi: 10.1016/s0031-9422(96)00721-2. [DOI] [PubMed] [Google Scholar]
  • 120.Borgi W., Recio M. C., Ríos J. L., Chouchane N. Anti-Inflammatory and Analgesic Activities of Flavonoid and Saponin Fractions From Zizyphus lotus (L.) Lam. South African Journal of Botany . 2008;74(2):320–324. doi: 10.1016/j.sajb.2008.01.009. [DOI] [Google Scholar]
  • 121.Wang F.-X., Zhu N., Zhou F., Lin D.-X. Natural Aporphine Alkaloids With Potential to Impact Metabolic Syndrome. Molecules . 2021;26(20):p. 6117. doi: 10.3390/molecules26206117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cao S., Liu M., Han Y., et al. Effects of Saponins on Lipid Metabolism: The Gut–Liver Axis Plays a Key Role. Nutrients . 2024;16(10):p. 1514. doi: 10.3390/nu16101514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.El Maaiden E., El Kharrassi Y., Moustaid K., Essamadi A. K., Nasser B. Comparative Study of Phytochemical Profile between Ziziphus Spina Christi and Ziziphus lotus From Morocco. Journal of Food Measurement and Characterization . 2019;13(1):121–130. doi: 10.1007/s11694-018-9925-y. [DOI] [Google Scholar]
  • 124.Elaloui M., Ennajah A., Ghazghazi H., et al. Quantification of Total Phenols, Flavonoides and Tannins From Ziziphus jujuba (mill.) and Ziziphus lotus (L.)(Desf). Leaf Extracts and Their Effects on Antioxidant and Antibacterial Activities. International Journal of Secondary Metabolite . 2016;4(1):18–26. doi: 10.21448/ijsm.275886. [DOI] [Google Scholar]
  • 125.Ghazghazi H., Aouadhi C., Riahi L., Maaroufi A., Hasnaoui B. Fatty Acids Composition of Tunisian Ziziphus lotus L.(Desf.) Fruits and Variation in Biological Activities Between Leaf and Fruit Extracts. Natural Product Research . 2014;28(14):1106–1110. doi: 10.1080/14786419.2014.913244. [DOI] [PubMed] [Google Scholar]
  • 126.Abdeddaim M., Lombarkia O., Bacha A., et al. Biochemical Characterization and Nutritional Properties of Zizyphus lotus L. Fruits in Aures Region, Northeastern of Algeria. Food Science and Technology . 2014;15:75–81. [Google Scholar]
  • 127.Benammar C., Hichami A., Yessoufou A., et al. Zizyphus lotus L.(Desf.) Modulates Antioxidant Activity and Human T-Cell Proliferation. BMC Complementary and Alternative Medicine . 2010;10(1):54–59. doi: 10.1186/1472-6882-10-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chouaibi M., Mahfoudhi N., Rezig L., Donsì F., Ferrari G., Hamdi S. Nutritional Composition of Zizyphus lotus L. Seeds. Journal of the Science of Food and Agriculture . 2012;92(6):1171–1177. doi: 10.1002/jsfa.4659. [DOI] [PubMed] [Google Scholar]
  • 129.Makhdar N., Anouar A., Bouyazza L. Composition in Fatty Acids, Sterols and Tocopherols of Vegetable Oil Extract From Kernels of Zizyphus lotus L. Journal of Materials and Environmental Science . 2019;10(11):1074–1082. [Google Scholar]
  • 130.Ourzeddine W., Fadel H., Mechehoud Y., et al. Chemical Composition and Antioxidant Activity of the Fruit Essential Oil of Zizyphus lotus (L.) Desf.(Rhamnaceae) International Journal of Pharmacognosy and Phytochemical Research . 2017;9:228–232. [Google Scholar]
  • 131.Snoussi A., Koubaier H. B. H., Chouaibi M., Bouzouita N. Comparative Study of the Fatty Acids and Tocopherol Profiles, Physical Properties, and Antioxidant Activities of Zizyphus lotus L. Seed Oils Based on the Geographical Origin. Chemistry Africa . 2021;5(1):69–78. doi: 10.1007/s42250-021-00294-0. [DOI] [Google Scholar]
  • 132.Boudraa S., Hambaba L., Zidani S., Boudraa H. Composition minérale et vitaminique des fruits de cinq espèces sous exploitées en Algérie: Celtis australis L., Crataegus azarolus L., Crataegus monogyna Jacq., Elaeagnus angustifolia L.et Zizyphus lotus L. Fruits . 2010;65(2):75–84. doi: 10.1051/fruits/20010003. [DOI] [Google Scholar]
  • 133.Bekkar N. E. H., Meddah B., Keskin B., Sonnet P. Oral Acute Toxicity, Influence on the Gastrointestinal Microbiota and In Vivo Anti-Salmonellosis Effect of Zizyphus lotus (l.) and Ruta chalepensis (L.) Essential Oils. Journal of Applied Biotechnology Reports . 2021;8(1):13–26. [Google Scholar]
  • 134.Letaief T., Garzoli S., Ovidi E., et al. Organ Dependency Variation of the Chemical Composition of Ziziphus lotus Volatile Fractions. European Journal of Biological Research . 2021;11(4):501–508. [Google Scholar]
  • 135.Kharchoufa L., Bouhrim M., Bencheikh N., et al. Potential Toxicity of Medicinal Plants Inventoried in Northeastern Morocco: An Ethnobotanical Approach. Plants . 2021;10(6):p. 1108. doi: 10.3390/plants10061108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Borgi W., Ghedira K., Chouchane N. Antiinflammatory and Analgesic Activities of Zizyphus lotus Root Barks. Fitoterapia . 2007;78(1):16–19. doi: 10.1016/j.fitote.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 137.Bencheikh N., Bouhrim M., Kharchoufa L., Choukri M., Bnouham M., Elachouri M. Protective Effect of Zizyphus lotus. L. (Desf.) Fruit Against CCl4-Induced Acute Liver Injury in Rat. Evidence-Based Complement Alternative Medicine . 2019;2019:p. 6161593. doi: 10.1155/2019/6161593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.El Hachimi F., Alfaiz C., Bendriss A., Cherrah Y., Alaoui K. Anti-Inflammatory Activity of the Seed Oil of Zizyphus lotus (L.) Desf. Phytothérapie . 2017;15(3):147–154. doi: 10.1007/s10298-016-1056-1. [DOI] [Google Scholar]
  • 139.Hani A. F., Zaouani M., Mimoune N., et al. Evaluation of Anti-Inflammatory and Antidiarrhoeal Activity of Leaf Aqueous Extracts of Zizyphus Lotus (L) in Albino Wistar Rats. Bulletin of University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca-Veterinary Medicine . 2020;77(1):53–60. doi: 10.15835/buasvmcn-vm:2019.0041. [DOI] [Google Scholar]
  • 140.Huang P. L. A Comprehensive Definition for Metabolic Syndrome. Disease Models & Mechanisms . 2009;2(5-6):231–237. doi: 10.1242/dmm.001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kaur J. Assessment and Screening of the Risk Factors in Metabolic Syndrome. Medical Science . 2014;2(3):140–152. doi: 10.3390/medsci2030140. [DOI] [Google Scholar]
  • 142.Bencheikh N., Bouhrim M., Kharchoufa L., et al. The Nephroprotective Effect of Zizyphus lotus L.(desf.) Fruits in a Gentamicin-Induced Acute Kidney Injury Model in Rats: A Biochemical and Histopathological Investigation. Molecules . 2021;26(16):p. 4806. doi: 10.3390/molecules26164806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Bencheikh N., Bouhrim M., Merrouni I. A., et al. Antihyperlipidemic and Antioxidant Activities of Flavonoid-Rich Extract of Ziziphus lotus (L.) Lam. Fruits. Applied Sciences . 2021;11(17):p. 7788. doi: 10.3390/app11177788. [DOI] [Google Scholar]
  • 144.Li Y.-W., Kao T.-W., Chang P.-K., Chen W.-L., Wu L.-W. Atherogenic Index of Plasma as Predictors for Metabolic Syndrome, Hypertension and Diabetes Mellitus in Taiwan Citizens: A 9-Year Longitudinal Study. Scientific Reports . 2021;11(1):p. 9900. doi: 10.1038/s41598-021-89307-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ghalem M., Murtaza B., Belarbi M., Akhtar Khan N., Hichami A. Antiinflammatory and Antioxidant Activities of a Polyphenol‐Rich Extract From Zizyphus lotus L Fruit Pulp Play a Protective Role Against Obesity. Journal of Food Biochemistry . 2018;42(6):p. e12689. doi: 10.1111/jfbc.12689. [DOI] [Google Scholar]
  • 146.Berrichi M., Benammar C., Murtaza B., Hichami A., Belarbi M., Khan N. A. Zizyphus lotus L. Fruit Attenuates Obesity-Associated Alterations: In Vivo Mechanisms. Archives of Physiology and Biochemistry . 2021;127(2):119–126. doi: 10.1080/13813455.2019.1621349. [DOI] [PubMed] [Google Scholar]
  • 147.Rodriguez-Ramiro I., Vauzour D., Minihane A. Polyphenols and Non-Alcoholic Fatty Liver Disease: Impact and Mechanisms. Proceedings of the Nutrition Society . 2016;75(1):47–60. doi: 10.1017/s0029665115004218. [DOI] [PubMed] [Google Scholar]
  • 148.Benammar C., Baghdad C., Belarbi M., Subramaniam S., Hichami A., Khan N. A. Antidiabetic and Antioxidant Activities of Zizyphus lotus L Aqueous Extracts in Wistar Rats. Journal of Nutrition & Food Sciences . 2014;8(4):1–6. [Google Scholar]
  • 149.Noce A., Di Lauro M., Di Daniele F., et al. Natural Bioactive Compounds Useful in Clinical Management of Metabolic Syndrome. Nutrients . 2021;13(2):p. 630. doi: 10.3390/nu13020630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lu Y., Bao T., Mo J., Ni J., Chen W. Research Advances in Bioactive Components and Health Benefits of Jujube (Ziziphus jujuba Mill.) Fruit. Journal of Zhejiang University-Science B . 2021;22(6):431–449. doi: 10.1631/jzus.b2000594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hasan S. M. K., Kabir M. R., Kabir M. R., Islam M. R., Akhter M. J., Moury J. Y. Proximate Composition, Minerals, Phytochemicals, and Functional Activities of Jujube Fruits Grown in Bangladesh. Journal of Agriculture and Food Research . 2022;8:p. 100302. doi: 10.1016/j.jafr.2022.100302. [DOI] [Google Scholar]
  • 152.Savova M. S., Vasileva L. V., Mladenova S. G., et al. Ziziphus jujuba Mill. Leaf Extract Restrains Adipogenesis by Targeting PI3K/AKT Signaling Pathway. Biomedicine & Pharmacotherapy . 2021;141:p. 111934. doi: 10.1016/j.biopha.2021.111934. [DOI] [PubMed] [Google Scholar]
  • 153.Alam A., Subhan N., Rahman M., Uddin S. J., Reza H. M., Sarker S. D. Effect of Citrus Flavonoids, Naringin and Naringenin, on Metabolic Syndrome and Their Mechanisms of Action. Advances in Nutrition . 2014;5(4):404–417. doi: 10.3945/an.113.005603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Arichi H., Kimura Y., Okuda H., Baba K., Kozawa M., Arichi S. Effects of Stilbene Components of the Roots of Polygonum Cuspidatum Sieb. et Zucc. on Lipid Metabolism. Chemical & Pharmaceutical Bulletin . 1982;30(5):1766–1770. doi: 10.1248/cpb.30.1766. [DOI] [PubMed] [Google Scholar]
  • 155.Gouveia H. J. C. B., Urquiza-Martínez M. V., Manhães-de-Castro R., et al. Effects of the Treatment With Flavonoids on Metabolic Syndrome Components in Humans: A Systematic Review Focusing on Mechanisms of Action. International Journal of Molecular Sciences . 2022;23(15):p. 8344. doi: 10.3390/ijms23158344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Niture N. T., Ansari A. A., Naik S. R. Anti-Hyperglycemic Activity of Rutin in Streptozotocin-Induced Diabetic Rats: An Effect Mediated Through Cytokines, Antioxidants and Lipid Biomarkers. Indian Journal of Experimental Biology . 2014;52(7):720–727. [PubMed] [Google Scholar]
  • 157.Prince P. S. M., Kamalakkannan N. Rutin Improves Glucose Homeostasis in Streptozotocin Diabetic Tissues by Altering Glycolytic and Gluconeogenic Enzymes. Journal of Biochemical and Molecular Toxicology . 2006;20(2):96–102. doi: 10.1002/jbt.20117. [DOI] [PubMed] [Google Scholar]
  • 158.Sun C., Wang L., Sun J., Wang Z., Tang Z. Hypoglycemic and Hypolipidemic Effects of Rutin on Hyperglycemic Rats. Journal of Traditional Chinese Medicine . 2020;40(4):640–645. doi: 10.19852/j.cnki.jtcm.2020.04.012. [DOI] [PubMed] [Google Scholar]
  • 159.Zhang L., Zhang S.-T., Yin Y.-C., Xing S., Li W.-N., Fu X.-Q. Hypoglycemic Effect and Mechanism of Isoquercitrin as an Inhibitor of Dipeptidyl Peptidase-4 in Type 2 Diabetic Mice. RSC Advances . 2018;8(27):14967–14974. doi: 10.1039/c8ra00675j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Jayachandran M., Zhang T., Ganesan K., Xu B., Chung S. S. M. Isoquercetin Ameliorates Hyperglycemia and Regulates Key Enzymes of Glucose Metabolism via Insulin Signaling Pathway in Streptozotocin-Induced Diabetic Rats. European Journal of Pharmacology . 2018;829:112–120. doi: 10.1016/j.ejphar.2018.04.015. [DOI] [PubMed] [Google Scholar]
  • 161.Zhang Y., Wang M., Dong H., Yu X., Zhang J. Anti-Hypoglycemic and Hepatocyte-Protective Effects of Hyperoside from Zanthoxylum Bungeanum Leaves in Mice With High-Carbohydrate/High-Fat Diet and Alloxan-Induced Diabetes. International Journal of Molecular Medicine . 2018;41(1):77–86. doi: 10.3892/ijmm.2017.3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zhang Y., Yu X., Wang M., et al. Hyperoside From Z. Bungeanum Leaves Restores Insulin Secretion and Mitochondrial Function by Regulating Pancreatic Cellular Redox Status in Diabetic Mice. Free Radical Biology and Medicine . 2021;162:412–422. doi: 10.1016/j.freeradbiomed.2020.10.320. [DOI] [PubMed] [Google Scholar]
  • 163.Verma N., Amresh G., Sahu P. K., Mishra N., Rao C. V., Singh A. P. Pharmacological Evaluation of Hyperin for Antihyperglycemic Activity and Effect on Lipid Profile in Diabetic Rats. Indian Journal of Experimental Biology . 2013;51(1):65–72. [PubMed] [Google Scholar]
  • 164.Rehman K., Saeed K., Munawar S. M., Akash M. S. H. Resveratrol Regulates Hyperglycemia-Induced Modulations in Experimental Diabetic Animal Model. Biomedicine & Pharmacotherapy . 2018;102:140–146. doi: 10.1016/j.biopha.2018.03.050. [DOI] [PubMed] [Google Scholar]
  • 165.Hsu C. Y., Shih H. Y., Chia Y. C., et al. Rutin Potentiates Insulin Receptor Kinase to Enhance Insulin‐Dependent Glucose Transporter 4 Translocation. Molecular Nutrition & Food Research . 2014;58(6):1168–1176. doi: 10.1002/mnfr.201300691. [DOI] [PubMed] [Google Scholar]
  • 166.Ahmed O. M., Moneim A. A., Yazid I. A., Mahmoud A. M. Antihyperglycemic, Antihyperlipidemic and Antioxidant Effects and the Probable Mechanisms of Action of Ruta Graveolens Infusion and Rutin in Nicotinamide-Streptozotocin-Induced Diabetic Rats. Diabetologia Croatica . 2010;39(1):15–35. [Google Scholar]
  • 167.Cai E. P., Lin J.-K. Epigallocatechin Gallate (EGCG) and Rutin Suppress the Glucotoxicity Through Activating IRS2 and AMPK Signaling in Rat Pancreatic β Cells. Journal of Agricultural and Food Chemistry . 2009;57(20):9817–9827. doi: 10.1021/jf902618v. [DOI] [PubMed] [Google Scholar]
  • 168.Lee C.-C., Hsu W.-H., Shen S.-R., Cheng Y.-H., Wu S.-C. Fagopyrum Tataricum (Buckwheat) Improved High-Glucose-Induced Insulin Resistance in Mouse Hepatocytes and Diabetes in Fructose-Rich Diet-Induced Mice. Experimental Diabetes Research . 2012;2012:1–10. doi: 10.1155/2012/375673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Jiang H., Horiuchi Y., Hironao K.-y., Kitakaze T., Yamashita Y., Ashida H. Prevention Effect of Quercetin and Its Glycosides on Obesity and Hyperglycemia Through Activating AMPKα in High-Fat Diet-Fed ICR Mice. Journal of Clinical Biochemistry & Nutrition . 2020;67(1):75–83. doi: 10.3164/jcbn.20-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Duan J.-Y., Chen W., Zhao Y.-Q., et al. Flavonoids From Hypericum Patulum Enhance Glucose Consumption and Attenuate Lipid Accumulation in HepG2 Cells. Journal of Food Biochemistry . 2021;45(9):p. e13898. doi: 10.1111/jfbc.13898. [DOI] [PubMed] [Google Scholar]
  • 171.Brasnyó P., Molnár G. A., Mohás M., et al. Resveratrol Improves Insulin Sensitivity, Reduces Oxidative Stress and Activates the Akt Pathway in Type 2 Diabetic Patients. British Journal of Nutrition . 2011;106(3):383–389. doi: 10.1017/s0007114511000316. [DOI] [PubMed] [Google Scholar]
  • 172.Liu K., Zhou R., Wang B., Mi M.-T. Effect of Resveratrol on Glucose Control and Insulin Sensitivity: A Meta-Analysis of 11 Randomized Controlled Trials. The American Journal of Clinical Nutrition . 2014;99(6):1510–1519. doi: 10.3945/ajcn.113.082024. [DOI] [PubMed] [Google Scholar]
  • 173.Timmers S., Konings E., Bilet L., et al. Calorie Restriction-Like Effects of 30 Days of Resveratrol Supplementation on Energy Metabolism and Metabolic Profile in Obese Humans. Cell Metabolism . 2011;14(5):612–622. doi: 10.1016/j.cmet.2011.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Goh K. P., Lee H. Y., Lau D. P., Supaat W., Chan Y. H., Koh A. F. Y. Effects of Resveratrol in Patients With Type 2 Diabetes Mellitus on Skeletal Muscle SIRT1 Expression and Energy Expenditure. International Journal of Sport Nutrition and Exercise Metabolism . 2014;24(1):2–13. doi: 10.1123/ijsnem.2013-0045. [DOI] [PubMed] [Google Scholar]
  • 175.Chi T.-C., Chen W.-P., Chi T.-L., et al. Phosphatidylinositol-3-Kinase is Involved in the Antihyperglycemic Effect Induced by Resveratrol in Streptozotocin-Induced Diabetic Rats. Life Sciences . 2007;80(18):1713–1720. doi: 10.1016/j.lfs.2007.02.002. [DOI] [PubMed] [Google Scholar]
  • 176.Oboh G., Ademosun A. O., Ayeni P. O., Omojokun O. S., Bello F. Comparative Effect of Quercetin and Rutin on α-Amylase, α-Glucosidase, and Some Pro-Oxidant-Induced Lipid Peroxidation in Rat Pancreas. Comparative Clinical Pathology . 2015;24(5):1103–1110. doi: 10.1007/s00580-014-2040-5. [DOI] [Google Scholar]
  • 177.Li Y. Q., Zhou F. C., Gao F., Bian J. S., Shan F. Comparative Evaluation of Quercetin, Isoquercetin and Rutin as Inhibitors of α-Glucosidase. Journal of Agricultural and Food Chemistry . 2009;57(24):11463–11468. doi: 10.1021/jf903083h. [DOI] [PubMed] [Google Scholar]
  • 178.Shen H., Wang J., Ao J., et al. Structure-Activity Relationships and the Underlying Mechanism of α-Amylase Inhibition by Hyperoside and Quercetin: Multi-Spectroscopy and Molecular Docking Analyses. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy . 2023;285:p. 121797. doi: 10.1016/j.saa.2022.121797. [DOI] [PubMed] [Google Scholar]
  • 179.Zhang A. J., Rimando A. M., Mizuno C. S., Mathews S. T. α-Glucosidase Inhibitory Effect of Resveratrol and Piceatannol. The Journal of Nutritional Biochemistry . 2017;47:86–93. doi: 10.1016/j.jnutbio.2017.05.008. [DOI] [PubMed] [Google Scholar]
  • 180.Barthel A., Schmoll D. Novel Concepts in Insulin Regulation of Hepatic Gluconeogenesis. American Journal of Physiology-Endocrinology and Metabolism . 2003;285(4):E685–E692. doi: 10.1152/ajpendo.00253.2003. [DOI] [PubMed] [Google Scholar]
  • 181.Ku S.-K., Kwak S., Kwon O.-J., Bae J.-S. Hyperoside Inhibits High-Glucose-Induced Vascular Inflammation In Vitro and In Vivo. Inflammation . 2014;37(5):1389–1400. doi: 10.1007/s10753-014-9863-8. [DOI] [PubMed] [Google Scholar]
  • 182.Palsamy P., Sivakumar S., Subramanian S. Resveratrol Attenuates Hyperglycemia-Mediated Oxidative Stress, Proinflammatory Cytokines and Protects Hepatocytes Ultrastructure in Streptozotocin–Nicotinamide-Induced Experimental Diabetic Rats. Chemico-Biological Interactions . 2010;186(2):200–210. doi: 10.1016/j.cbi.2010.03.028. [DOI] [PubMed] [Google Scholar]
  • 183.Soufi F. G., Vardyani M., Sheervalilou R., Mohammadi M., Somi M. H. Long-Term Treatment With Resveratrol Attenuates Oxidative Stress Pro-Inflammatory Mediators and Apoptosis in Streptozotocin-Nicotinamide-Induced Diabetic Rats. General Physiology and Biophysics . 2012;31(04):431–438. doi: 10.4149/gpb_2012_039. [DOI] [PubMed] [Google Scholar]
  • 184.Ku C. R., Lee H. J., Kim S. K., Lee E. Y., Lee M.-K., Lee E. J. Resveratrol Prevents Streptozotocin-Induced Diabetes by Inhibiting the Apoptosis of Pancreatic β-Cell and the Cleavage of Poly (ADP-Ribose) Polymerase. Endocrine Journal . 2012;59(2):103–109. doi: 10.1507/endocrj.ej11-0194. [DOI] [PubMed] [Google Scholar]
  • 185.Gao M., Ma Y., Liu D. Rutin Suppresses Palmitic Acids-Triggered Inflammation in Macrophages and Blocks High Fat Diet-Induced Obesity and Fatty Liver in Mice. Pharmaceutical Research . 2013;30(11):2940–2950. doi: 10.1007/s11095-013-1125-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Muvhulawa N., Dludla P. V., Ziqubu K., et al. Rutin Ameliorates Inflammation and Improves Metabolic Function: A Comprehensive Analysis of Scientific Literature. Pharmacological Research . 2022;178:p. 106163. doi: 10.1016/j.phrs.2022.106163. [DOI] [PubMed] [Google Scholar]
  • 187.Muvhulawa N., Dludla P. V., Mthembu S. X. H., Ziqubu K., Tiano L., Mazibuko-Mbeje S. E. Rutin Attenuates Tumor Necrosis Factor-α-Induced Inflammation and Initiates Fat Browning in 3T3-L1 Adipocytes: Potential Therapeutic Implications for Anti-Obesity Therapy. South African Journal of Botany . 2023;160:697–704. doi: 10.1016/j.sajb.2023.07.043. [DOI] [Google Scholar]
  • 188.Kim J. H., Lee S., Cho E. J. The Protective Effects of Acer Okamotoanum and Isoquercitrin on Obesity and Amyloidosis in a Mouse Model. Nutrients . 2020;12(5):p. 1353. doi: 10.3390/nu12051353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Abdelhameed R. F., Ibrahim A. K., Elfaky M. A., et al. Antioxidant and Anti-Inflammatory Activity of Cynanchum acutum L. Isolated Flavonoids Using Experimentally Induced Type 2 Diabetes Mellitus: Biological and In Silico Investigation for Nf-Κb Pathway/mir-146a Expression Modulation. Antioxidants . 2021;10(11):p. 1713. doi: 10.3390/antiox10111713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Kang L., Heng W., Yuan A., Baolin L., Fang H. Resveratrol Modulates Adipokine Expression and Improves Insulin Sensitivity in Adipocytes: Relative to Inhibition of Inflammatory Responses. Biochimie . 2010;92(7):789–796. doi: 10.1016/j.biochi.2010.02.024. [DOI] [PubMed] [Google Scholar]
  • 191.Cullberg K. B., Olholm J., Paulsen S. K., et al. Resveratrol has Inhibitory Effects on the Hypoxia-Induced Inflammation and Angiogenesis in Human Adipose Tissue In Vitro. European Journal of Pharmaceutical Sciences . 2013;49(2):251–257. doi: 10.1016/j.ejps.2013.02.014. [DOI] [PubMed] [Google Scholar]
  • 192.Zagotta I., Dimova E. Y., Debatin K.-M., Wabitsch M., Kietzmann T., Fischer-Posovszky P. Obesity and Inflammation: Reduced Cytokine Expression Due to Resveratrol in a Human In Vitro Model of Inflamed Adipose Tissue. Frontiers in Pharmacology . 2015;6:p. 79. doi: 10.3389/fphar.2015.00079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Yuan X., Wei G., You Y., et al. Rutin Ameliorates Obesity Through Brown Fat Activation. The FASEB Journal . 2017;31(1):333–345. doi: 10.1096/fj.201600459rr. [DOI] [PubMed] [Google Scholar]
  • 194.Zhou J., Yoshitomi H., Liu T., et al. Isoquercitrin Activates the AMP–Activated Protein Kinase (AMPK) Signal Pathway in Rat H4IIE Cells. BMC Complementary and Alternative Medicine . 2014;14(1):p. 42. doi: 10.1186/1472-6882-14-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kim S. H., Yun C., Kwon D., Lee Y.-H., Kwak J.-H., Jung Y.-S. Effect of Isoquercitrin on Free Fatty Acid-Induced Lipid Accumulation in HepG2 Cells. Molecules . 2023;28(3):p. 1476. doi: 10.3390/molecules28031476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Berkoz M. Effect of Hyperoside on the Inhibition of Adipogenesis in 3t3-L1 Adipocytes. Acta Endocrinologica . 2019;15(2):165–172. doi: 10.4183/aeb.2019.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Cheng S., Ni X., Yao Y., et al. Hyperoside Prevents High-Fat Diet-Induced Obesity by Increasing White Fat Browning and Lipophagy via CDK6-TFEB Pathway. Journal of Ethnopharmacology . 2023;307:p. 116259. doi: 10.1016/j.jep.2023.116259. [DOI] [PubMed] [Google Scholar]
  • 198.Chen S., Li Z., Li W., Shan Z., Zhu W. Resveratrol Inhibits Cell Differentiation in 3T3-L1 Adipocytes via Activation of AMPK. Canadian Journal of Physiology and Pharmacology . 2011;89(11):793–799. doi: 10.1139/y11-077. [DOI] [PubMed] [Google Scholar]
  • 199.Imamura H., Nagayama D., Ishihara N., et al. Resveratrol Attenuates Triglyceride Accumulation Associated With Upregulation of Sirt1 and Lipoprotein Lipase in 3T3-L1 Adipocytes. Molecular Genetics and Metabolism Reports . 2017;12:44–50. doi: 10.1016/j.ymgmr.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Gomez-Zorita S., Tréguer K., Mercader J., Carpéné C. Resveratrol Directly Affects In Vitro Lipolysis and Glucose Transport in Human Fat Cells. Journal of Physiology & Biochemistry . 2013;69(3):585–593. doi: 10.1007/s13105-012-0229-0. [DOI] [PubMed] [Google Scholar]
  • 201.Szkudelska K., Nogowski L., Szkudelski T. Resveratrol, a Naturally Occurring Diphenolic Compound, Affects Lipogenesis, Lipolysis and the Antilipolytic Action of Insulin in Isolated Rat Adipocytes. The Journal of Steroid Biochemistry and Molecular Biology . 2009;113(1-2):17–24. doi: 10.1016/j.jsbmb.2008.11.001. [DOI] [PubMed] [Google Scholar]
  • 202.Carpéné C., Les F., Cásedas G., et al. Resveratrol Anti-Obesity Effects: Rapid Inhibition of Adipocyte Glucose Utilization. Antioxidants . 2019;8(3):p. 74. doi: 10.3390/antiox8030074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Ma B., Hao J., Xu H., et al. Rutin Promotes White Adipose Tissue “Browning” and Brown Adipose Tissue Activation Partially Through the Calmodulin-Dependent Protein Kinase Kinase β/AMP-Activated Protein Kinase Pathway. Endocrine Journal . 2022;69(4):385–397. doi: 10.1507/endocrj.ej21-0441. [DOI] [PubMed] [Google Scholar]
  • 204.Lee S.-H., Kim B., Oh M. J., et al. Persicaria hydropiper (L.) Spach and its Flavonoid Components, Isoquercitrin and Isorhamnetin, Activate the Wnt/β‐Catenin Pathway and Inhibit Adipocyte Differentiation of 3T3‐L1 Cells. Phytotherapy Research . 2011;25(11):1629–1635. doi: 10.1002/ptr.3469. [DOI] [PubMed] [Google Scholar]
  • 205.Qiao Y., Sun J., Xia S., Tang X., Shi Y., Le G. Effects of Resveratrol on Gut Microbiota and Fat Storage in a Mouse Model With High-Fat-Induced Obesity. Food & Function . 2014;5(6):1241–1249. doi: 10.1039/c3fo60630a. [DOI] [PubMed] [Google Scholar]
  • 206.Chen L., Shen T., Zhang C., et al. Quercetin and Isoquercitrin Inhibiting Hepatic Gluconeogenesis Through Lkb1-Ampkα Pathway. Acta Endocrinologica . 2020;16(1):p. 9. doi: 10.4183/aeb.2020.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Micháliková D., Tyukos Kaprinay B., Lipták B., et al. Natural Substance Rutin Versus Standard Drug Atorvastatin in a Treatment of Metabolic Syndrome-Like Condition. Saudi Pharmaceutical Journal . 2019;27(8):1196–1202. doi: 10.1016/j.jsps.2019.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Prince P. S. M., Kannan N. K. Protective Effect of Rutin on Lipids, Lipoproteins, Lipid Metabolizing Enzymes and Glycoproteins in Streptozotocin-Induced Diabetic Rats. Journal of Pharmacy and Pharmacology . 2006;58(10):1373–1383. doi: 10.1211/jpp.58.10.0011. [DOI] [PubMed] [Google Scholar]
  • 209.Zhang R., Yao Y., Wang Y., Ren G. Antidiabetic Activity of Isoquercetin in Diabetic KK-A Y Mice. Nutrition and Metabolism . 2011;8:85–86. doi: 10.1186/1743-7075-8-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Manzoor M., Muroi M., Ogawa N., et al. Isoquercitrin From Apocynum Venetum L. Produces an Anti-Obesity Effect on Obese Mice by Targeting C-1-Tetrahydrofolate Synthase, Carbonyl Reductase, and Glutathione S-Transferase P and Modification of the AMPK/SREBP-1c/FAS/CD36 Signaling Pathway in Mice In Vivo. Food & Function . 2022;13(21):10923–10936. doi: 10.1039/d2fo02438a. [DOI] [PubMed] [Google Scholar]
  • 211.Jayachandran M., Wu Z., Ganesan K., Khalid S., Chung S., Xu B. Isoquercetin Upregulates Antioxidant Genes, Suppresses Inflammatory Cytokines and Regulates AMPK Pathway in Streptozotocin-Induced Diabetic Rats. Chemico-Biological Interactions . 2019;303:62–69. doi: 10.1016/j.cbi.2019.02.017. [DOI] [PubMed] [Google Scholar]
  • 212.Jayachandran M., Zhang T., Wu Z., Liu Y., Xu B. Isoquercetin Regulates SREBP-1C via AMPK Pathway in Skeletal Muscle to Exert Antihyperlipidemic and Anti-Inflammatory Effects in STZ Induced Diabetic Rats. Molecular Biology Reports . 2020;47(1):593–602. doi: 10.1007/s11033-019-05166-y. [DOI] [PubMed] [Google Scholar]
  • 213.Zhu L., Luo X., Jin Z. Effect of Resveratrol on Serum and Liver Lipid Profile and Antioxidant Activity in Hyperlipidemia Rats. Asian-Australasian Journal of Animal Sciences . 2008;21(6):890–895. doi: 10.5713/ajas.2008.70638. [DOI] [Google Scholar]
  • 214.Xie H., Han H.-P., Chen Z., He J.-P. A Study on the Effect of Resveratrol on Lipid Metabolism in Hyperlipidemic Mice. African Journal of Traditional, Complementary and Alternative Medicines . 2013;11(1):209–212. doi: 10.4314/ajtcam.v11i1.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Rivera L., Morón R., Zarzuelo A., Galisteo M. Long-Term Resveratrol Administration Reduces Metabolic Disturbances and Lowers Blood Pressure in Obese Zucker Rats. Biochemical Pharmacology . 2009;77(6):1053–1063. doi: 10.1016/j.bcp.2008.11.027. [DOI] [PubMed] [Google Scholar]
  • 216.Bhatt J. K., Thomas S., Nanjan M. J. Resveratrol Supplementation Improves Glycemic Control in Type 2 Diabetes Mellitus. Nutrition Research . 2012;32(7):537–541. doi: 10.1016/j.nutres.2012.06.003. [DOI] [PubMed] [Google Scholar]
  • 217.Huang W., Ye X., Li X., et al. The Inhibition Activity of Chemical Constituents in Hawthorn Fruit and Their Synergistic Action to HMG-CoA Reductase. Zhongguo Zhongyao Zazhi . 2010;35(18):2428–2431. [PubMed] [Google Scholar]
  • 218.Do G.-M., Kwon E.-Y., Kim H.-J., et al. Long-Term Effects of Resveratrol Supplementation on Suppression of Atherogenic Lesion Formation and Cholesterol Synthesis in Apo E-Deficient Mice. Biochemical and Biophysical Research Communications . 2008;374(1):55–59. doi: 10.1016/j.bbrc.2008.06.113. [DOI] [PubMed] [Google Scholar]
  • 219.Penumathsa S. V., Thirunavukkarasu M., Koneru S., et al. Statin and Resveratrol in Combination Induces Cardioprotection Against Myocardial Infarction in Hypercholesterolemic Rat. Journal of Molecular and Cellular Cardiology . 2007;42(3):508–516. doi: 10.1016/j.yjmcc.2006.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Chen Q., Wang E., Ma L., Zhai P. Dietary Resveratrol Increases the Expression of Hepatic 7α-Hydroxylase and Ameliorates Hypercholesterolemia in High-Fat Fed C57BL/6J Mice. Lipids in Health and Disease . 2012;11(1):p. 56. doi: 10.1186/1476-511x-11-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Park S.-Y., Bok S.-H., Jeon S.-M., et al. Effect of Rutin and Tannic Acid Supplements on Cholesterol Metabolism in Rats1 1Abbreviations: HDL, High Density Lipoprotein; HMG-CoA, 3-Hydroxy-3-Methylglutaryl-CoA; ACAT, Acyl CoA:cholesterol Acyltransferase. Nutrition Research . 2002;22(3):283–295. doi: 10.1016/s0271-5317(01)00398-0. [DOI] [Google Scholar]
  • 222.Qin X., Wang W., Chu W. Antioxidant and Reducing Lipid Accumulation Effects of Rutin in Caenorhabditis elegans. BioFactors . 2021;47(4):686–693. doi: 10.1002/biof.1755. [DOI] [PubMed] [Google Scholar]
  • 223.Yashiro T., Nanmoku M., Shimizu M., Inoue J., Sato R. Resveratrol Increases the Expression and Activity of the Low Density Lipoprotein Receptor in Hepatocytes by the Proteolytic Activation of the Sterol Regulatory Element-Binding Proteins. Atherosclerosis . 2012;220(2):369–374. doi: 10.1016/j.atherosclerosis.2011.11.006. [DOI] [PubMed] [Google Scholar]
  • 224.Zhu X., Yang J., Zhu W., et al. Combination of Berberine With Resveratrol Improves the Lipid-Lowering Efficacy. International Journal of Molecular Sciences . 2018;19(12):p. 3903. doi: 10.3390/ijms19123903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Ganga R. M., Goud P. P., Nvl S. R. Antihypertensive Effect of Rutin: Pharmacological and Computational Approach. Asian Journal of Pharmaceutical and Clinical Research . 2019;12(8):87–92. doi: 10.22159/ajpcr.2019.v12i18.34118. [DOI] [Google Scholar]
  • 226.Panchal S. K., Poudyal H., Arumugam T. V., Brown L. Rutin Attenuates Metabolic Changes, Nonalcoholic Steatohepatitis, and Cardiovascular Remodeling in High-Carbohydrate, High-Fat Diet-Fed Rats. The Journal of Nutrition . 2011;141(6):1062–1069. doi: 10.3945/jn.111.137877. [DOI] [PubMed] [Google Scholar]
  • 227.da Rocha Lapa F., Soares K. C., Rattmann Y. D., et al. Vasorelaxant and Hypotensive Effects of the Extract and the Isolated Flavonoid Rutin Obtained From Polygala Paniculata L. Journal of Pharmacy and Pharmacology . 2011;63(6):875–881. doi: 10.1111/j.2042-7158.2010.01240.x. [DOI] [PubMed] [Google Scholar]
  • 228.Guerrero L., Castillo J., Quinones M., et al. Inhibition of Angiotensin-Converting Enzyme Activity by Flavonoids: Structure-Activity Relationship Studies. PLoS One . 2012;7(11):p. e49493. doi: 10.1371/journal.pone.0049493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Gasparotto Junior A., Gasparotto F. M., Lourenço E. L. B., et al. Antihypertensive Effects of Isoquercitrin and Extracts From Tropaeolum majus L.: Evidence for the Inhibition of Angiotensin Converting Enzyme. Journal of Ethnopharmacology . 2011;134(2):363–372. doi: 10.1016/j.jep.2010.12.026. [DOI] [PubMed] [Google Scholar]
  • 230.Balasuriya B. N., Rupasinghe H. V. Plant Flavonoids as Angiotensin Converting Enzyme Inhibitors in Regulation of Hypertension. Functional Foods in Health and Disease . 2011;1(5):172–188. [Google Scholar]
  • 231.Gasparotto Junior A., Gasparotto F. M., Boffo M. A., et al. Diuretic and Potassium-Sparing Effect of Isoquercitrin—An Active Flavonoid of Tropaeolum majus L. Journal of Ethnopharmacology . 2011;134(2):210–215. doi: 10.1016/j.jep.2010.12.009. [DOI] [PubMed] [Google Scholar]
  • 232.Gasparotto Junior A., Prando T. B. L., Leme T. D. S. V., et al. Mechanisms Underlying the Diuretic Effects of Tropaeolum majus L. Extracts and its Main Component Isoquercitrin. Journal of Ethnopharmacology . 2012;141(1):501–509. doi: 10.1016/j.jep.2012.03.018. [DOI] [PubMed] [Google Scholar]
  • 233.Koçyõldõz Z. Ç., Birman H., Olgaç V., Akgün-Dar K., Melikoğlu G., Meriçli A. H. Crataegus Tanacetifolia Leaf Extract Prevents L-NAME-Induced Hypertension in Rats: A Morphological Study. Phytotherapy Research . 2006;20(1):66–70. doi: 10.1002/ptr.1808. [DOI] [PubMed] [Google Scholar]
  • 234.Xia M.-l., Yao H., Wei E.-q., Zhou X.-m., Bruce I. C., Xia Q. Rutin-Induced Endothelium-Dependent Vasorelaxation in Rat Aortic Rings and the Underlying Mechanism. 2005 IEEE Engineering in Medicine and Biology 27th Annual Conference; 2006; IEEE; pp. 5595–5597. [DOI] [PubMed] [Google Scholar]
  • 235.Gasparotto Junior A., dos Reis Piornedo R., Assreuy J., Da Silva-Santos J. E. Nitric Oxide and Kir6.1 Potassium Channel Mediate Isoquercitrin-Induced Endothelium-Dependent and Independent Vasodilation in the Mesenteric Arterial Bed of Rats. European Journal of Pharmacology . 2016;788:328–334. doi: 10.1016/j.ejphar.2016.08.006. [DOI] [PubMed] [Google Scholar]
  • 236.Han J., Xuan J., Hu H., Chen Z. Effects and Mechanisms of Hyperoside on Vascular Endothelium Function in Middle Cerebral Arteries of Rats Ex Vivo. Zhongguo Zhongyao Zazhi . 2014;39(24):4849–4855. [PubMed] [Google Scholar]
  • 237.Ibarra-Alvarado C., Rojas A., Luna F., Rojas J. I., Rivero-Cruz B., Rivero-Cruz J. F. Vasorelaxant Constituents of the Leaves of Prunus Serotina Capulín. Revista Latinoamericana de Quimica . 2009;37(2):164–173. [Google Scholar]
  • 238.Li X., Dai Y., Yan S., et al. Resveratrol Lowers Blood Pressure in Spontaneously Hypertensive Rats via Calcium-Dependent Endothelial No Production. Clinical and Experimental Hypertension . 2016;38(3):287–293. doi: 10.3109/10641963.2015.1089882. [DOI] [PubMed] [Google Scholar]
  • 239.Dolinsky V. W., Chakrabarti S., Pereira T. J., et al. Resveratrol Prevents Hypertension and Cardiac Hypertrophy in Hypertensive Rats and Mice. Biochimica et Biophysica Acta-Molecular Basis of Disease . 2013;1832(10):1723–1733. doi: 10.1016/j.bbadis.2013.05.018. [DOI] [PubMed] [Google Scholar]
  • 240.DiNatale J., Crowe-White K. Effects of Resveratrol Supplementation on Nitric Oxide-Mediated Vascular Outcomes in Hypertension: A Systematic Review. Nitric Oxide . 2022;129:74–81. doi: 10.1016/j.niox.2022.10.005. [DOI] [PubMed] [Google Scholar]
  • 241.Sarkar O., Li Y., Anand-Srivastava M. B. Resveratrol Prevents the Development of High Blood Pressure in Spontaneously Hypertensive Rats Through the Inhibition of Enhanced Expression of Giα Proteins. Canadian Journal of Physiology and Pharmacology . 2019;97(9):872–879. doi: 10.1139/cjpp-2019-0040. [DOI] [PubMed] [Google Scholar]
  • 242.Devarajan S., Yahiro E., Uehara Y., et al. Depressor Effect of the Young Leaves of Polygonum Hydropiper Linn. In High-Salt Induced Hypertensive Mice. Biomedicine & Pharmacotherapy . 2018;102:1182–1187. doi: 10.1016/j.biopha.2018.03.055. [DOI] [PubMed] [Google Scholar]
  • 243.Malheiros J., Simões D. M., Antunes P. E., Figueirinha A., Cotrim M. D., Fonseca D. A. Vascular Effects of Polyphenols From Agrimonia eupatoria L. and Role of Isoquercitrin in Its Vasorelaxant Potential in Human Arteries. Pharmaceuticals . 2022;15(5):p. 638. doi: 10.3390/ph15050638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Liu L., Huang S., Xu M., et al. Isoquercitrin Protects HUVECs Against High Glucose-Induced Apoptosis Through Regulating P53 Proteasomal Degradation. International Journal of Molecular Medicine . 2021;48(1):122–211. doi: 10.3892/ijmm.2021.4955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Mendes-Junior L. D. G., Monteiro M. M. D. O., Carvalho A. D. S., Queiroz T. M. D., Braga V. D. A. Oral Supplementation With the Rutin Improves Cardiovagal Baroreflex Sensitivity and Vascular Reactivity in Hypertensive Rats. Applied Physiology Nutrition and Metabolism . 2013;38(11):1099–1106. doi: 10.1139/apnm-2013-0091. [DOI] [PubMed] [Google Scholar]
  • 246.Wang W., Wu Q.-h., Sui Y., Wang Y., Qiu X. Rutin Protects Endothelial Dysfunction by Disturbing Nox4 and ROS-Sensitive NLRP3 Inflammasome. Biomedicine & Pharmacotherapy . 2017;86:32–40. doi: 10.1016/j.biopha.2016.11.134. [DOI] [PubMed] [Google Scholar]
  • 247.Sattanathan K., Dhanapal C., Umarani R., Manavalan R. Beneficial Health Effects of Rutin Supplementation in Patients With Diabetes Mellitus. Journal of Applied Pharmaceutical Science . 2011;1(8):227–231. [Google Scholar]
  • 248.Kreft I., Golob A., Germ M. A Crop of High Nutritional Quality and Health Maintenance Value: The Importance of Tartary Buckwheat Breeding. Agriculture . 2023;13(9):p. 1783. doi: 10.3390/agriculture13091783. [DOI] [Google Scholar]
  • 249.Kianersi F., Abdollahi M. R., Mirzaie-asl A., Dastan D., Rasheed F. Identification and Tissue-Specific Expression of Rutin Biosynthetic Pathway Genes in Capparis Spinosa Elicited With Salicylic Acid and Methyl Jasmonate. Scientific Reports . 2020;10(1):p. 8884. doi: 10.1038/s41598-020-65815-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Yang Q., Song Z., Dong B., et al. Hyperoside Regulates its Own Biosynthesis via MYB30 in Promoting Reproductive Development and Seed Set in Okra. Plant Physiology . 2021;185(3):951–968. doi: 10.1093/plphys/kiaa068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Feng C., Chen J., Ye W., et al. Synthetic Biology-Driven Microbial Production of Resveratrol: Advances and Perspectives. Frontiers in Bioengineering and Biotechnology . 2022;10:p. 833920. doi: 10.3389/fbioe.2022.833920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Boyle S. P., Dobson V. L., Duthie S. J., Hinselwood D. C., Kyle J. A., Collins A. R. Bioavailability and Efficiency of Rutin as an Antioxidant: A Human Supplementation Study. European Journal of Clinical Nutrition . 2000;54(10):774–782. doi: 10.1038/sj.ejcn.1601090. [DOI] [PubMed] [Google Scholar]
  • 253.Farha A. K., Gan R. Y., Li H., et al. The Anticancer Potential of the Dietary Polyphenol Rutin: Current Status, Challenges, and Perspectives. Critical Reviews in Food Science and Nutrition . 2020;62(3):832–859. doi: 10.1080/10408398.2020.1829541. [DOI] [PubMed] [Google Scholar]
  • 254.Manach C., Morand C., Demigné C., Texier O., Régérat F., Rémésy C. Bioavailability of Rutin and Quercetin in Rats. FEBS Letters . 1997;409(1):12–16. doi: 10.1016/s0014-5793(97)00467-5. [DOI] [PubMed] [Google Scholar]
  • 255.Yang C.-Y., Hsiu S.-L., Wen K.-C., et al. Bioavailability and Metabolic Pharmacokinetics of Rutin and Quercetin in Rats. Journal of Food and Drug Analysis . 2020;13(3):p. 5. doi: 10.38212/2224-6614.2517. [DOI] [Google Scholar]
  • 256.Murray C. W., Booth A. N., DeEds F., Jones F. T. Absorption and Metabolism of Rutin and Quercetin in the Rabbit. Journal of the American Pharmaceutical Association . 1954;43(6):361–364. doi: 10.1002/jps.3030430613. [DOI] [PubMed] [Google Scholar]
  • 257.Alshahrani S. M. Development and Optimization of Oral Nanoemulsion of Rutin for Enhancing its Dissolution Rate, Permeability, and Oral Bioavailability. Pharmaceutical Development and Technology . 2022;27(5):588–597. doi: 10.1080/10837450.2022.2090957. [DOI] [PubMed] [Google Scholar]
  • 258.Remanan M. K., Zhu F. Encapsulation of Rutin Using Quinoa and Maize Starch Nanoparticles. Food Chemistry . 2021;353:p. 128534. doi: 10.1016/j.foodchem.2020.128534. [DOI] [PubMed] [Google Scholar]
  • 259.Raju M. G., Srivani M., Suvarchala Reddy N. V. L., Priya G. S. Formulation and Characterization of Rutin Loaded Chitosan Nanoparticles. Asian Journal of Research in Medical and Pharmaceutical Sciences . 2022;11(1):33–44. doi: 10.9734/ajrimps/2022/v11i130180. [DOI] [Google Scholar]
  • 260.Miyake K., Arima H., Hirayama F., et al. Improvement of Solubility and Oral Bioavailability of Rutin by Complexation With 2-Hydroxypropyl-β-Cyclodextrin. Pharmaceutical Development and Technology . 2000;5(3):399–407. doi: 10.1081/pdt-100100556. [DOI] [PubMed] [Google Scholar]
  • 261.Wang K., Zhang H., Yuan L., Li X., Cai Y. Potential Implications of Hyperoside on Oxidative Stress-Induced Human Diseases: A Comprehensive Review. Journal of Inflammation Research . 2023;16:4503–4526. doi: 10.2147/jir.s418222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Ji L., Shi W., Li Y., et al. Systematic Identification, Fragmentation Pattern, and Metabolic Pathways of Hyperoside in Rat Plasma, Urine, and Feces by UPLC-Q-Exactive Orbitrap MS. Journal of Analytical Methods in Chemistry . 2022;2022:1–14. doi: 10.1155/2022/2623018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Xu S., Chen S., Xia W., Sui H., Fu X. Hyperoside: A Review of its Structure, Synthesis, Pharmacology, Pharmacokinetics and Toxicity. Molecules . 2022;27(9):p. 3009. doi: 10.3390/molecules27093009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Guo J., Xue C.-f., Shang E.-x., Duan J., Tang Y., Qian D. Identification of Hyperoside Metabolites in Rat Using Ultra Performance Liquid Chromatography/Quadrupole-Time-of-Flight Mass Spectrometry. Journal of Chromatography B . 2011;879:1987–1992. doi: 10.1016/j.jchromb.2011.04.031. [DOI] [PubMed] [Google Scholar]
  • 265.Yang J., Qian D., Guo J., et al. Identification of the Major Metabolites of Hyperoside Produced by the Human Intestinal Bacteria Using the Ultra Performance Liquid Chromatography/Quadrupole-Time-of-Flight Mass Spectrometry. Journal of Ethnopharmacology . 2013;147(1):174–179. doi: 10.1016/j.jep.2013.02.029. [DOI] [PubMed] [Google Scholar]
  • 266.Paulke A., Eckert G. P., Schubert-Zsilavecz M., Wurglics M. Isoquercitrin Provides Better Bioavailability Than Quercetin: Comparison of Quercetin Metabolites in Body Tissue and Brain Sections After Six Days Administration of Isoquercitrin and Quercetin. Die Pharmazie . 2012;67(12):991–996. [PubMed] [Google Scholar]
  • 267.Valentová K., Vrba J., Bancířová M., Ulrichová J., Křen V. Isoquercitrin: Pharmacology, Toxicology, and Metabolism. Food and Chemical Toxicology . 2014;68:267–282. doi: 10.1016/j.fct.2014.03.018. [DOI] [PubMed] [Google Scholar]
  • 268.Lu L., Qian D., Yang J., et al. Identification of Isoquercitrin Metabolites Produced by Human Intestinal Bacteria Using UPLC-Q-TOF/MS. Biomedical Chromatography . 2013;27(4):509–514. doi: 10.1002/bmc.2820. [DOI] [PubMed] [Google Scholar]
  • 269.Owczarek-Januszkiewicz A., Magiera A., Olszewska M. A. Enzymatically Modified Isoquercitrin: Production, Metabolism, Bioavailability, Toxicity, Pharmacology, and Related Molecular Mechanisms. International Journal of Molecular Sciences . 2022;23(23):p. 14784. doi: 10.3390/ijms232314784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Kapoor M. P., Moriwaki M., Uguri K., Timm D., Kuroiwa Y. Bioavailability of Dietary Isoquercitrin-γ-Cyclodextrin Molecular Inclusion Complex in Sprague–Dawley Rats and Healthy Humans. Journal of Functional Foods . 2021;85:p. 104663. doi: 10.1016/j.jff.2021.104663. [DOI] [Google Scholar]
  • 271.Walle T., Hsieh F., DeLegge M., Oatis J. E., Walle U. K. High Absorption but Very Low Bioavailability of Oral Resveratrol in Humans. Drug Metabolism and Disposition . 2004;32(12):1377–1382. doi: 10.1124/dmd.104.000885. [DOI] [PubMed] [Google Scholar]
  • 272.Wenzel E., Somoza V. Metabolism and Bioavailability of Trans-resveratrol. Molecular Nutrition & Food Research . 2005;49(5):472–481. doi: 10.1002/mnfr.200500010. [DOI] [PubMed] [Google Scholar]
  • 273.Smoliga J. M., Blanchard O. Enhancing the Delivery of Resveratrol in Humans: If Low Bioavailability Is the Problem, what Is the Solution? Molecules . 2014;19(11):17154–17172. doi: 10.3390/molecules191117154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Andlauer W., Kolb J., Siebert K., Fürst P. Assessment of Resveratrol Bioavailability in the Perfused Small Intestine of the Rat. Drugs Under Experimental and Clinical Research . 2000;26(2):47–55. [PubMed] [Google Scholar]
  • 275.Cottart C.-H., Nivet-Antoine V., Laguillier-Morizot C., Beaudeux J. L. Resveratrol Bioavailability and Toxicity in Humans. Molecular Nutrition & Food Research . 2010;54(1):7–16. doi: 10.1002/mnfr.200900437. [DOI] [PubMed] [Google Scholar]
  • 276.Kapetanovic I. M., Muzzio M., Huang Z., Thompson T. N., Mccormick D. L. Pharmacokinetics, Oral Bioavailability, and Metabolic Profile of Resveratrol and its Dimethylether Analog, Pterostilbene, in Rats. Cancer Chemotherapy and Pharmacology . 2011;68(3):593–601. doi: 10.1007/s00280-010-1525-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Johnson J. J., Nihal M., Siddiqui I. A., et al. Enhancing the Bioavailability of Resveratrol by Combining It With Piperine. Molecular Nutrition & Food Research . 2011;55(8):1169–1176. doi: 10.1002/mnfr.201100117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Tamura T., Mitsumori K., Muto S., et al. Fifty-Two Week Chronic Toxicity of Enzymatically Decomposed Rutin in Wistar Rats. Food and Chemical Toxicology . 2010;48(8-9):2312–2318. doi: 10.1016/j.fct.2010.05.065. [DOI] [PubMed] [Google Scholar]
  • 279.Valentová K., Vrba J., Bancířová M., Ulrichová J., Křen V. Isoquercitrin: Pharmacology, Toxicology, and Metabolism. Food and Chemical Toxicology . 2014;68:267–282. doi: 10.1016/j.fct.2014.03.018. [DOI] [PubMed] [Google Scholar]
  • 280.Owczarek-Januszkiewicz A., Magiera A., Olszewska M. A. Enzymatically Modified Isoquercitrin: Production, Metabolism, Bioavailability, Toxicity, Pharmacology, and Related Molecular Mechanisms. International Journal of Molecular Sciences . 2022;23(23):p. 14784. doi: 10.3390/ijms232314784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Crowell J. A., Korytko P. J., Morrissey R. L., Booth T. D., Levine B. S. Resveratrol-Associated Renal Toxicity. Toxicological Sciences . 2004;82(2):614–619. doi: 10.1093/toxsci/kfh263. [DOI] [PubMed] [Google Scholar]
  • 282.Shaito A., Posadino A. M., Younes N., et al. Potential Adverse Effects of Resveratrol: A Literature Review. International Journal of Molecular Sciences . 2020;21(6):p. 2084. doi: 10.3390/ijms21062084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Xu S., Chen S., Xia W., Sui H., Fu X. Hyperoside: A Review of its Structure, Synthesis, Pharmacology, Pharmacokinetics and Toxicity. Molecules . 2022;27(9):p. 3009. doi: 10.3390/molecules27093009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Ai G., Huang Z., Wang D., Zhang H. Acute Toxicity and Genotoxicity Evaluation of Hyperoside Extracted From Abelmoschus Manihot (L.) Medic. Journal of Chinese Pharmaceutical Sciences . 2012;21(5):2811–2816. doi: 10.5246/jcps.2012.05.063. [DOI] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from Advances in Pharmacological and Pharmaceutical Sciences are provided here courtesy of Wiley

RESOURCES