Next Article in Journal
Tropylium Ion, an Intriguing Moiety in Organic Chemistry
Previous Article in Journal
Preparation of PO43−-Intercalated Calcium–Aluminum Hydrotalcites via Coprecipitation Method and Its Flame-Retardant Effect on Bamboo Scrimber
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ethnobotany, Biological Activities and Phytochemical Compounds of Some Species of the Genus Eryngium (Apiaceae), from the Central-Western Region of Mexico

by
Jeanette G. Cárdenas-Valdovinos
,
Ignacio García-Ruiz
,
María V. Angoa-Pérez
and
Hortencia G. Mena-Violante
*
Instituto Politécnico Nacional, Department of Research, CIIDIR IPN Unidad Michoacán, Jiquilpan 59510, Mexico
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(10), 4094; https://doi.org/10.3390/molecules28104094
Submission received: 7 April 2023 / Revised: 28 April 2023 / Accepted: 10 May 2023 / Published: 15 May 2023
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
There are approximately 250 species of Eryngium L. distributed throughout the world, with North America and South America being centers of diversity on this continent. In the central-western region of Mexico there may be around 28 species of this genus. Some Eryngium species are cultivated as leafy vegetables, ornamental, and medicinal plants. In traditional medicine they are used to treat respiratory and gastrointestinal conditions, diabetes, and dyslipidemia, among others. This review addresses the phytochemistry and biological activities, as well as traditional uses, distribution, and characteristics of the eight species of Eryngium reported as medicinal in the central-western region of Mexico: E. cymosum, E. longifolium, E. fluitans (or mexicanum), E. beecheyanum, E. carlinae, E. comosum, E. heterophyllum, and E. nasturtiifolium. The extracts of the different Eryngium spp. have shown biological activities such as hypoglycemic, hypocholesterolemic, renoprotective, anti-inflammatory, antibacterial, and antioxidant, among others. E. carlinae is the most studied species, and phytochemical analyses, performed mainly by high-performance liquid chromatography (HPLC) and gas chromatography coupled with mass spectrometry (GC-MS), have shown its content of terpenoids, fatty acids, organic acids, phenolic acids, flavonoids, sterols, saccharides, polyalcohols, and aromatic and aliphatic aldehydes. According to the results of this review on Eryngium spp., they constitute a relevant alternative as a source of bioactive compounds for pharmaceutical, food, and other industries. However, there is a lot of research to be conducted regarding phytochemistry, biological activities, cultivation, and propagation, in those species with few or no reports.

1. Introduction

Plants constitute a valuable resource in the health systems of developing countries, and in this sense, the World Health Organization (WHO) has estimated that more than 80% of the world population uses traditional medicine for primary health care, mainly through treatments with plant extracts or their active ingredients [1]. According to the WHO (1979) [2] a medicinal plant is defined as any plant species that contains substances which can be used for therapeutic purposes or whose active ingredients can serve as precursors for the synthesis of new drugs. Therefore, drugs derived from plants have gained relevance both in traditional medicine and in modern medicine [3]. However, Ref. [1] the massive use of medicinal plants is limited by procedures such as the classification, identification and characterization of these and their active principles; therefore, the generation of knowledge about medicinal plants in different latitudes is highly relevant.
In Mexico, medicinal plants have been historically important even before colonization of the New World; thus, knowledge of plants and their usefulness as medicine, have passed from generation to generation as part of inherited traditions. There are people who master the empirical knowledge of medicinal plants, while others only sell them unaware of their properties and health effects [4]. The diversity of plants is very high in Mexico, and between 3000 and 5000 species are plants with therapeutic potential and only 1% have been studied in depth to discover their medicinal properties [5]. Among the plants with medicinal properties are those belonging to the genus Eryngium, the largest and most complex of the Apiaceae family. There are 250 species of which 28 are distributed in the central-western region of the country and 21–22 in Michoacán [6]. Therapeutic uses have been reported for some of these Eryngium species mostly in rural communities, and some of them have been studied due to their beneficial properties, mainly to treat diabetes, dyslipidemia, and kidney conditions [7,8,9,10]. Extracts from some Eryngium spp. have been shown to have biological activities [11,12] and various bioactive compounds [13]. However, only a few species have been investigated to ensure the efficacy and safety of the treatments for which they are prescribed.
Consistent with the above information, the main objectives of the present work were to review (1) the traditional uses associated with the Eryngium spp. distributed in the central-western region of the country; (2) the advances in the detection and identification of their bioactive compounds; and (3) the biological activities that reveal their healing potential. Having this information available will allow us to propose future research to contribute to the development of safe products and therapies aimed at treating chronic degenerative diseases, among others.

2. Ethnobotany: Description, Distribution, and Traditional Uses

2.1. The Genus Eryngium Worldwide and in Mexico

There are approximately 230–250 species of Eryngium L. distributed in tropical and temperate regions in Eurasia, North Africa, North and South America, and Australia [14,15]. Except in the tropics and Southern Africa [16,17], Eryngium L. is the largest genus and possibly the most taxonomically complex of the Apiaceae family [18]. In his treatise, Wolff (1913) [19] considered Eryngium one of the most complex, and he recognized two groups within the genus: “Species Gerontogeae” representing 12 sections of the Old World (Africa, Europe, and Asia) and “Species Americanae and Australienses” including 22 sections from the New World (America and Australia). According to Wörz (1999) [15], and in agreement with updated information, there are about 100 South American and 85 North American species. In Mexico, there are around 50–60 species: between 27 and 28 in the central-western region of the country, and 21–22 in the state of Michoacán [6].

2.2. The Genus Eryngium Worldwide and in Mexico

Eryngium L.

Creeping to erect, herbaceous, caulescent or acaulescent, these plants are usually glabrous biennials or perennials, which grow from stout taproots or rootstocks bearing fibrous roots. The leaves are coriaceous or membranaceous, entire, or pinnately or palmately lobed to divided, often ciliate to spinose, and the venation parallel or reticulate. The petiole’s sheathing is sometimes septate. It has an inflorescence capitate, with the heads either solitary or in cymes or racemes. It has an involucre of one or more series of entire or lobed bracts subtending the head and an involucel of entire or lobed bractlets subtending the flowers. The flowers are white to purple and sessile; the petals ovate to oblong with variously inflexed and lobed to fimbriate tips; the sepals ovate to lanceolate, acute to obtuse, entire, or rarely spinescent; the styles are shorter and do not exceed the sepals, as they lack a stylopodium; and the carpophore is absent. The fruit are globose to obovoid, scarcely flattened laterally, and variously covered with scales or tubercles; the ribs are obsolete; the commissure is broad; and the five oil-tubes are mostly inconspicuous. The seeds subterete in cross-section, and the face is plane or slightly concave [20].
Worldwide, several species of the genus Eryngium have been used in traditional medicine, especially to treat cholesterol and diabetes problems. In Mexico there are about 50 species, of which about 27 taxa are considered for the central-west region, which includes the states of Michoacán and Jalisco, as well as areas adjacent to the neighboring states: State of Mexico, Guanajuato, and the southern zone of Nayarit [20, 21,22] (Table 1). Various properties have been reported for only eight taxa since they are traditionally used as an auxiliary or complement to family health.
The species reported in this review fall into the section and subsection at the infrageneric level, according to Wolff (1913) [19] and Calviño et al. (2008) [18], as “American and Australian species (new world)” as follows (Table 2).
Some characteristics on the biology and distribution of the Eryngium species reported in the central-western region of Mexico are shown in Table 3 [6].

2.3. Traditional Uses

Many Eryngium species have traditionally been used as ornamental, edible, or medicinal plants [24]. Various Eryngium spp. are used for the treatment of different inflammatory diseases around the world [25,26,27]. In Mexico, the use of different Eryngium species in traditional medicine is very important, some of them are distributed in the central-western region (Table 4). In general, they are known as “hierba del sapo” (toad grass) regardless of the species. Such is the case of Eryngium beecheyanum Hook. F. and Arn., a species mostly preferred by the population of the Purépecha Plateau in Michoacán, Mexico, to treat skin inflammation; consumed orally as an infusion of the aerial part of the plant, it is also applied in fomentation form to the affected area [28]. It has been reported as an antipyretic [29], and the farmers of the Sierra de Huautla, Morelos, use it to treat renal inflammation [10].
Eryngium carlinae F. Delaroche, also known as “hierba del sapo” or “mosquitas” [12,30,31], whose medicinal properties have been widely studied, is also one of the most used in indigenous communities in central Mexico as well as in Michoacán, Hidalgo, State of Mexico and Querétaro [32,33]. It is mainly used in the treatment of kidney problems such as cystitis known as “mal de orín” in Puebla and Tlaxcala, kidney pain in Hidalgo, and as a diuretic in the State of Mexico. In general, the whole plant is used with or without the roots and taken orally as an infusion. It is also used to reduce inflammation of the stomach in the State of Mexico, and to treat biliary disease, taken on an empty stomach. In the case of inflammation caused by blows, it is topically applied to the affected area, using hot fomentations, and adding salt to the infusion. E. carlinae is also used to treat inflammatory conditions of the bowel, back pain, pain of the bones, chest, ear, and hernia, and against snake bites and fever [9,32]. One of the most popular uses is in the treatment of diabetes and dyslipidemias as an infusion taken throughout the day called “agua de uso” [9,32]. Additional uses have been reported, including as an aphrodisiac, an anticrotalic antidote, antispasmodic, antipodagric, antitumor, carminative, diuretic, expectorant, increased heat in the stomach, cold-caused illnesses, asystole, and gastroenteritis, among others [29].
Eryngium comosum F. Delaroche, is used as an aphrodisiac, antigonorrheal, antipyretic, antipodagric, diuretic and oxytocic [29], for lipid-lowering, and to treat “mal de orín” (cysititis) related to urinary tract infections and kidney pain [34].
Eryngium longifolium is another of the species used to treat diabetes in Hidalgo, where it is given the name “piñuela”, where the form of use is as an infusion of the dried plant (aerial part) taken throughout the day (“agua de uso”) [35,36]. In addition, its use as a diuretic, emmenagogue, and alexiteric has been reported [37], similar to Eryngium fluitans [38]. Another empirically well-known species is Eryngium heterophyllum Engel, also called “toad grass”, which is considered useful for the treatment of diabetes, arthritis, and hypercholesterolemia, among other diseases. The commonly recommended form of use is to prepare the aerial part of the plant in its natural state, pulverized and boiled in water, and the infusion is supplied according to the condition [4,8]. It has also been reported to be useful for the control of gallstones [23].
Eryngium nasturtiifolium is known to be used locally as a traditional medicine against type 2 diabetes mellitus and “mal de orín” [39], similar to other Eryngium species.
The traditional medicinal uses of Eryngium species reviewed are summarized in Table 4.
Table 4. Eryngium spp. distributed in the central-western region of Mexico with reports of medicinal use.
Table 4. Eryngium spp. distributed in the central-western region of Mexico with reports of medicinal use.
Scientific NameCommon NameMedicinal UsesPreparationReferences
E. beecheyanumHierba del sapoAntipyreticInfusion of the whole plant; fomento[29]
For kidney inflammationNR[10]
For skin inflammationInfusion of the aerial part,
taken orally; additionally, it is used in the fomentation form on skin
[28]
E. carlinaeHierba del sapo,
mosquitas
Aphrodisiac, anticrotalic, antispasmodic, antipodagric, antitumor, asystole, carminative, diuretic, tonic, and expectorant; in cold-caused diseases, it increases heat in the stomach; and to treat gastroenteritisInfusion of the whole plant[29]
To treat type 2 diabetes, dyslipidemias, and digestive problems; and to control blood pressureInfusion[8,9]
Diuretic and antipyretic; to treat kidney problems, “mal de orín” (cystitis), and kidney pain; to control bile (taken on an empty stomach); to treat stomach and intestine inflammation, pain in the back, bones, chest and hernia; for snake bites; and in piercing earsInfusion, whole plant with or without root, it is taken orally[33]
For inflammations due to blowsIt is applied topically, through hot fomentations adding salt to the infusion[33]
E. comosumHierba del sapo, piñitasAphrodisiac, antigonorrheal, antipyretic, antipodagric, diuretic, and oxytocicInfusion of the whole plant[29]
Hypolipidic; to treat cystitis (“mal de orín”)NR[34]
E. cymosum To treat type 2 diabetes; and as a hypoglycemicInfusion: as “agua de uso”[8,9,40]
E. fluitans Diuretic, emmenagogue, and alexitericNR[38]
E. heterophyllumHierba del SapoTo treat diabetes, arthritis, and hypercholesterolemia; and
to control bile and reduce gallstones related to emotional problems (e.g., anger)
Infusion, whole plant[4,41]
To control of gallstonesNR[23]
E. longifoliumPiñuelaTo treat type 2 diabetes; and as a diuretic, emmenagogue, and alexitericInfusion aerial part of the plant: as “agua de uso”[35,36,37,38]
E. nasturtiifoliumHierba del sapoTo treat type 2 diabetes and cystitis (“mal de orín”)Infusion whole plant: as “agua de uso”[39]
NR—Not reported.

3. Biological and Pharmacological Activity

The use of plants in medicines ranges from crude preparations or extracts, to refined extracts and single molecular species. In terms of categories of use, these encompass food supplements, herbal medicines, botanical drugs, and prescription medicines. There is an increasing interest in plants as a source of novel pharmacophores [42].
In this context, pharmacological studies of medicinal plants have been carried out, addressing various extract evaluation strategies in vitro or in vivo, using different extractive solvents or following traditional preparation practices. The plants of the genus Eryngium have not been the exception; thus, the evaluation of the extracts of Eryngium spp. distributed around the world, have shown multiple beneficial effects [43], such as anti-inflammatory [44], against snake and scorpion venoms [45,46], antibacterial, antioxidant [47], antihyperglycemic [48], and cytotoxic against human tumor cell lines [49], among others.
Regarding the Eryngium spp. reported in the central-western region of Mexico, records of its use in traditional medicine were found for eight species, but reports of biological activities were found only for five of these species.

3.1. Eryngium carlinae

There has been great interest in learning about its effects on diabetes control; thus, Noriega-Cisneros et al. (2012) [50] investigated the effect of chronic administration of ethanolic extract of E. carlinae on glucose, creatinine, uric acid, total cholesterol, and triglyceride levels in the serum of streptozotocin (STZ)-induced diabetic rats. Treatment with ethanolic extract of E. carlinae prevented the increase in glucose, triglycerides, total cholesterol, and uric acid in serum; it also reduced the levels of creatinine, uric acid, total cholesterol, and triglycerides in healthy rats compared to those with diabetes. Additionally, ethanolic extract significantly decreased glycosylated hemoglobin (HbA1c) in the serum of diabetic rats. The authors concluded that administration of E. carlinae reduced cardiovascular-risk-related hyperlipidemia in diabetes mellitus. Subsequently, Noriega-Cisneros (2013) [51] analyzed the chemical composition of the ethanolic extract of E. carlinae and studied the effect of its consumption in STZ-induced diabetic rats, and its antioxidant activity was assayed. The results showed that ethanolic extract had no hypoglycemic effect when administered orally to diabetic rats (45 mg/kg); however, it did reduce cholesterol and triglyceride levels, improving the lipid profile and reducing the cardiovascular risk index. The in vitro analysis showed antioxidant activity and a considerable amount of flavonoids and phenolic compounds related to it; however, the in vivo analysis did not have a significant effect on lipid peroxidation, and antioxidant enzymatic activity of the superoxide dismutase (SOD) and catalase (CAT) only showed an effect on reducing the nitric oxide levels. Histological analysis of the kidney showed that although the ethanolic extract of E. carlinae did not control hyperglycemia, it may offer benefits on lipid profile and progression of renal damage. Later, Noriega-Cisneros et al. (2020) [31] investigated the mechanism of action of the hypolipidemic effect of the ethanolic extract of E. carlinae, analyzing its composition and lipid-lowering activity. The extract was administered orally to STZ-induced diabetic rats (30 mg/kg) for more than 40 days, and its effect was compared with that of atorvastatin (a drug used to lower cholesterol levels). The analyzed extract reduced total cholesterol and non-high-density lipoprotein cholesterol (C-HDL) levels and increased the C-HDL levels reduced in diabetes, decreasing the atherogenic index and, therefore, the risk of suffering cardiovascular disease risk at the same level as atorvastatin. The results demonstrated the hypolipidemic potential of ethanolic extract of E. carlinae and support its use in traditional medicine as a hypolipidemic agent. On the other hand, García-Cerrillo et al. (2018) [52] demonstrated that the hexanic extract of E. carlinae had in vitro and in vivo antioxidant activity associated with the decrease in glucose and triacylglyceride levels during hyperglycemia and suggested that this effect could reduce the risk of developing diabetic cardiomyopathy. The authors administered hexanic extract of E. carlinae (30 mg/kg) to STZ-induced diabetic rats for seven weeks and found that serum levels of glucose, triacylglycerides, and TBARS (thiobarbituric acid reactive substances) were significantly reduced in diabetic rats supplemented with the extract. Peña-Montes et al. (2019) [30] also evaluated the in vitro antioxidant activity of the hexanic extract of E. carlinae inflorescences in Saccharomyces cerevisiae under stress induced by hydrogen peroxide, and later, they tested the extract in STZ-induced diabetic male Wistar rats. The hexanic extract showed in vitro antioxidant activity at different concentrations compared to ascorbic acid (positive control). Oral administration (30 mg/kg) of the hexanic extract reduced blood glucose levels; lipid peroxidation in the liver, kidney, and brain; protein carbonylation; and reactive oxygen species (ROS) production in normoglycemic and hyperglycemic rats. CAT activity in the brain, kidneys, and liver also increased. These findings showed the antioxidant properties of the hexanic extract of E. carlinae inflorescences.
Regarding active metabolites, Castro-Torres et al. (2017) [53] determined the hypocholesterolemic activity of the hydroalcoholic extract of aerial parts of E. carlinae and demonstrated the presence of hexa-O-acetyl-d-mannitol and its acetylated derivatives by gas chromatography coupled with mass spectrometry (GC-MS) analysis. The authors concluded that mannitol promoted osmotic diuresis, which may favor cholesterol transport, preventing it from accumulating in enterocytes and the development of hypercholesterolemia; in this sense, mannitol-based drugs are used to promote diuresis (before irreversible renal failure) and urinary excretion of toxic substances as an antiglaucoma agent, and as an aid in the diagnosis of renal function [54]. In the same way the diuretic effect and the excretion of toxic substances, the renoprotective activity of E. carlinae has been reported by Pérez-Ramírez et al. (2016) [55]. The authors studied the effect of plant decoctions on renal dysfunction in high-fructose and high-fat fed rats. Decoction consumption reduced serum uric acid, urine albumin and urea, and increased creatinine clearance, which was associated with reduced hyperglycemia, renal lipid accumulation, and oxidative stress. These results suggested that E. carlinae could be used as an ingredient of functional beverages with renoprotective effects.
The only clinical study of E. carlinae found during this review was reported by Montes-Moreno (2017) [32]. The authors evaluated the effect of consuming aqueous extracts of toad grass on serum triglycerides, body composition, and anthropometric values in adults. A randomized, parallel blind clinical trial was carried out, and anthropometric measurements, body composition, and blood biochemistry were taken. Individuals with triglycerides >150 mg/dL were selected to determine the effect of drinking the aqueous extract on the baseline parameters of the participants after four weeks. The consumption of the extract reduced weight and body fat (approx. 1 kg) and triglycerides and VLDL cholesterol (21%). The results obtained suggested that drinking the infusion at a 1% concentration at least once a day could reduce and/or control high serum triglyceride levels and be an adjuvant in reducing the percentage of body fat and weight.
Another reported use of Eryngium spp. is the treatment of cholelithiasis; therefore, Valdivia-Mares (2021) [56] evaluated the effectiveness of a 50% hydroalcoholic extract of E. carlinae to treat cholelithiasis by an in vitro dissolution model using 30 stones formed by ≥70% cholesterol selected from 1597 stones obtained by cholecystectomy. To improve solubility and resemble gallbladder conditions, the test temperature was between 35 and 37 °C, and the extract was renewed every hour for 20 h. Solutions of 50% ethanol and 99% ethyl ether were used as negative and positive controls, respectively. The dissolution rate of the media was estimated as the reduction in the mass of the treated stones (g/mL/h). The extract showed a higher dissolution rate (0.00280–0.00285 g/mL/h) than that shown by ethanol (0.00255 g/mL/h) and six times lower than that shown by ethyl ether (0.00715 g/mL/h). The authors suggested that these results could contribute to the development of a safer, cheaper, and less invasive therapy, such as a product containing E. carlinae.
Another of the benefits attributed to E. carlinae is its antispasmodic activity, which was confirmed in vivo by Pérez-Gutiérrez et al. (2006) [57]. This activity was attributed to the presence of two γ-lactones from the methanol fraction isolated and characterized by the authors.
Regarding the antimicrobial activity of E. carlinae, tests performed in vitro have not shown significant growth inhibition of human pathogenic bacteria [11]. On the other hand, Galindo-Hernández (2018) [58] evaluated the antifungal activity of the acetonic extract of E. carlinae against Candida spp. strains isolated from pediatric dental patients. The extract did not show strong antimicrobial activity against C. albicans (ATCC 90029). In contrast to the above antimicrobial studies, Espino-Garibay (2010) [59] evaluated the antimicrobial effect of E. carlinae metabolites, identifying 21 metabolites in ethanolic extracts (leaves, peduncles, and flowers) by GC-MS. Regarding the volatile compounds, germacrene showed antifungal activity against Colletotrichum lindemuthianum (49.6%) and Botrytis cinerea (39.1%). The highest antifungal activity against C. lindemuthianum (almost 100%) was shown by spathulenol (50 mg/mL) and piperitone oxide (500 mg/mL). While spathulenol, piperitone oxide, and menthol (100 mg/mL) exerted a less inhibitory effect against B. cinerea (37.8%), only piperitone oxide (250 mg/mL) had an inhibitory effect against Fusarioum oxysporum (28.8%). On the other hand, the antimicrobial activity of E. carlinae terpenoids was lower; thus, pulegone and borneol, with a dose of 500 mg/mL, inhibited the oomycete Phytophthora cinammomis 32.1 and 30.8%, respectively. Meanwhile, spathulenol (500 mg/mL) and myrcene (250 mg/mL) exerted an inhibitory effect of 18.1 and 15.3%, respectively. The crude extracts showed higher activity against P. cinnamomi (34%).

3.2. Eryngium comosum

There are few scientific reports on the biological activities of Eryngium comosum, Delaroche F.; for example, the work of Ronquillo de Jesús (2013) [60] who determined the antioxidant activity of ethanolic, aqueous, hexanic, and ether of petroleum extracts of E. comosum using the DPPH assay. In addition, the extracts cytotoxicity was assayed in vitro in peripheral blood mononuclear cells and in vivo in Artemia salina. Ethanolic and aqueous extracts at a concentration of 1000 ppm showed IC50 values of 4.93 µg/mL and 49.52 µg/mL, respectively. None of the extracts showed toxicity in mononuclear cells, while the extract with petroleum ether did show a cytotoxic effect in A. salina (IC50 2.92 ppm).
The antimicrobial activity has also been studied in E. comosum, in addition to the antioxidant activity. Díaz-Alvarado et al. (2020) [61] evaluated the antibacterial activity by the disk diffusion method (DDM), using reference strains of equine pathogenic bacteria: Listeria monocytogenes ATCC 19115, Staphylococcus sp., Escherichia coli ATCC 25922, and Salmonella enterica serotype Enteritidis ATCC 13076. Ethanolic extract of E. comosum (50%) prepared with dried tissue (125 mg/mL) inhibited the growth of Staphylococcus sp., S. enterica, and L. monocytogenes, showing a greater effect on the latter strain. The results suggested the extract of E. comosum as a source of antimicrobial agents to treat equine infections, although further in vitro and in vivo research is required to achieve its application. In the same way, Díaz-Alvarado (2020) [62] analyzed the bioactive compounds in aqueous and ethanolic extracts (50 and 70%) of E. comosum, and assayed antioxidant capacity and antimicrobial activity in 50% ethanolic extracts of this medicinal plant. The 50% ethanolic extract of E. comosum showed antioxidant capacity (1973.42 μM ETCA/g) and antibacterial activity against Enterococcus sp. and Salmonella sp. (inhibition zone diameter = 11.3 mm).
Regarding in vivo studies, Pérez-Reyes (2016) [13] reported that the aqueous extract of E. comosum reduced cholesterol and triglyceride levels in rats with dyslipidemia, induced with a hypercholesterolemic and hypertriglyceridemic diet. The extract was administered intragastrically for 3 weeks, testing three doses: 100, 200, and 400 mg/kg. After the treatment, the influence of the aqueous extract on the weight of adipose and muscle tissues was observed; however, a body weight reduction was not reported. On the other hand, the decrease in serum cholesterol levels was recorded at a dose of 100 mg/kg, with a serum concentration of 189.45 mg/dL in the control group and 99.16 mg/dL in the treated group; while the serum concentration of triglycerides decreased only with the 200 mg/kg dose, being 403.1 mg/dL in the control group and 337.8 mg/dL in the treated group.

3.3. Eryngium cymosum

One of the most widespread traditional uses of Eryngium ssp. is the treatment of type 2 diabetes (T2D), and there are some reports about its hypoglycemic effect. For example, the study carried out by Espinoza-Hernández et al. (2021) [8], in which the aqueous extract of aerial parts of the E. cymosum plant was administered via gavage to Wistar rats with streptozotocin-nicotinamide-induced hypoglycemia (STZ-NA). The authors reported the antihyperglycemic effect of the extract in the pyruvate tolerance test and the significant reduction of postprandial hyperglycemia in the maltose tolerance tests. As the main mechanism of action, the extract suppressed gluconeogenesis by inhibition (almost 100%) of the enzymes glucose-6-phosphatase (G6Pase) and fructose-1,6-bisphosphatase (FBPase), which is the altered pathway that causes fasting and postprandial hyperglycemia in patients with T2D; the extract also reduced the activity of a-glucosidases by 32%. In addition, it decreased insulin levels when it was administered orally in healthy rats in both nutritional states, without affecting normoglycemia in normal curves and reducing the postprandial peak in glucose load curves. The authors concluded that the traditional form of consumption of E. cymosum is safe and regulates glucose levels both fasting and in the postprandial state.
Subsequently, the same research group published a study that evaluated the chronic effects of traditional extracts on hyperglycemia and hypertriglyceridemia of some Mexican medicinal plants, including E. cymosum [9]. The aqueous extract was administered via gavage to hyperglycemic STZ-NA Wistar rats, daily for 42 days. For the preparation of the extract, 20 g of dried and ground plant material (aerial parts) were added to 500 mL of boiling, distilled water for 15 min. Non-fasting blood glucose (NFBG), HbA1c, and blood triglycerides were determined. The authors confirmed the long-term efficacy of the extract, as E. cymosum prevented the worsening of hyperglycemia by avoiding the significant increase in glucose levels shown by the negative control group and the increase in HbA1c (2.98%). Despite its antihyperglycemic effects, the extract was less effective in controlling triglycerides. The authors generated evidence of the antihyperglycemic effect of this Mexican medicinal plant, as well as its long-term efficacy in the control of T2D.
Research to reveal the mechanisms of action of the hypoglycemic effect of E. cymosum has led to the description of a new metabolite, acylated flavonol, and the isolation of known compounds both in aqueous extract and butanolic extract [40], whose chemical structures were elucidated using spectroscopic techniques, as described in the following section. Additionally, the role of the acylated flavonol glucoside on the inhibition of G6Pase and FBPase has been demonstrated.

3.4. Eryngium heterophyllum

Some studies have been carried out in E. heterophyllum to confirm its anti-inflammatory, hypoglycemic, and hypocholesterolemic activity. Navarrete et al. (1990) [63] reported the decrease in rat serum cholesterol when the aqueous extract of E. heterophyllum was administered orally. For his part, Miranda-Velásquez (2010) [64] tested the hypocholesterolemic activity of crude extracts dissolved in water or in a Tween 80/saline solution at two doses of 50 and 100 mg/kg of weight administered to hypercholesterolemic mice for five days, which at the end of this period were fasted for 12 h. The results showed that only the aqueous extracts of E. heterophyllum at 100 mg/kg showed a cholesterol reduction (20.7%). Therefore, this extract was subsequently evaluated in vitro using Vero cells to determine the inhibitory effect of the HMG-CoA enzyme. The results showed that indeed the mechanism of serum cholesterol reduction was related to the inhibition of said enzyme, as in the case of statin drugs. In the same way, García-Gómez et al. (2019) [65], in a 1-month clinical study, showed that combined treatment of E. heterophyllum and Amphipterygium adstringens with proven hypocholesterolemic activity tested in rats, reduced triglyceride levels by an average of 20%. On the other hand, Carreón-Sánchez et al. (2013) [4] showed that the ethanolic extract of E. heterophyllum, after being administered to mice by oral gavage in a single dose of 100 mg/kg of weight in a volume of 0.2 mL/30 g, had no hypoglycemic effect or acute or chronic anti-inflammatory effect; nor did it cause visible toxic effects in the acute poisoning model in mice.
Molina-Garza et al. (2014) [66] conducted a study to screen the trypanocidal activity of the Eryngium heterophyllum plants used in traditional Mexican medicine for the treatment of various diseases related to parasitic infections. Cultured Trypanosoma cruzi epimastigotes were incubated for 96 h with different concentrations of methanolic extract of E. heterophyllum, and the inhibitory concentration (IC50) was determined. The methanolic extracts exhibited the highest trypanocidal activity (88–100%) at a concentration of 150 µg/mL.

3.5. Eryngium longifolium

A dose-independent hypoglycemic effect has been reported for E. longifolium. Andrade-Cetto et al. (2021) [35] evaluated aqueous (30 and 310 mg/kg doses) and ethanolic (32 and 318 mg/kg doses) extracts of the aerial parts of the plant in hyperglycemic STZ-NA Wistar rats. Previously, the authors determined the basic phytochemical profiles (see next section) and acute toxicity tests, which did not show any physical problems or behavioral changes after oral administration of the maximum dose of 2000 mg/kg body weight (b.w.) of each extract; no deaths were reported and the LD50 was higher than the maximum dose used. In addition, they tested the inhibition of the G6Pase and FBPase enzymes involved in glucose metabolism. This study validated for the first time the traditional use of the aerial part of E. longifolium as a hypoglycemic agent in a hyperglycemic animal model; the results indicated that the in vitro inhibition of G6Pase and FBPase could be associated with the hypoglycemic effect in vivo. Therefore, the authors concluded that the ability to regulate hyperglycemia could involve inhibition of hepatic glucose production, which primarily controls fasting glucose levels, and that the doses traditionally consumed did not generate toxic effects.
According to the previous information, the main potential of the Mexican species of Eryngium to promote health, is related to lipid metabolism, which has been proven by the capacity of its extracts to decrease cholesterol, triglycerides, and body fat levels; this has also been reported for other species of Eryngium [67].
It is important to highlight the potential of the aqueous extracts of E. cymosum and E. longifolium for the control of diabetes, as reported for E. foetidum and E. billardieri [67,68]. The information found about hypoglycemic activity shows the differences between the biological and pharmacological activity that different Mexican species of Eryngium show; in addition, such differences could be associated with the type of extract evaluated since the content and nature of the active ingredients will also vary. For instance, the acetonic and methanolic extracts of E. foetidum did not show antibacterial activity against Escherichia coli, Salmonella infantis, Listeria monocytogenes, Staphylococcus aureus, or Bacillus cereus [69], while the essential oil of E. maritimum showed a significant antibacterial activity against L. monocytogenes and E.coli due to its content of oxygenated sesquiterpenes [70], and the leaf hydromethanolic extract of E. maritimun showed antimicrobial activity against S. aureus, B. cereus, Salmonella enterica, Pseudomonas aeruginosa, P. fluorescens, P. marginalis, E. coli, and Erwinia carotovora subsp. carotovora [71]. Although many species of Eryngium have shown antimicrobial activity against Gram-positive and Gram-negative bacteria, some species of fungi and yeasts, and viruses, it has been suggested that multi-target antimicrobial experiments should be carried out using extracts of Eymgium spp. as antimicrobial agents in order to expand the knowledge about its antimicrobial potential [72].
Additionally, it is important to point out that the study of the anticholelithiasis and trypanocidal activity shown by E. carlinae and E. heterophyllum, respectively, could extend to other species of Eryngium; likewise, other activities such as anticlastogenic, anticarcinogenic, antihelmintic, and larvicidal, amongst others reported for E. foetidum could be evaluated [43].
The biological activities reported for E. carlinae, E. comosum, E. cymosum, E. heterophyllum, and E. longifolium are summarized in Table 5.

4. Phytochemistry

4.1. Screening, Detection, and Identification of Metabolites

Phytochemical characterization is valuable to reveal the presence and identity of secondary metabolites in a plant as well as a helpful tool in the search for bioactive compounds useful for the synthesis of new drugs and other products. The presence of various groups of metabolites has been described in plant extracts (ethanolic, aqueous, methanolic, and hexane) of aerial parts (stems, leaves, and inflorescences) qualitatively analyzed to determine the presence of flavonoids, tannins, terpenoids, and saponins. Additionally, more precise techniques such as GC-MS, nuclear magnetic resonance (NMR), and high-performance liquid chromatography (HPLC) are performed to detect, quantify, and identify metabolites in plant extracts. Regarding the genus Eryngium, investigations have been carried out on the phytochemical profile of some Eryngium spp. found in the central-western region of México, mainly E. carlinae, E. comosum, E. cymosum, and E. longifolium.
The most widely used methods to determine the general phytochemical composition of a plant are screening and the colorimetric techniques of ultraviolet-visible spectrophotometry (UV-Vis). Phytochemical screening involves a series of chemical reactions that make it possible to qualitatively detect the groups of secondary metabolites present in plant extracts [73,74,75,76,77,78]. For example, E. carlinae, one of the most common species used along the region delimited for this review, was qualitatively analyzed by Knauth et al. (2018) [11] to determine the main groups of metabolites in methanolic extract from the aerial part of the plant. The authors reported a strong presence of tannins and saponins, as well as a slight presence of flavonoids. Galindo-Hernández (2018) [58] evaluated the phytochemical profile of E. carlinae, confirming the presence of triterpenoids, sterols, tannins, coumarins, carboxyls, flavonoids, and carbohydrates. In the same way, Pérez-Reyes (2016) [13] performed phytochemical screening of an aqueous extract of E. comosum, reporting the presence of alkaloids, flavonoids (flavones and xanthones), triterpenoid saponins, reducing sugars, tannins derived from catechol, phenolic compounds, and benzoquinones; in addition, the author reported in vivo hypolipidemic activity.
Using UV-Vis, it is possible to determine compound concentration in a solution; this is a simple, reliable and low-cost analytical technique [79]. In this sense, Lemus-de la Cruz-Hurtado et al. (2023) [12] determined and quantified total phenolic compounds (TPC), total flavonoids (TF), and total terpenoids (TT) in aqueous extracts of aerial parts of E. carlinae. The authors reported concentrations of 0.0038 mg gallic acid equivalents per mL (mg GAE/mL) for TPC, 3.3032 mg quercetin equivalents per mL (mg QE/mL) for TF, and 0.0424 mg linalool equivalents per mL (mg LE/mL) for TT. Montes-Moreno (2017) [32] reported higher TPC values in aqueous infusions and decoctions of the same species. The authors evaluated by UV-Vis, up to 2.5 mg GAE/mL of TPC and, in the case of TF, the author reported less than 0.2 mg QE/mL. Similarly, Díaz-Alvarado et al. (2020) [61] reported the total phenolic content (4.33 mg GAE/g dry weight) and total saponins (62.2 mg/g dry weight) of the ethanolic extract of aerial parts of E. comosum. In the same sense, Díaz-Alvarado (2020) [62] determined the phytochemical profile of ethanolic extracts (50 and 70%) of aerial parts of E. comosum by UV-Vis, confirming the presence and abundance of phenolic compounds (up to 4.3 mg GAE/mL), TPC (up to 13.33 mg/g DW), and total saponins (up to 29.33 mg/g DW), with 70% ethanol being the best extractor.
Within the phenols group, phenolic acids (caffeic, rosmarinic, and chlorogenic) and their derivatives are the most abundant in the genus Eryngium; these compounds have been analyzed and identified by more precise techniques, such as chromatography [80]. Andrade-Cetto et al. (2021) [35] described the chromatographic profile of the ethanolic extract of aerial parts of E. longifolium analyzed by HPLC, the authors detected three signals at wavelengths of 254 and 320 nm with retention time (RT) between 6 and 13 min, which were identified as caffeic, chlorogenic, and rosmarinic acids by comparing them to their commercial standards, rosmarinic acid being the most abundant in the analyzed extract. Likewise, the authors reported other minor intensity peaks with RT between 16 and 24 min, which were attributed to isoflavone-type compounds and glycosylated flavonoids. In a similar work, Espinoza-Hernández et al. (2021) [8] analyzed aqueous extracts of E. cymosum by HPLC, finding caffeic, chlorogenic, and rosmarinic acids between 6 and 13 min of RT in the same UV-spectrum (320 nm). Additionally, the chromatographic profile of fractions isolated from aqueous, methanolic, and organic extracts of the same species has been analyzed by Romo-Pérez et al. (2022) [40], confirming that chlorogenic and rosmarinic phenolic acids were the most abundant in aqueous and methanolic extracts, while caffeic and protocatechuic acid were more abundant in the butanolic organic extract; in which, the authors reported for the first time the presence of the flavonoid kaempferol-3-O-(2,6-di-O-trans-ρ-coumaryl)-β-d-glucopyranoside in E. cymosum, associated directly with the hypoglycemic activity shown by the extracts. The identity and structure of the above compounds were corroborated by NMR.
Montes-Moreno (2017) [32] analyzed, by HPLC with a diode array detector coupled to mass spectrometry (HPLC-DAD-MS, detection wavelengths: 280, 320, and 370 nm) aqueous extract obtained by infusion and decoction at different concentrations (1 and 2%) and cooking times (5 and 10 min) of aerial parts of E. carlinae. The author reported the presence of hydroxybenzoic acids, mainly gallic acid (1300–2600 µg·mL−1, RT = 12.5 min), and to a lesser extent 4-hydroxybenzoic and protocatechuic acids. Likewise, the next hydroxycinnamic acids were found: chlorogenic acid (400–700 µg·mL−1, RT = 2.1 min), rosmarinic acid (40–58 µg·mL−1, RT = 25 min), and caffeic acid (12–21 µg·mL−1, RT = 16.3 min), while ellagic, p-coumaric, ferulic, and sinapic acids were present in the extract in smaller amounts; flavanols such as gallocatechin gallate (600–1600 µg·mL−1, RT = 20.4 min), catechin, epicatechin, and epigallocatechin gallate; and flavonols such as quercetin (89–179 µg·mL−1, RT = 25.5 min), rutin, and kaempferol were found in the extract as well. Additionally, the presence of flavanones and hydroxybenzaldehydes such as eriocitrin, naringenin, hesperidin, and vanillin were detected in minimal amounts. Using the same method, the author detected β-sitosterol, stigmastanol, sitosteryl-3-β-glucoside, and campesteryl-3-β-glucoside, phytosterols that could help control cholesterol levels in diseases such as hyperlipidemia, hypercholesterolemia, and atherosclerosis [81]. Likewise, the presence of glycosylated saponins involved in anti-inflammatory and antioxidant activities such as α-l-arabinopyranoside phytolaccagenic acid and β-d-glucopyranoside hederagenin was confirmed [82].
Pérez-Ramírez et al. (2016) [55] evaluated the phytochemical composition of the E. carlinae decoction by HPLC with a diode array detector (HPLC-DAD), as well as its participation in the modulating activity of renal dysfunction. Ellagic acid was the most abundant phenolic compound (38.3 areas under the curve, mA), followed by caffeic (20.3 mA), protocatechuic (11.9 mA), and p-hydroxybenzoic (9.8 mA) acids; likewise, flavonoids such as rutin (14.1 mA), catechin (12.1 mA), and epicatechin (11.7 mA) were detected and quantified. Regarding the presence of sterols, the phytosterol α-7-stigmasterol (18.7 mA) was detected as the most abundant, followed by β-sitosterol (11.1 mA), β-campesterol (8.7 mA), and stigmastanol (8.4 mA). Finally, the authors also detected and quantified two major saponins, campesteryl β-d-glucopyranoside (28.9 mA) and sitosteril β-d-glucopyranoside (20.1 mA).
The GC-MS technique is also a useful tool that allows the identification of volatile or semi-volatile compounds present in plant tissues. In this sense, mainly aromatic aldehydes, sesquiterpenes, and fatty acids have been detected in extracts of roots, stems, leaves, and inflorescences of different Eryngium spp. using GC-MS. Components such as 2-dodecenal, 2,4,6-trimethyl-benzaldehyde, d-elemene, a-bisabolol, α, and β-selinene, and fatty acids such as palmitic and stearic acid are common in a wide variety of Eryngium spp. found around the world, and their direct and synergistic participation in biological activities such as antiprotozoal [83], antibacterial [47,70,84,85], antifungal [47], antioxidant [84,86], and antidiabetic have been reported [86].
In relation to the species found in Mexico, Peña-Montes et al. (2016) [30] reported the presence and abundance of terpenes and sesquiterpenes identified in hexanic extracts of E. carlinae inflorescences. The major constituents identified were (Z)β-farnesene (38.79%), β-pinene (17.53%), calamenene (13.30%), and α-farnesene (10.38%). The authors attributed the ability to reduce lipid peroxidation and protein carbonylation to farnesene and pinene, in addition to the probable synergistic effect with other compounds present in the analyzed extract.
Likewise, Espino Garibay (2010) [59] found 21 terpenoid compounds in the ethanolic extract of leaves and inflorescences of E. carlinae. The main terpenoids that the author reported were borneol (367 mg/L), α-pinene (278 mg/L), myrcene (256 mg/L), caryophyllene (225 mg/L), and β-pinene (120 mg/L). In addition, the author reported differences in the presence and abundance through different phenological stages of the plant (before, during, and after flowering and fruiting), with the extracts of leaves and inflorescences during and after flowering being those that presented a higher content of metabolites; the isomers of pinene and farnesene were constant in both mentioned stages.
Noriega-Cisneros et al. (2019) [31] reported sesquiterpenes α-selinene (17.54%) and β-selinene (26.04%) as main components of ethanolic extract of aerial parts of E. carlinae, in addition to palmitic (14.43%) and stearic acid (14.53%), and others in proportions of less than 5%, such as humulene, stigmasterol, elemol, elemene, and α-cedrene; the authors conclude that these compounds, individually or synergistically, could be involved in the hypolipidemic and hypoglycemic response demonstrated in in vivo studies.
Likewise, some oligosaccharides and polyalcohols have been detected in extracts of leaves and stems of E. carlinae. Arana-Argáez et al. (2021) [87] described the presence of D-(−)-fructofuranose, D-(−)-fructopyranose, D-(−)-tagatofuranose, and sucrose, in addition to the polyalcohol 1,5-anhydro-d-sorbitol and cinnamic acid; the authors attributed the anti-inflammatory response, shown in vivo by the extract, to cinnamic acid.
The above information is summarized in Table 6.
According to the literature reviewed, the most studied species is E. carlinae, one of the most common in the central-western region of Mexico. However, research is needed on the phytochemical composition of the other species described in the region. In this sense, in the working group, a preliminary analysis of the phytochemical profile of several Eryngium species found in Michoacán and Jalisco, including E. carlinae, E. heterophyllum, E. nasturtiifolium, and E. beecheyanum, has been performed using HPTLC, and it has been possible to determine the presence of chlorogenic, rosmarinic, and caffeic acids in ethanolic extracts of inflorescences and stems.
An important aspect to consider when studying or proposing the application of Eryngium spp. bioactive compounds, is the extraction methods. Eryngium extracts contain various metabolites (e.g., tannins, phenolic acids, saponins, and terpenoids) associated with biological activities that support their use in traditional medicine or reveal new therapeutic potential, serving as targets for developing novel drugs. In order to extract bioactive compounds from plants, different methods are used [108]. Thus, multiple solvents chosen based on the polarity have been used to extract phytochemicals. While the ultrasound-assisted extraction (greater than 20 kHz) is used to disrupt plant cell walls, which helps improve the solvent’s ability to penetrate the cells and obtain a higher extraction yield, the use of a low operating temperature through processing allows the procurement of a high extract quality reducing the amount of solvent and energy used [109,110]. Unfortunately, there are still some concerns related to experimental repeatability and reproducibility [108]. Microwave-assisted extraction (MAE) has been used as an alternative to conventional techniques for the extraction of biocompounds due to its important advantages, among which are controllable and effective heating, faster energy transfer, the reduction of extraction time and use of solvents, higher selectivity, and enhanced yield [111,112,113]. Furthermore, new nonconventional technologies are emerging offering superior extraction efficiency in terms of cost, yield, extraction time, and/or selectivity [114]. In this sense, green synthesis of nanoparticles using plant extracts is a promising alternative to chemical methods [115,116,117].

4.2. Properties of Phytochemical Compounds in Species of the Genus Eryngium

Rosmarinic, caffeic, and chlorogenic acids are amongst the phenolic compounds reported in the genus Eryngium, regardless of the studied species. Rosmarinic acid (RA, molecular formula C18H16O8) named (R)-α-[[3-(3,4-dihydroxyphenyl)-1-oxo-2E-propenyl]oxy]-3,4-dihydroxy-enzenepropanoic acid is an ester of caffeic acid and (R)-(+)-3-(3,4-dihydroxyphenyl) lactic acid and originated from the amino acids L-phenylalanine and L-tyrosine, respectively (Hitl et al., 2020) [118]. RA has been proven to act as (1) an anticarcinogenic, as it inhibits the gene expression, the growth, and the proliferation of tumor cells [119]; (2) an antidiabetic, as it prevents hyperglycemia by increasing the insulin sensitivity index and reducing the levels of blood glucose [120]; (3) an antimicrobial, as it inhibits the growth of Methicillin-resistant Staphylococcus aureus (MRSA), E. coli, Pseudomonas spp., and L. monocytogenes, amongst others, and in addition to the formation of biofilm, it kills planktonic cells [121]; and (4) an antioxidant, as it inhibits the formation of free radicals, the generation of ROS, and lipid peroxidation [120]. In addition, RA has shown hepatoprotective, cardioprotective, antiallergic, antidepressant, anti-aging, nephroprotective, and anti-inflammatory activity [118,122]. Budzianowski et al. (2023) [93] investigated the cytotoxic effect of RA obtained from 50% ethanolic extracts of seedlings of different Eryngium species. The authors concluded that the rosmarinic acid-4′-O-β-glucopyranoside fraction did not show cytotoxic effects on cell lines involved in cancer development (MCF-7, MDA-MB-231, MCF-12A, HT-29, Caco-2, and OVCAR-3) with an IC50 average of 400–700 µM in 24–72 h. RA is used as a bioactive ingredient in supplements, and in its isolated or semipurified form, has not shown toxicity in humans. Jia et al. (2010) concluded that the administration of the RA isolated fraction from depsides salts of Salvia miltiorrhiza in only one dose (100–200 mg/kg), did not show toxic effects in humans [123].
Caffeic acid (CA, empirical formula C9H8O4, 3,4-dihydroxycinnamic acid) is a phenolic compound abundant in medicinal plants, with multiple biological activities, such as antioxidant, anti-inflammatory, anticarcinogenic, and neuroprotective [124]. Alam et al. (2022) [125] suggested that CA can intervene in the reduction of oxidative stress by blocking or preventing the formation of ROS molecules in the organism, having a potential antioxidant and anti-aging effect. The authors also determined that the CA could inhibit the formation and migration of tumor cells, decreasing the probability of metastasis in different types of cancer, suggesting that CA alone or in combination with other chemotherapeutic agents/drugs might be suggested to treat and manage cancer.
The chlorogenic acid (CGA) is directly involved in the mitochondrial function, the reduction and prevention of oxidative stress, apoptosis, inflammation, obesity, and diabetes. Hernandes et al. (2020) [124] investigated the cytotoxicity and genotoxicity of CA and CGA on human leukemic cell lines and determined that they showed neither cytotoxicity nor genotoxicity over the analyzed cells. However, CGA induces specific hypomethylation on Jukart cells, which can be beneficial against hematologic malignancies, since ADN altered methylation plays an important role in the pathogenic process. RA, CA, and CGA have been reported in different species of Eryngium, such as E. campestre, E. maritimum, E. plannum, E. creticum, and E. alpinum [75,76,77,91,126], and in some regional species addressed in this review, such as E. carlinae, E. longifolium, and E. cymosum.
Kaempferol (KAE, general empirical formula C15H10O6, 3,4′,5,7-Tetrahydroxyflavone) is part of the frequent appearance of flavonoids in different species of Eryngium. KAE is synthesized by condensation of 4-coumaroyl-CoA (C6-C3) with three molecules of malonyl-CoA (C6). KAE and its multiple glycosylated forms have been reported as agents with antitumor, anti-inflammatory, and antioxidant activity. Wang et al. (2018) [127] analyzed the antiproliferative activity of hepatic tumor cells of KAE and its glycosides (Kae-3-O-rha, Kae-7-O-glu and Kae-3-O-rut); the authors showed that KAE in its pure form has antiproliferative concentration-dependent activity (0–100 μM), whereas the glycosylated forms did not show such activity. Likewise, the authors reported that KAE has higher antioxidant and anti-inflammatory activity than its glycosylated forms. The safe use of KAE is being discussed since it could have antimutagenic and genotoxic activity; it also has prooxidant action since at high concentrations it can produce ROS [128]. Additionally, in order to overcome its poor bioavailability and improve its pharmacokinetics, the use of kaempferol nanosuspensions, solid dispersions, and complexes of polysaccharides and oligosaccharides has been developed [129].
The ellagic acid (EA, empirical formula C14H6O8, 4, 4′, 5, 5′, 6, 6′-hexahydrodiphenic acid 2, 6, 2′, 6′-dilactone), a flavonoid compound, has shown activity as a regulator of proinflammatory mediators and it also normalizes the lipid metabolism, in addition to having neuroprotective activity by starting several cell signaling pathways, preventing mitochondrial disfunction, and eliminating free radicals [130]. Likewise, catechin (CAT, empirical formula C22H18O10, catechin 5-O-gallate) and its derivatives, ((−)-CAT), ECAT ((−)-epicatechin), ECG ((−)-epicatechingallate), EGC ((−)-epigallocatechin), and EGCG ((−)-epigallocatechin gallate), have antioxidant and anti-aging activity, eliminating free radicals and delaying the extracellular matrix degradation induced by ultraviolet radiation (UV) and skin contamination, activating the collagen synthesis and promoting the inhibition of matrix metalloproteinase [131]. Beyond food and biomedical applications, EA and ellagitannins’ (ET) scientific relevance can be linked to advanced materials such as copolymers, chelating reagents, ion-exchange resins, and materials for electrochemical devices, among others [132].
On its part, the rutin and its derivatives (Rut, C27H30O16, quercetin 3-rutinoside) are flavonoids that have antioxidant, antimicrobial, anticarcinogenic, antidiabetic, antiallergic, antidepressant, antihypertensive, and other effects [133]. In the genus Eryngium, the flavonoids KAE, EA, CAT, and Rut have been reported in E. longifolium and E. carlinae. Quercetin and Rut could be applied in nutritional supplements and innovative complexes and formulations for pharmaceuticals [129].
Among the terpenic compounds in the Eryngium species of the central-western region of Mexico (described in E. carlinae, Table 6), are the borneol and α- and β-pinene, which are present in the essential oils of several aromatic plants and show activity on blood pressure regulation [134] in addition to showing cytogenetic, gastroprotective, anxiolytic, cytoprotective, anticonvulsant, and neuroprotective effects as well as their effects against H2O2-stimulated oxidative stress, pancreatitis, stress-stimulated hyperthermia, and pulpal pain [135]. Regarding caryophyllene (β-Caryophyllene (BCP)) and farnesene, they are natural sesquiterpenes that have several biological activities such as antimicrobial, anticarcinogenic, anti-inflammatory, antioxidant, anxiolytic-like, and local anesthetic effects. However, its volatility and poor water solubility limit its application in the pharmaceutical field [136].

4.3. Tocixity of Phytochemical Compounds in Species of the Genus Eryngium

Some findings about the toxicity of bioactive compounds detected in the Eryngium species reviewed in this work are described below. Knauth et al. (2018) [11] studied the cytotoxicity of extracts from various medicinal plants including E. carlinae using the Caco-2 enterocyte cell line. Most of the methanolic extracts from the tested plants showed low cytotoxicity.
Agiorgiti et al. (2018) [137] analyzed the cytotoxic and antitumor activities of five organotin complexes (1–5) with o-hydroxybenzoic or p-hydroxybenzoic acids in vitro and in vivo (in Wistar rats). All complexes exhibited strong cytotoxic activity against all cancer cell lines tested, so they could be used as potential chemotherapeutic agents. However, there is controversy about the safety of p-hydroxybenzoic acid in human health, as there is no evidence of its toxicity in humans [138]. In this sense, Downs et al. (2023) [139] evaluated the relation between accumulation of p-hydroxybenzoic acid and methyl, ethyl, propyl, and butyl esters and the incidence of both malignant and benign breast tumors in patients. The authors concluded that propyl and butyl paraben concentrations are higher on metastatic tissue compared to non-metastatic breast cancer tissue. Likewise, the authors suggested that factors that determine the formation of tumors in breast cancer such as age, menopause status, and family history of cancer could be controlled by changing to a paraben-free lifestyle, especially those present in cosmetics as preservatives, although more extensive studies must be carried out to confirm this association.
Budzianowski et al. (2023) [93] reported a similar effect for AR. The authors evaluated the effect of rosmarinic acid 4′-O-β-glucopyranoside (RAG4′) on five human carcinoma cell lines and one non-tumorigenic mammary epithelial cell line (MCF-12A). The highest cytotoxic activity of RAG4′ was observed in the Caco-2 cell line and RAG4′ did not show any effect on the non-tumorigenic cell line MCF-12A, indicating that it might be safe as a cosmetic and pharmaceutical ingredient.
D-mannitol found in E. carlinae was evaluated in a model of hypercholesterolemia in mice, and a chronic toxicity test was performed. The mice did not show evident signs of toxicity (e.g., lack of motor coordination, piloerection, or pupil dilation) after receiving doses of 100 and 500 mg/kg of the extract for 4 weeks [53].
Hameed et al. (2016) [140] evaluated the cytotoxic profiles of sinapic acid (SA) belonging to the phenylpropanoid family also present in Eryngium species. SA is assumed to be therapeutically beneficial and generally non-toxic. The authors tested a wide range of concentrations in Chinese hamster lung fibroblasts (V79) and human cervical carcinoma (HeLa) cells. Concentrations up to 500 μM and 2000 μM had no effect on the viability of V79 and HeLa cells, respectively, demonstrating that SA had no cytotoxic effects in two different cell lines except at very high concentrations. The dichotomous effect of SA on concentration-dependent cell viability has also been reported: at low SA concentrations cell viability was enhanced, while at high SA concentrations cell proliferation was almost completely inhibited [141,142].
Little is known about the genotoxic/antigenotoxic effects of SA, although there are several studies on the effects of different phenolic compounds [140,142]. The genotoxic effect and clastogenic potential of caffeic, cinnamic, and ferulic acids was examined by Maistro et al. (2011) [143]. The authors used in vitro comet and micronucleus (MN) assays in rat hepatoma tissue (HTC) cells at three different concentrations, for 24 h. No genotoxicity was observed by the comet assay, but clastogenic effects were found in HTC by the MN assay.
The subchronic toxicity of ellagic acid, also present in Eryngium species, was analyzed by Tasaki et al. (2008) [144] in F344 rats at five doses in a powdered basal diet for 90 days. No mortality or clinical signs related to the treatments were observed during the entire experimental period. The no-observed effect level (NOEL) was estimated to be 5% (3011 mg/kg b.w./day) for males and the no-observed-adverse-effect level (NOAEL) and NOEL in females were estimated to be 5% (3254 mg/kg b.w./day) and <1.25% (778 mg/kg b.w./day), respectively.
Marcarini et al. (2011) [145] studied the cytotoxic, apoptosis-inducing, genotoxic, and protective effects of rutin in HTC cells and demonstrated that at low concentrations (810 μM) the flavonoid decreased the viability and proliferation of cells after 72 h of treatment; however, at 24 h the authors observed induction of DNA damage, with genotoxic effects but without inducing apoptosis. The authors also observed a protective DNA effect by reducing damage induced by procarcinogenic agents such as benzo(a)pyrene suggesting an important biological activity for this compound, which can contribute to human health through diet. A subsequent report noted that rutin is considered a non-toxic and selective modulator of hypercholesterolemia [146].
It has been reported that the administration of β-sitosterol (BS) in rats does not cause genotoxicity or cytotoxicity [147]. A high level of BS concentrations in blood has been correlated with increased severity of heart disease in men who have previously suffered heart attacks [148]. An extensive toxicological study of BS showed a high LD50 in rats (>2 g/kg) [149]. A subsequent study showed that BS had no effect on the reproductive system [150]. It has also been shown that high BS exposure is related to alteration of the G5/8 transporters of the hepatic and intestinal ATP-binding cassette and promotes potential risks to the integrity of the blood–brain barrier in diabetic rats [151].
Several in vitro and in vivo studies have shown that CGA can protect cells against toxicities induced by chemicals of different classes (e.g., fungal/bacterial toxins, pharmaceuticals, and metals) by controlling the overproduction of nitric oxide and ROS and suppressed pro-apoptotic signaling [152].
Cos et al. (2001) [153] determined the kaempferol genotoxicity and attributed it mainly to its prooxidative activity. For their part, Ren et al. (2019) generated an antioxidant selectivity index (ASI), which is the maximum non-toxic dose divided by the IC50 value, which is used to assess the toxicity of flavonoids. If the ASI of the flavonoids is >100, they can be considered safe with a good antioxidant activity profile; however, further research is required to standardize it.
Spagnuolo et al., 2012 [154] carried out a study of quercetin toxicity in male F344/N rats fed 2 g per kg body weight per day of quercetin. The results showed severe chronic nephropathy, hyperplasia, and kidney tubular epithelium neoplasm after the treatment. In another study, it was reported that maternal intake of quercetin during pregnancy could increase the risk of mixed lineage leukemia (MLL) gene rearrangements, which is common in childhood leukemia, especially in the presence of compromised DNA repair [155]. However, Utesch et al. (2008) [156] demonstrated that orally administered quercetin (up to 2 g/kg body weight/day) did not induce unscheduled DNA synthesis in male or female rat hepatocytes, suggesting that quercetin was not genotoxic. Additionally, in the only phase I clinical trial of quercetin found, renal toxicity was detected without signs of nephritis or obstructive uropathy when a dose of 50 mg/kg (approximately 3.5 g/70 kg) was administered by intravenous infusion at three weeks or at weekly intervals [157].

5. Eryngium spp. Propagation Strategies

Given the wide variety of uses that different species of the genus Eryngium have, it is important to develop strategies to cultivate them, but some limitations should be faced, such as non-synchronized or uniform seed germination, low germination rates, lack of seed availability, and the need to reach higher sowing rates [158]. In this sense, due to the scarce reports found in literature about the propagation of the Eryngium species located in Mexico, particularly in the central-western region, some works in other species were reviewed as a starting point for future development in this matter.
Mozumder et al. (2010) [159] achieved an increase of E. foetidum germination rate, this species is of great importance in Latin America and other parts of the world. The authors reported that gibberellic acid (GA3, 1000 ppm) and kinetin (50 ppm) were effective to promote germination up to 28.54%. In a complementary work, Mozumder et al. (2017a) [158] evaluated different storage conditions, application of growth regulators, and soaking to increase E. foetidum seeds’ germination rate. Seeds kept at low temperature (3–5 °C) had a germination percentage of 18.4% and increased to 32.3% with 12 h of soaking and the addition of growth regulators. Mozumder et al. (2017 b) [160] evaluated seed germination and field performance of E. foetidum using soaking, growth regulators, and pesticide (0.2% copper oxychloride + 1000 ppm tetracycline) combinations. The maximum percentage of germination (74.7%) and early germination (12 days) was obtained using growth regulators (GA3 500 ppm and kinetin 50 ppm) with 96 h of soaking. Additionally, Mozumder et al. (2017c) [161] evaluated the growth and biomass production observing that the profitability increased, and the seed production cost decreased; the authors reported the highest values of the following variables: germination percentage, number of seedlings, number of harvestable plants/m2, number of leaves per plant, leaf width and biomass, at 30 and 60 days with the application of growth regulators.
On the other hand, Mozumder et al. (2020) [162] used three different levels of shades and two planting methods at two locations to cultivate E. foetidum. The maximum gross yield (4944.2 thousand TK/ha) and the net yield (4438.2 thousand TK/ha) were obtained with the shade of nylon netting in broadcast planting, which was better to produce leaves and obtain higher profits. These results coincided with those reported by Kuttan (2008) [163] who cultivated E. foetidum at three shade levels and four plant spacings. The maximum yield was obtained with a shade level of 75% (1411.04 g/plot size 120 × 150 cm). From storage studies it was concluded that at room temperature the leaves could be stored for a maximum of 5.2 days with a shade level of 75%, while under cold storage conditions and a shade level of 75% the leaves could be stored for a maximum of 109.65 days without any deterioration. The highest cost-benefit ratio (1.28) was obtained with a shade level of 75% and 15 cm × 15 cm spacing.
One of the few published works reporting the propagation of Eryngium species in Mexico is that of Carrera-Quirino and Colohua-Citlahua (2014) [164] who generated a reproduction strategy for Eryngium proteaeflorum in a nursery. The authors established recommendations for the collection of germplasm, characteristics of the mother plants, collection strategies to avoid damaging the habitat of the species and allow natural regeneration, as well as for propagation in seedbeds and transplant to planting bags.
Another propagation method reported for Eryngium spp. is the vegetative tissue culture (e.g., leaf, stem, or root segments) using culture media such as Murashige and Skoog (MS). The use of basal medium supplemented or not with sucrose, macro and micronutrients has been suggested [165]. Additionally, different explants and growth regulators, such as indole butyric acid (IBA) and indole acetic acid (IAA), have been used in various combinations [166], and the treatment with IBA 500 ppm and 500 ppm IAA using 7 to 10 cm shoots was the best to promote growth and obtain commercial seedlings. On the other hand, Ayuso et al. (2019) [167] reported the use of different growth inducers, sucrose, and salt concentrations for the propagation of E. viviparum, achieving a 96% survival rate in plants. In the same way, Martin (2006) [168], Nagananda et al. (2012) [169], and Jena et al. (2020) [170] reported protocols for the fastest and most successful clonal propagation of E. foetidum reaching an 85–90% plant survival rate using different explants, doses of inducers, and nitrogen sources.

6. Conclusions

The use of plants belonging to the genus Eryngium is widely distributed around the world, especially for medicinal and culinary purposes. The species of the genus whose presence and use in phytotherapy have been reported in central-western Mexico, especially in the states of Michoacán and Jalisco, are E. cymosum, E. longifolium, E. fluitans (or mexicanum), E. beecheyanum, E. carlinae, E. comosum, E. heterophyllum, and E. nasturtiifolium. The use of which has been recorded for the treatment of at least 27 ailments, since diverse benefits are attributed to them, such as antibacterial, anticrotalic, antispasmodic, antigonorrheal, anti-inflammatory, antioxidant, antipodagric, antitumor, carminative, digestive, hypoglycemic, hypocholesterolemic, and diuretic, among others; therefore, they are considered of great significance in the local populations’ health care. However, not all of them have been validated by scientific studies of the corresponding biological activities. Studies of biological activities have been reported only for five of them, and some of the metabolites that four of these Eryngium spp. contain, have been identified.
E. carlinae is the most studied species, and its hypoglycemic and hypolipidemic properties have been confirmed both in vitro and in vivo models. In addition, it has shown antioxidant, antimicrobial, and anti-inflammatory activities. Regarding its phytochemistry, more than 30 different compounds belonging to various groups of metabolites have been identified in E. carlinae extracts: hydroxybenzoic acids, hydroxycinnamic acids, phenolic acids, flavonoids, flavanols, flavanones, hydroxybenzaldehydes, phytosterols, saponins, terpenes, and sesquiterpenes. Much less information about the other species is published, and there are fewer toxicity studies. Therefore, more research is required on its phytochemistry and in vitro and in vivo biological activities. It is recommended to carry out in vivo tests with the extracts and metabolites of the Eryngium species that have been studied in vitro, and to later carry out clinical trials on inflammatory, infectious, and chronic diseases. An important aspect to investigate is the elucidation of the mechanisms of action associated with the beneficial effects reported. Additionally, the separation, isolation, and identification of the active metabolites or bioactive compounds must be performed since the greatest advances are made only in E. carlinae, followed by E. cymosum and E. longifolium. Having this information will allow the development of effective and safe treatments, drugs, and supplements, in addition to validate and promote the use of this plant resource for medicinal purposes in the communities that possess them. However, toxicity issues need to be addressed.
This literature review did not show any reports on the propagation or cultivation strategies of the Eryngium spp. addressed in this review, so it is necessary to start developing them to achieve the sustainable use of this important plant resource since they are obtained mainly by collection, causing populations to decline.

Author Contributions

Conceptualization, H.G.M.-V. and I.G.-R.; methodology, J.G.C.-V., H.G.M.-V. and I.G.-R.; investigation, J.G.C.-V., I.G.-R., M.V.A.-P. and H.G.M.-V.; resources H.G.M.-V. and I.G.-R.; writing—original draft preparation, J.G.C.-V., I.G.-R., M.V.A.-P. and H.G.M.-V.; writing—review and editing, J.G.C.-V. and H.G.M.-V.; project administration, H.G.M.-V. and I.G.-R.; funding acquisition, H.G.M.-V. and I.G.-R. All authors have read and agreed to the published version of the manuscript.

Funding

The first author thanks the Consejo Nacional de Ciencia y Tecnología (CONACYT) in Mexico for the economic support through national scholarship 558794. The rest of the authors appreciate the financial support from Instituto Politécnico Nacional through the projects SIP-20220666 and SIP-20220677, and the academic fellowships COFAA–IPN and EDI–IPN.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Sample Availability

Not applicable.

References

  1. Farnsworth, N.R.; Akerele, O.; Bingel, A.S.; Soejarto, D.D.; Zhengang, G. Medicinal Plants in Therapy. Bull. WHO 1985, 63, 965–981. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization. The Use of Essential Drugs Report of a WHO Expert Committee; World Health Organization Technical Report Series 685; WHO: Geneva, Switzerland, 1979; Volume 641, pp. 5–50. [Google Scholar]
  3. Akerele, O. Las Plantas Medicinales: Un Tesoro Que No Debemos Desperdiciar. Foro Mund. Salud 1993, 14, 390–395. [Google Scholar]
  4. Carreón-Sánchez, R.; Marroquín-Segura, R.; Mora-Guevara, J.L.A.; Valadez-Sánchez, C.S.; Flores-Cabrera, Y.; Flores-Pimentel, M.; Hernández-Abad, V.J. Estudio Del Extracto Etanólico de Eryngium heterophyllum (Hierba Del Sapo); Para Comprobar Su Actividad Hipoglucemiante y Anti-Inflamatoria. Rev. Mex. Cienc. Farm. 2013, 44, 41–45. [Google Scholar]
  5. Esquivel-Gutiérrez, E.R.; Noriega-Cisneros, R.; Bello-González, M.A.; Saavedra-Molina, A.; Salgado-Garciglia, R. Plantas Utilizadas En La Medicina Tradicional Mexicana Con Propiedades Antidiabéticas y Antihipertensivas. Biológicas 2012, 14, 45–52. [Google Scholar]
  6. García-Ruiz, I. Contribución al Conocimiento Del Género Eryngium (Apiaceae) En El Estado de Michoacán, México. Acta Bot. Mex. 2013, 118, 65–118. [Google Scholar] [CrossRef]
  7. Pérez-Muñoz, E.P.; Antunes-Ricardo, M.; Martínez-Ávila, M.; Guajardo-Flores, D. Eryngium Species as a Potential Ally for Treating Metabolic Syndrome and Diabetes. Front. Nutr. 2022, 9, 878306. [Google Scholar] [CrossRef]
  8. Espinoza-Hernández, F.; Andrade-Cetto, A.; Escandón-Rivera, S.; Mata-Torres, G.; Mata, R. Contribution of Fasting and Postprandial Glucose-Lowering Mechanisms to the Acute Hypoglycemic Effect of Traditionally Used Eryngium cymosum F. Delaroche. J. Ethnopharmacol. 2021, 279, 114339. [Google Scholar] [CrossRef]
  9. Espinoza-Hernández, F.A.; Andrade-Cetto, A. Chronic Antihyperglycemic Effect Exerted by Traditional Extracts of Three Mexican Medicinal Plants. Evid.-Based Complement. Altern. Med. 2022, 2022, 5970358. [Google Scholar] [CrossRef]
  10. Maldonado-Almanza, B.J. Aprovechamiento de Los Recursos Florísticos de La Sierra de Huautla Morelos, México. Master’s Thesis, Universidad Nacional Autónoma de México, Mexico City, Mexico, 1997. [Google Scholar]
  11. Knauth, P.; Acevedo-Hernández, G.J.; Cano, M.E.; Gutiérrez-Lomelí, M.; López, Z. In Vitro Bioactivity of Methanolic Extracts from Amphipterygium adstringens (Schltdl.) Schiede Ex Standl., Chenopodium ambrosioides, L., Cirsium mexicanum DC., Eryngium carlinae F. Delaroche, and Pithecellobium dulce (Roxb.) Benth. Used in Traditional Medi. Evid.-Based Complement. Altern. Med. 2018, 2018, 3610364. [Google Scholar] [CrossRef]
  12. Lemus-de la Cruz-Hurtado, J.; Trejo-Hurtado, M.; Landa-Moreno, C.; Peña-Montes, D.; Landeros-Páramo, J.L.; Cortés-Rojo, C.; Montoya-Pérez, R.; Rosas, G.; Saavedra-Molina, A. Antioxidant Effects of Silver Nanoparticles Obtained by Green Synthesis from the Aqueous Extract of Eryngium carlinae on the Brain Mitochondria of Streptozotocin-Induced Diabetic Rats. J. Bioenerg. Biomembr. 2023. preprint. [Google Scholar] [CrossRef]
  13. Pérez-Reyes, M.V. Determinación de la Probable Disminución de los Niveles de Colesterol y Triglicéridos en Rata Tratada con el Extracto Acuoso de Hierba de Sapo (Eryngium comosum Delar F). Bachelor’s Thesis, Investigación Curricular. Instituto Politécnico Nacional, Mexico City, Mexico, 2016. [Google Scholar]
  14. Mabberley, D.J. Mabberley’s Plant-Book, 4th ed.; Cambridge University Press: New York, NY, USA, 2017; ISBN 9781107115026. [Google Scholar]
  15. Wörz, A. A Taxonomic Index of the Species of Eryngium L. (Apiaceae: Saniculoideae). Stuttg. Beitr. Naturkd. A Biologie 1999, 48, 1–48. [Google Scholar]
  16. Wörz, A. On the Distribution and Relationships of the South-West Asian Species of Eryngium L. (Apiaceae-Saniculoideae). Turk. J. Bot. 2004, 28, 85–92. [Google Scholar]
  17. Wörz, A. A New Subgeneric Classification of the Genus Eryngium L. (Apiaceae, Saniculoideae). Bot. Jahrb. Syst. Pflanzengesch. Pflanzengeogr. 2005, 126, 253–259. [Google Scholar] [CrossRef]
  18. Calviño, C.I.; Martínez, S.G.; Downie, S.R. The Evolutionary History of Eryngium (Apiaceae, Saniculoideae): Rapid Radiations, Long Distance Dispersals, and Hybridizations. Mol. Phylogenet. Evol. 2008, 46, 1129–1150. [Google Scholar] [CrossRef]
  19. Wolff, H. Umbelliferae-Saniculoidae. In Das Pflanzenreich, Regni Vegetabilis Conspectus; Engelmann, V.v.W., Ed.; Auftrage der Königl. preuss; Akademie der Wissenschaften: Leipzig, Germany; Berlin, Germany, 1913. [Google Scholar]
  20. Mathias, M.E.; Constance, L. North American Flora; The New York Botanical Garden: New York, NY, USA, 1945; Volume 28B. [Google Scholar]
  21. Cházaro, B.M.; Lomelí, M.E.; Flores, H.M.; Ellembergm, R.S. Antología Botánica Del Occidente de México. Dep. Geogr. Ord. Territ. 2002, 1, 184. [Google Scholar]
  22. Mora-Navarro, M.R.; Vargas-Rodríguez, Y.L.; Hernández-Herrera, R.M. Algas del Occidente de México: Florística y Ecología; Universidad de Guadalajara: Guadalajara, Mexico, 2006. [Google Scholar]
  23. Astudillo-Vásquez, A.; Ortega-Delgado, M.L.; Hernández-Xolocotzi, E.; Estrada-Lugo, E.; Ramírez-Torres, L. Estudio Químico-Farmacológico de Eryngium heterophyllum (Yerba Del Sapo), Relacionado Con La Prevención de Cálculos Biliares. C. Bot. 1985, 7–19. [Google Scholar]
  24. Khoshbakht, K.; Hammer, K.; Pistrick, K. Eryngium caucasicum Trautv. Cultivated as a Vegetable in the Elburz Mountains (Northern Iran). Genet. Resour. Crop. Evol. 2006, 54, 445–448. [Google Scholar] [CrossRef]
  25. Paul, J.H.A.; Seaforth, C.E.; Tikasingh, T. Eryngium foetidum L.: A Review. Fitoterapia 2011, 82, 302–308. [Google Scholar] [CrossRef]
  26. Medbouhi, A.; Benbelaïd, F.; Djabou, N.; Beaufay, C.; Bendahou, M.; Quetin-Leclercq, J.; Tintaru, A.; Costa, J.; Muselli, A. Essential Oil of Algerian Eryngium campestre: Chemical Variability and Evaluation of Biological Activities. Molecules 2019, 24, 2575. [Google Scholar] [CrossRef]
  27. Redouan, F.Z.; Cheikh, Y.; Crisafulli, A.; Picone, R.M.; Merzouki, A. Ethnopharmacological Preparations Used for Digestive System Disorders by the Population in Talassemtane National Park (North of Morocco). Ethnobot. Res. Appl. 2022, 24, 1–25. [Google Scholar] [CrossRef]
  28. Esquivel-García, R.; Pérez-Calix, E.; Ochoa-Zarzosa, A.; García-Pérez, M.E. Ethnomedicinal Plants Used for the Treatment of Dermatological Affections on the Purépecha Plateau, Michoacán, Mexico. Acta Bot. Mex. 2018, 2018, 95–132. [Google Scholar] [CrossRef]
  29. Díaz, J.L. Usos de Las Plantas Medicinales de México; Instituto Mexicano para el Estudio de las Plantas Medicinales, IMEPLAM, A.C.: México City, Mexico, 1976. [Google Scholar]
  30. Peña-Montes, D.J.; Huerta-Cervantes, M.; Ríos-Silva, M.; Trujillo, X.; Huerta, M.; Noriega-Cisneros, R.; Salgado-Garciglia, R.; Saavedra-Molina, A. Protective Effect of the Hexanic Extract of Eryngium carlinae Inflorescences In Vitro, in Yeast, and in Streptozotocin-Induced Diabetic Male Rats. Antioxidants 2019, 8, 73. [Google Scholar] [CrossRef] [PubMed]
  31. Noriega-Cisneros, R.; Pena-Montes, D.J.; Huerta-Cervantes, M.; Torres-Martinez, R.; Huerta, M.; Manzo-Avalos, S.; Salgado-Garciglia, R.; Saavedra-Molina, A. Eryngium carlinae Ethanol Extract Corrects Lipid Abnormalities in Wistar Rats with Experimental Diabetes. J. Med. Food 2020, 23, 827–833. [Google Scholar] [CrossRef] [PubMed]
  32. Montes-Moreno, L.A.R. Efecto Del Extracto Acuoso de Eryngium carlinae (Hierba Del Sapo) Sobre Marcadores Bioquímicos de Enfermedades No Transmisibles. Master’s Thesis, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Santiago de Querétaro, Mexico, 2017. [Google Scholar]
  33. Atlas de las Plantas de la Medicina Tradicional Mexicana. Hierba Del Sapo. Available online: http://www.medicinatradicionalmexicana.unam.mx/apmtm/termino.php?l=3&t=eryngium-carlinae (accessed on 30 March 2023).
  34. Ávila-Uribe, M.M.; García-Zárate, S.N.; Sepúlveda-Barrera, A.S.; Godínez-Rodríguez, M.A. Plantas Medicinales En Dos Poblados Del Municipio De San Martín De Las Pirámides, Estado De México. Polibotanica 2016, 1, 215–245. [Google Scholar] [CrossRef]
  35. Andrade-Cetto, A.; Espinoza-Hernández, F.; Mata-Torres, G.; Escandón-Rivera, S. Hypoglycemic Effect of Two Mexican Medicinal Plants. Plants 2021, 10, 2060. [Google Scholar] [CrossRef]
  36. Andrade-Cetto, A. Ethnobotanical Study of the Medicinal Plants from Tlanchinol, Hidalgo, México. J. Ethnopharmacol. 2009, 122, 163–171. [Google Scholar] [CrossRef]
  37. Palá-Paúl, J. Contribución al Conocimiento de Los Aceites Esenciales Del Género “Eryngium” L., en la Península Ibérica. Ph.D. Thesis, Universidad Complutense de Madrid, Madrid, Spain, 2002. [Google Scholar]
  38. Morales, M.M.L. Interés Terapéutico de Eryngium (Cardo Corredor) En Oncología. Rev. Médica Homeopat. 2012, 5, 18–21. [Google Scholar]
  39. Lehman, R.L. Eryngium Nasturtiifolium; Texas A & M University Press: College Station, TX, USA, 2005. [Google Scholar]
  40. Romo-Pérez, A.; Escandón-Rivera, S.M.; Miranda, L.D.; Andrade-Cetto, A. Phytochemical Study of Eryngium cymosum F. Delaroche and the Inhibitory Capacity of Its Main Compounds on Two Glucose-Producing Pathway Enzymes. Plants 2022, 11, 992. [Google Scholar] [CrossRef]
  41. García-Hernández, K.Y.; Vibrans, H.; Rivas-Guevara, M.; Aguilar-Contreras, A. This Plant Treats That Illness? The Hot-Cold System and Therapeutic Procedures Mediate Medicinal Plant Use in San Miguel Tulancingo, Oaxaca, Mexico. J. Ethnopharmacol. 2015, 163, 12–30. [Google Scholar] [CrossRef]
  42. Fowler, M.W. Plants, Medicines and Man. J. Sci. Food Agric. 2006, 86, 1797–1804. [Google Scholar] [CrossRef]
  43. Rodrigues, T.L.M.; Silva, M.E.P.; Gurgel, E.S.C.; Oliveira, M.S.; Lucas, F.C.A. Eryngium foetidum L. (Apiaceae): A Literature Review of Traditional Uses, Chemical Composition, and Pharmacological Activities. Evid.-Based Complement. Altern. Med. 2022, 2022, 2896895. [Google Scholar] [CrossRef]
  44. Cortés-Fernández, I.; Sureda, A.; Adrover, M.; Caprioli, G.; Maggi, F.; Gil-Vives, L.; Capó, X. Antioxidant and Anti-Inflammatory Potential of Rhizome Aqueous Extract of Sea Holly (Eryngium maritimum L.) on Jurkat Cells. J. Ethnopharmacol. 2023, 305. [Google Scholar] [CrossRef]
  45. Price III, J.A. An in Vitro Evaluation of the Native American Ethnomedicinal Plant Eryngium Yuccifolium as a Treatment for Snakebite Envenomation. J. Intercult. Ethnopharmacol. 2016, 5, 219–225. [Google Scholar] [CrossRef]
  46. González, J.A.; Vallejo, J.R. The Scorpion in Spanish Folk Medicine: A Review of Traditional Remedies for Stings and Its Use as a Therapeutic Resource. J. Ethnopharmacol. 2013, 146, 62–74. [Google Scholar] [CrossRef]
  47. Kremer, D.; Končić, M.Z.; Kosalec, I.; Košir, I.J.; Potočnik, T.; Čerenak, A.; Srečec, S.; Dunkić, V.; Vuko, E. Phytochemical Traits and Biological Activity of Eryngium amethystinum and E. Alpinum (Apiaceae). Horticulturae 2021, 7, 364. [Google Scholar] [CrossRef]
  48. Sadiq, A.; Rashid, U.; Ahmad, S.; Zahoor, M.; AlAjmi, M.F.; Ullah, R.; Noman, O.M.; Ullah, F.; Ayaz, M.; Khan, I.; et al. Treating Hyperglycemia from Eryngium Caeruleum M. Bieb: In-Vitro α-Glucosidase, Antioxidant, in-Vivo Antidiabetic and Molecular Docking-Based Approaches. Front. Chem. 2020, 8, 558641. [Google Scholar] [CrossRef]
  49. Forouhandeh, H.; Rezaei Param, Z.; Molavi, O.; Asgharian, P.; Tarhriz, V. Evaluation of Anti-Proliferative Activity of Eryngium caucasicum on Melanoma Cancer Cells. BMC Complement. Med. Ther. 2022, 22, 134. [Google Scholar] [CrossRef]
  50. Noriega-Cisneros, R.; Ortiz-Vila, O.; Esquivel-Gutiérrez, E.; Clemente-Guerrero, M.; Manzo-Avalos, S.; Salgado-Garciglia, R.; Cortés-Rojo, C.; Boldogh, I.; Saavedra-Molina, A. Hypolipidemic Activity of Eryngium carlinae on Streptozotocin-Induced Diabetic Rats. Biochem. Res. Int. 2012, 2012, 603501. [Google Scholar] [CrossRef]
  51. Noriega-Cisneros, R. Evaluación de la Actividad Hipoglucémica, Hipolipidémica y Antioxidante de Eryngium carlinae en Ratas Diabéticas. Ph.D. Thesis, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico, 2013. [Google Scholar]
  52. García-Cerrillo, D.; Noriega-Cisneros, R.; Peña-Montes, D.; Huerta-Cervantes, M.; Ríos-Silva, M.; Salgado-Garciglia, R.; Montoya-Pérez, R.; Saavedra-Molina, A. Antioxidant Effects of Eryngium carlinae in Diabetic Rats. Asian J. Appl. Sci. 2018, 6, 308–314. [Google Scholar] [CrossRef]
  53. Castro-Torres, I.G.; De la O-Arciniega, M.; Naranjo-Rodríguez, E.B.; Castro-Torres, V.A.; Domínguez-Ortíz, M.Á.; Martínez-Vázquez, M. The Hypocholesterolemic Effects of Eryngium carlinae F. Delaroche Are Mediated by the Involvement of the Intestinal Transporters ABCG5 and ABCG. Evid.-Based Complement. Altern. Med. 2017, 2017, 3176232. [Google Scholar] [CrossRef]
  54. DrugBank Online Mannitol: Uses, Interactions, Mechanism of Action. Available online: https://go.drugbank.com/drugs/DB00742 (accessed on 23 March 2023).
  55. Pérez-Ramírez, I.F.; Enciso-Moreno, J.A.; Guevara-González, R.G.; Gallegos-Corona, M.A.; Loarca-Piña, G.; Reynoso-Camacho, R. Modulation of Renal Dysfunction by Smilax Cordifolia and Eryngium carlinae, and Their Effect on Kidney Proteome in Obese Rats. J. Funct. Foods 2016, 20, 545–555. [Google Scholar] [CrossRef]
  56. Valdivia-Mares, L.E. Capacidad Disolutiva Sobre Cálculos Biliares de Colesterol de Cuatro Especies Medicinales in Vitro. Master’s Thesis, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico, 2021. [Google Scholar]
  57. Pérez-Gutiérrez, R.M.; Vargas-Solís, R. γ-Lactone Isolated from Methanol Extract of the Leaves of Eryngium carlinae and Their Antispasmodic Effect on Rat Ileum. Bol. Latinoam. Caribe Plantas Med. Aromat. 2006, 5, 51–56. [Google Scholar]
  58. Galindo-Hernández, M.E. Obtención y Caracterización Fitoquímica de Extractos Vegetales y Su Evaluación Antimicótica para Su Aplicación en Odontopediatría. Master’s Thesis, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico, 2018. [Google Scholar]
  59. Espino-Garibay, D.F. Clasificación Molecular de Satureja Macrostema (Benth) Briq. y Eryngium carlinae Delar F. y Evaluación de La Actividad Antimicrobiana de Sus Terpenoides. Master’s Thesis, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico, 2010. [Google Scholar]
  60. Ronquillo de Jesus, E. Estudio de la Actividad Antioxidante y Toxicológica de Diferentes Extractos de Plantas Medicinales. Ph.D. Thesis, Tecnología Avanzada. Instituto Politécnico Nacional, Mexico City, Mexico, 2013. [Google Scholar]
  61. Díaz-Alvarado, T.; Mariezcurrena-Berasain, M.D.; Salem, A.Z.M.; Pinzón-Martínez, D.L. Antimicrobial and Antioxidant Activities of Two Medicinal Plants Cuphea aequipetala var. Hispida (Cav.) Koehne and Eryngium comosum Delaroche F against Bacteria Related to Equine Infections. J. Equine Vet. Sci. 2020, 94, 103269. [Google Scholar] [CrossRef] [PubMed]
  62. Díaz Alvarado, T. Análisis de los Compuestos Bioactivos, Actividad Antimicrobiana y Relación con el Territorio de Tres Plantas Medicinales de la Región Mazahua del Estado de México. Master’s Thesis, Universidad Autónoma del Estado de México, Toluca, Mexico, 2020. [Google Scholar]
  63. Navarrete, A.; Niño, D.; Reyes, B.; Sixtos, C.; Aguirre, E.; Estrada, E. On the Hypocholesteremic Effect of Eryngium heterophyllum. Fitoterapia 1990, 61, 182–184. [Google Scholar]
  64. Miranda-Velázquez, L.G. Actividad Hipocolesterolémica de Plantas de Uso Etnobotánico en México. Ph.D. Thesis, Ciencias, con Acentuación en Química de Productos Naturales. Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico, 2010. [Google Scholar]
  65. García-Gómez, Y.; Mendieta-Alcántara, M.R.; Mendieta-Zerón, H. Eryngium heterophyllum and Amphipterygium Adstringens Tea Effect on Triglyceride Levels: A Clinical Trial. Tradit. Integr. Med. 2019, 4, 22–27. [Google Scholar] [CrossRef]
  66. Molina-Garza, Z.J.; Bazaldúa-Rodríguez, A.F.; Quintanilla-Licea, R.; Galaviz-Silva, L. Anti-Trypanosoma Cruzi Activity of 10 Medicinal Plants Used in Northeast Mexico. Acta Trop. 2014, 136, 14–18. [Google Scholar] [CrossRef]
  67. Khani, S.; Abdollahi, M.; Asadi, Z.; Nazeri, M.; Nasiri, M.; Yusefi, H.; Moghadam, A.; Heidari, H. Hypoglycemic, Hepatoprotective, and Hypolipidemic Effects of Hydroalcoholic Extract of Eryngium Billardieri Root on Nicotinamide/Streptozotocin-Induced Type II Diabetic Rats. Res. Pharm. Sci. 2021, 16, 193–202. [Google Scholar] [CrossRef]
  68. Chandira, R.M.; Jaykar, B. Extraction, Pharmacological Evaluation and Formulation of Selected Medicinal Herbs for Antidiabetic Activity Pharmacological Evaluation and Formulation of Selected Medicinal Herbs for Antidiabetic Activity. Int. J. Pharm. Teach. Pract. 2013, 4, 458–482. [Google Scholar]
  69. Alzoreky, N.S.; Nakahara, K. Antibacterial Activity of Extracts from Some Edible Plants Commonly Consumed in Asia. Int. J. Food Microbiol. 2003, 80, 223–230. [Google Scholar] [CrossRef]
  70. Darriet, F.; Bendahou, M.; Desjobert, J.M.; Costa, J.; Muselli, A. Bicyclo[4.4.0]Decane Oxygenated Sesquiterpenes from Eryngium maritimum Essential Oil. Planta Med. 2012, 78, 386–389. [Google Scholar] [CrossRef]
  71. Meot-Duros, L.; Le Floch, G.; Magné, C. Radical Scavenging, Antioxidant and Antimicrobial Activities of Halophytic Species. J. Ethnopharmacol. 2008, 116, 258–262. [Google Scholar] [CrossRef]
  72. Erdem, S.A.; Nabavi, S.F.; Orhan, I.E.; Daglia, M.; Izadi, M.; Nabavi, S.M. Blessings in Disguise: A Review of Phytochemical Composition and Antimicrobial Activity of Plants Belonging to the Genus Eryngium. DARU J. Pharm. Sci. 2015, 23, 53. [Google Scholar] [CrossRef]
  73. Pant, D.R.; Pant, N.D.; Saru, D.B.; Yadav, U.N.; Khanal, D.P. Phytochemical Screening and Study of Antioxidant, Antimicrobial, Antidiabetic, Anti-Inflammatory and Analgesic Activities of Extracts from Stem Wood of Pterocarpus Marsupium Roxburgh. J. Intercult. Ethnopharmacol. 2017, 6, 170–176. [Google Scholar] [CrossRef]
  74. Farhan, H.; Malli, F.; Rammal, H.; Hijazi, A.; Bassal, A.; Ajouz, N.; Badran, B. Phytochemical Screening and Antioxidant Activity of Lebanese Eryngium creticum L. Asian Pac. J. Trop. Biomed. 2012, 2, S1217–S1220. [Google Scholar] [CrossRef]
  75. Kikowska, M.; Chanaj-Kaczmarek, J.; Derda, M.; Budzianowska, A.; Thiem, B.; Ekiert, H.; Szopa, A. The Evaluation of Phenolic Acids and Flavonoids Content and Antiprotozoal Activity of Eryngium Species Biomass Produced by Biotechnological Methods. Molecules 2022, 27, 363. [Google Scholar] [CrossRef]
  76. Thiem, B.; Goślińska, O.; Kikowska, M.; Budzianowski, J. Antimicrobial Activity of Three Eryngium L. Species (Apiaceae). Herba Pol. 2010, 56, 53–58. [Google Scholar]
  77. Kikowska, M.; Kruszka, D.; Derda, M.; Hadas, E.; Thiem, B. Phytochemical Screening and Acanthamoebic Activity of Shoots from in Vitro Cultures and in Vivo Plants of Eryngium alpinum L.—The Endangered and Protected Species. Molecules 2020, 25, 1416. [Google Scholar] [CrossRef]
  78. Mejri, H.; Tir, M.; Feriani, A.; Ghazouani, L.; Salah Allagui, M.; Saidani-Tounsi, M. Does Eryngium maritimum Seeds Extract Protect against CCl4 and Cisplatin Induced Toxicity in Rats: Preliminary Phytochemical Screening and Assessment of Its in Vitro and in Vivo Antioxidant Activity and Antifibrotic Effect. J. Funct. Foods 2017, 37, 363–372. [Google Scholar] [CrossRef]
  79. Weber, F.; Passon, M. Characterization and Quantification of Polyphenols in Fruits. In Polyphenols in Plants; Elsevier: Amsterdam, The Netherlands, 2019; pp. 111–121. [Google Scholar]
  80. Meyer, V.R. Practical High-Performance Liquid Chromatography, 5th ed.; John Wiley and Sons: St. Gallen, Switzerland, 2013; ISBN 9780470682180. [Google Scholar]
  81. López-Luengo, M.T. Fitoesteroles y Fitoestanoles. Offarm 2005, 24, 90–94. [Google Scholar]
  82. Juang, Y.-P.; Liang, P.-H. Biological and Pharmacological Effects of Synthetic Saponins. Molecules 2020, 25, 4974. [Google Scholar] [CrossRef]
  83. Rojas-Silva, P.; Graziose, R.; Vesely, B.; Poulev, A.; Mbeunkui, F.; Grace, M.H.; Kyle, D.E.; Lila, M.A.; Raskin, I. Leishmanicidal Activity of a Daucane Sesquiterpene Isolated from Eryngium foetidum. John M. Pezzuto Pharm. Biol. 2014, 52, 398–401. [Google Scholar] [CrossRef] [PubMed]
  84. Merghache, D.; Boucherit-Otmani, Z.; Merghache, S.; Chikhi, I.; Selles, C.; Boucherit, K. Chemical Composition, Antibacterial, Antifungal and Antioxidant Activities of Algerian Eryngium Tricuspidatum L. Essential Oil. Nat. Prod. Res. 2014, 28, 795–807. [Google Scholar] [CrossRef] [PubMed]
  85. Landoulsi, A.; Roumy, V.; Duhal, N.; Skhiri, F.H.; Ere, R.; Sahpaz, S.; Neut, C.; Benhamida, J.; Hennebelle, T. Chemical Composition and Antimicrobial Activity of the Essential Oil from Aerial Parts and Roots of Eryngium Barrelieri BOISS. and Eryngium Glomeratum LAM. from Tunisia. Chem. Biodivers. 2016, 13, 1720–1729. [Google Scholar] [CrossRef] [PubMed]
  86. Paw, M.; Gogoi, R.; Sarma, N.; Saikia, S.; Kumar Chanda, S.; Lekhak, H.; Lal, M. Antimicrobial, Antioxidant, Antidiabetic Study of Leaf Essential Oil of Eryngium foetidum L. Along with the Chemical Profiling Collected from North East India. J. Essent. 2022, 25, 1229–1241. [Google Scholar] [CrossRef]
  87. Arana-Argáez, V.; Alonso-Castro, A.J.; Yáñez-Barrientos, E.; Euan-Canto, A.; Torres-Romero, J.C.; Isiordia-Espinoza, M.A.; Brennan-Bourdon, L.M.; Juárez-Vázquez, M.d.C.; González-Ibarra, A.A. In Vitro and in Vivo Anti-Inflammatory Effects of an Ethanol Extract from the Aerial Parts of Eryngium carlinae F. Delaroche (Apiaceae). J. Ethnopharmacol. 2021, 266, 113406. [Google Scholar] [CrossRef]
  88. Nejati, M.; Masoudi, S.; Dastan, D.; Masnabadi, N. Phytochemical Analysis and Antibacterial Activity of Eryngium Boiss. & Hausskn. J. Chil. Chem. Soc. 2021, 66, 5230–5236. [Google Scholar]
  89. Singh, S.; Singh, D.R.; Banu, S.; Salim, K.M. Determination of Bioactives and Antioxidant Activity in Eryngium foetidum L.: A Traditional Culinary and Medicinal Herb. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2013, 83, 453–460. [Google Scholar] [CrossRef]
  90. Rjeibi, I.; Saad, A.B.; Ncib, S.; Souid, S. Phenolic Composition and Antioxidant Properties of Eryngium maritimum (Sea Holly). J. Coast. 2017, 5, 212–215. [Google Scholar] [CrossRef]
  91. Kikowska, M.; Budzianowski, J.; Krawczyk, A.; Thiem, B. Accumulation of Rosmarinic, Chlorogenic and Caffeic Acids in in Vitro Cultures of Eryngium planum L. Acta Physiol. Plant. 2012, 34, 2425–2433. [Google Scholar] [CrossRef]
  92. Kikowska, M.; Thiem, B.; Szopa, A.; Ekiert, H. Accumulation of Valuable Secondary Metabolites: Phenolic Acids and Flavonoids in Different in Vitro Systems of Shoot Cultures of the Endangered Plant Species—Eryngium alpinum L. Plant. Cell Tissue Organ. Cult. 2020, 141, 381–391. [Google Scholar] [CrossRef]
  93. Budzianowski, J.; Romaniuk-Drapała, A.; Kikowska, M.; Budzianowska, A.; Thiem, B.; Lisiak, N.; Rubiś, B.; Jacczak, B.; Kosmalska, I.; Totoń, E. Rosmarinic Acid 4′-O-β-Glucoside—A Compound with Prospective Medicinal and Cosmetological Applications—Its Isolation from Callus and Root Cultures of Eryngium Species and Biological Activity. Ind. Crops Prod. 2023, 193, 116138. [Google Scholar] [CrossRef]
  94. Dalar, A.; Türker, M.; Zabaras, D.; Konczak, I. Phenolic Composition, Antioxidant and Enzyme Inhibitory Activities of Eryngium bornmuelleri Leaf. Plant. Foods Hum. Nutr. 2014, 69, 30–36. [Google Scholar] [CrossRef]
  95. García, M.D.; Sáenz, M.T.; Gómez, M.A.; Fernández, M.A. Topical Antiinflammatory Activity of Phytosterols Isolated from Eryngium foetidum on Chronic and Acute Inflammation Models. Phytother. Res. 1999, 13, 78–80. [Google Scholar] [CrossRef]
  96. Hatami, M.; Karimi, M.; Aghaee, A.; Bovand, F.; Ghorbanpour, M. Morphological Diversity, Phenolic Acids, and Antioxidant Properties in Eryngo (Eryngium caucasicum Trautv): Selection of Superior Populations for Agri-Food Industry. Food Sci. Nutr. 2022, 10, 3905–3919. [Google Scholar] [CrossRef]
  97. Palá-Paúl, J.; Pérez-Alonso, M.J.; Velasco-Negueruela, A.; Varadé, J.; Villa, A.M.; Sanz, J.; Brophy, J.J. Analysis of the Essential Oil Composition from the Different Parts of Eryngium glaciale Boiss. from Spain. J. Chromatogr. 2005, 1094, 179–182. [Google Scholar] [CrossRef]
  98. Flamini, G.; Tebano, M.; Cioni, P.L. Composition of the Essential Oils from Leafy Parts of the Shoots, Flowers and Fruits of Eryngium amethystinum from Amiata Mount (Tuscany, Italy). Food Chem. 2008, 107, 671–674. [Google Scholar] [CrossRef]
  99. Celik, A.; Aydınlık, N.; Arslan, I. Phytochemical Constituents and Inhibitory Activity towards Methicillin-Resistant Staphylococcus aureus Strains of Eryngium Species (Apiaceae). Chem. Biodivers. 2011, 8, 454–459. [Google Scholar] [CrossRef]
  100. Silva Fernandes, L.; Ferreira Garcia da Costa, Y.; Eunice de Bessa, M.; Lucia Pires Ferreira, A.; Otávio do Amaral Corrêa, J.; Del-Vechio Vieira, G.; Vieira de Sousa, O.; Lúcia Santos de Matos Araújo, A.; Castilho, P.C.; Silvana Alves, M.; et al. Metabolic Profiling and Antibacterial Activity of Eryngium Pristis Cham. & Schltdl.—Prospecting for Its Use in the Treatment of Bacterial Infections. Arch. Pharm. Pharm. Sci. 2021, 5, 020–028. [Google Scholar] [CrossRef]
  101. Kikowska, M.; Kalemba, D.; Dlugaszewska, J.; Thiem, B. Chemical Composition of Essential Oils from Rare and Endangered Species—Eryngium maritimum L. and E. Alpinum L. Plants 2020, 9, 417. [Google Scholar] [CrossRef]
  102. Palá-Paúl, J.; Copeland, L.M.; Brophy, J.J.; Goldsack, R.J. Essential Oil Composition of Eryngium rosulatum P.W. Michael Ined.: A New Undescribed Species from Eastern Australia. Biochem. Syst. Ecol. 2006, 34, 796–801. [Google Scholar] [CrossRef]
  103. Nacef, S.; Jannet, H.B.; Hamza, M.A.; Mighri, Z. Contribution to the Phytochemical Investigation of the Plant Eryngium dichotomum Desf. (Apiaceae) from Tunisia. J. Soc. Chim. Tunisie 2008, 10, 141–148. [Google Scholar]
  104. Cádiz-Gurrea, M.d.l.L.; Fernández-Arroyo, S.; Joven, J.; Segura-Carretero, A. Comprehensive Characterization by UHPLC-ESI-Q-TOF-MS from an Eryngium bourgatii Extract and Their Antioxidant and Anti-Inflammatory Activities. Food Res. Int. 2013, 50, 197–204. [Google Scholar] [CrossRef]
  105. Dunkić, V.; Vuko, E.; Bezić, N.; Kremer, D.; Ruščić, M. Composition and Antiviral Activity of the Essential Oils of Eryngium alpinum and E. amethystinum. Chem. Biodivers. 2013, 10, 1894–1902. [Google Scholar] [CrossRef] [PubMed]
  106. Mohammadhosseini, M.; Mahdavi, B.; Akhlaghi, H. Characterization and Chemical Composition of the Volatile Oils from Aerial Parts of Eryngium bungei Bioss. (Apiaceae) by Using Traditional Hydrodistillation, Microwave Assisted Hydrodistillation and Head Space Solid Phase Microextraction Methods Prior to GC and GC/MS Analyses: A Comparative Approach. J. Essent. 2013, 16, 613–623. [Google Scholar] [CrossRef]
  107. Dawilai, S.; Muangnoi, C.; Praengamthanachoti, P.; Tuntipopipat, S. Anti-Inflammatory Activity of Bioaccessible Fraction from Eryngium foetidum Leaves. BioMed Res. Int. 2013, 2013, 958567. [Google Scholar] [CrossRef]
  108. Altemimi, A.; Lakhssassi, N.; Baharlouei, A.; Watson, D.G.; Lightfoot, D.A. Phytochemicals: Extraction, Isolation, and Identification of Bioactive Compounds from Plant Extracts. Plants 2017, 6, 42. [Google Scholar] [CrossRef]
  109. Williams, O.J.; Raghavan, G.S.V.; Orsat, V.; Dai, J. Microwave-Assisted Extraction of Capsaicinoids from Capsicum Fruit. J. Food Biochem. 2004, 28, 113–122. [Google Scholar] [CrossRef]
  110. Garcia-Salas, P.; Morales-Soto, A.; Segura-Carretero, A.; Fernández-Gutiérrez, A. Phenolic-Compound-Extraction Systems for Fruit and Vegetable Samples. Molecules 2010, 15, 8813–8826. [Google Scholar] [CrossRef]
  111. Routray, W.; Orsat, V. Microwave-Assisted Extraction of Flavonoids: A Review. Food Bioprocess Technol. 2012, 5, 409–424. [Google Scholar] [CrossRef]
  112. Tzanova, M.; Atanasov, V.; Yaneva, Z.; Ivanova, D.; Dinev, T. Selectivity of Current Extraction Techniques for Flavonoids from Plant Materials. Processes 2020, 8, 1222. [Google Scholar] [CrossRef]
  113. Coelho, J.P.; Robalo, M.P.; Boyadzhieva, S.; Stateva, R.P. Microwave-Assisted Extraction of Phenolic Compounds from Spent Coffee Grounds. Process Optimization Applying Design of Experiments. Molecules 2021, 26, 7320. [Google Scholar] [CrossRef]
  114. Zhang, Y.; Cai, P.; Cheng, G.; Zhang, Y. A Brief Review of Phenolic Compounds Identified from Plants: Their Extraction, Analysis, and Biological Activity. Nat. Prod. Commun. 2022, 17, 1934578X211069721. [Google Scholar] [CrossRef]
  115. Ajitha, B.; Reddy, Y.A.K.; Lee, Y.; Kim, M.J.; Ahn, C.W. Biomimetic Synthesis of Silver Nanoparticles Using Syzygium aromaticum (Clove) Extract: Catalytic and Antimicrobial Effects. Appl. Organomet. 2019, 33, e4867. [Google Scholar] [CrossRef]
  116. Vanlalveni, C.; Lallianrawna, S.; Biswas, A.; Selvaraj, M.; Changmai, B.; Rokhum, S.L. Green Synthesis of Silver Nanoparticles Using Plant Extracts and Their Antimicrobial Activities: A Review of Recent Literature. RSC Adv. 2021, 11, 2804–2837. [Google Scholar] [CrossRef]
  117. Begum, S.; Ahmaruzzaman, M.; Adhikari, P.P. Ecofriendly Bio-Synthetic Route to Synthesize ZnO Nanoparticles Using Eryngium foetidum L. and Their Activity against Pathogenic Bacteria. Mater. Lett. 2018, 228, 37–41. [Google Scholar] [CrossRef]
  118. Hitl, M.; Kladar, N.; Gavarić, N.; Božin, B. Rosmarinic Acid-Human Pharmacokinetics and Health Benefits. Planta Med. 2020, 87, 273–282. [Google Scholar] [CrossRef]
  119. Lin, D.; Jin, B.; Liu, J.; Gao, D.; Xu, Y.; He, L.; Zang, Y.; Li, N. Detailed Studies on the Anticancer Action of Rosmarinic Acid in Human Hep-G2 Liver Carcinoma Cells: Evaluating Its Effects on Cellular Apoptosis, Caspase Activation and Suppression of Cell Migration and Invasion. J. BUON 2020, 25, 1383–1389. [Google Scholar]
  120. Cardullo, N.; Catinella, G.; Floresta, G.; Muccilli, V.; Rosselli, S.; Rescifina, A.; Bruno, M.; Tringali, C. Synthesis of Rosmarinic Acid Amides as Antioxidative and Hypoglycemic Agents. J. Nat. Prod. 2019, 82, 573–582. [Google Scholar] [CrossRef]
  121. Ivanov, M.; Kostić, M.; Stojković, D.; Soković, M. Rosmarinic Acid–Modes of Antimicrobial and Antibiofilm Activities of a Common Plant Polyphenol. S. Afr. J. Bot. 2022, 146, 521–527. [Google Scholar] [CrossRef]
  122. Nadeem, M.; Imran, M.; Gondal, T.A.; Imran, A.; Shahbaz, M.; Amir, R.M.; Sajid, M.W.; Qaisrani, T.B.; Atif, M.; Hussain, G.; et al. Therapeutic Potential of Rosmarinic Acid: A Comprehensive Review. Appl. Sci. 2019, 2019, 3139. [Google Scholar] [CrossRef]
  123. Jia, J.Y.; Lu, Y.L.; Li, X.C.; Liu, G.Y.; Li, S.J.; Liu, Y.; Liu, Y.M.; Yu, C.; Wang, Y.P. Pharmacokinetics of Depside Salts from Salvia Miltiorrhiza in Healthy Chinese Volunteers: A Randomized, Open-Label, Single-Dose Study. Curr. Ther. Res. Clin. Exp. 2010, 71, 260–271. [Google Scholar] [CrossRef] [PubMed]
  124. Hernandes, L.C.; Thomazela Machado, A.R.; Tuttis, K.; Ribeiro, D.L.; Ferro Aissa, A.; Pícoli Dévoz, P.; Greggi Antunes, L.M. Caffeic Acid and Chlorogenic Acid Cytotoxicity, Genotoxicity and Impact on Global DNA Methylation in Human Leukemic Cell Lines. Genet. Mol. Biol. 2020, 43, 20190347. [Google Scholar] [CrossRef] [PubMed]
  125. Alam, M.; Ahmed, S.; Elasbali, A.M.; Adnan, M.; Alam, S.; Hassan, M.I.; Pasupuleti, V.R. Therapeutic Implications of Caffeic Acid in Cancer and Neurological Diseases. Front. Oncol. 2022, 12, 860508. [Google Scholar] [CrossRef] [PubMed]
  126. Elsbaey, M.; Ibrahim, M.A.A.; Shawky, A.M.; Miyamoto, T. Eryngium creticum L.: Chemical Characterization, SARS-CoV-2 Inhibitory Activity, and In Silico Study. ACS Omega 2022, 7, 22725–22734. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, J.; Fang, X.; Ge, L.; Cao, F.; Zhao, L.; Wang, Z.; Xiao, W. Antitumor, Antioxidant and Anti-Inflammatory Activities of Kaempferol and Its Corresponding Glycosides and the Enzymatic Preparation of Kaempferol. PLoS ONE 2018, 13, e0197563. [Google Scholar] [CrossRef] [PubMed]
  128. Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A Review on the Dietary Flavonoid Kaempferol. Med. Chem. Rev. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  129. Gervasi, T.; Calderaro, A.; Barreca, D.; Tellone, E.; Trombetta, D.; Ficarra, S.; Smeriglio, A.; Mandalari, G.; Gattuso, G. Biotechnological Applications and Health-Promoting Properties of Flavonols: An Updated View. Int. J. Mol. Sci. 2022, 23, 1. [Google Scholar] [CrossRef]
  130. Gupta, A.; Singh, A.K.; Kumar, R.; Jamieson, S.; Pandey, A.K.; Bishayee, A. Neuroprotective Potential of Ellagic Acid: A Critical Review. Adv. Nutr. 2021, 12, 1211–1238. [Google Scholar] [CrossRef]
  131. Bae, J.; Kim, N.; Shin, Y.; Kim, S.-Y.; Kim, Y.-J. Activity of Catechins and Their Applications. Biomed. Derm. 2020, 4, 8. [Google Scholar] [CrossRef]
  132. Evtyugin, D.D.; Magina, S.; Evtuguin, D.V. Recent Advances in the Production and Applications of Ellagic Acid and Its Derivatives. A Review. Molecules 2020, 25, 2745. [Google Scholar] [CrossRef]
  133. Gullón, B.; Lú-Chau, T.A.; Moreira, M.T.; Lema, J.M.; Eibes, G. Rutin: A Review on Extraction, Identification and Purification Methods, Biological Activities and Approaches to Enhance Its Bioavailability. Trends Food Sci. Technol. 2017, 67, 220–235. [Google Scholar] [CrossRef]
  134. Luz, M.S.; Gadelha, D.D.A.; Andrade, K.J.S.; Travassos, R.A.; Ribeiro, J.D.; Carvalho-Galvão, A.; Cruz, J.C.; Balarini, C.M.; Braga, V.A.; França-Falcão, M.S. Borneol Reduces Sympathetic Vasomotor Hyperactivity and Restores Depressed Baroreflex Sensitivity in Rats with Renovascular Hypertension. Hypertens. Res. 2022, 45, 802–813. [Google Scholar] [CrossRef]
  135. Salehi, B.; Upadhyay, S.; Orhan, I.E.; Jugran, A.K.; Jayaweera, S.L.D.; Dias, D.A.; Sharopov, F.; Taheri, Y.; Martins, N.; Baghalpour, N.; et al. Therapeutic Potential of α- and β-Pinene: A Miracle Gift of Nature. Biomolecules 2019, 9, 738. [Google Scholar] [CrossRef]
  136. Gupta, S.; Variyar, P.S. Nanoencapsulation of Essential Oils for Sustained Release: Application as Therapeutics and Antimicrobials. In Encapsulations; Elsevier: Amsterdam, The Netherlands, 2016; pp. 641–672. [Google Scholar]
  137. Agiorgiti, M.S.; Evangelou, A.; Vezyraki, P.; Hadjikakou, S.K.; Kalfakakou, V.; Tsanaktsidis, I.; Batistatou, A.; Zelovitis, J.; Simos, Y.V.; Ragos, V.; et al. Cytotoxic Effect, Antitumour Activity and Toxicity of Organotin Derivatives with Ortho- or Para-Hydroxy-Benzoic Acids. Med. Chem. Res. 2018, 27, 1122–1130. [Google Scholar] [CrossRef]
  138. Petric, Z.; Ruzic, J.; Zuntar, I. The Controversies of Parabens-an Overview Nowadays. Acta Pharm. 2021, 17–32. [Google Scholar] [CrossRef]
  139. Downs, C.A.; Amin, M.M.; Tabatabaeian, M.; Chavoshani, A.; Amjadi, E.; Afshari, A.; Kelishadi, R. Parabens Preferentially Accumulate in Metastatic Breast Tumors Compared to Benign Breast Tumors and the Association of Breast Cancer Risk Factors with Paraben Accumulation. Environ. Adv. 2023, 11, 100325. [Google Scholar] [CrossRef]
  140. Hameed, H.; Aydin, S.; Başaran, N. Sinapic Acid: Is It Safe for Humans? J. Pharm. Sci. 2016, 41, 39–49. [Google Scholar]
  141. Zheng, L.F.; Dai, F.; Zhou, B.; Yang, L.; Liu, Z.L. Prooxidant Activity of Hydroxycinnamic Acids on DNA Damage in the Presence of Cu(II) Ions: Mechanism and Structure-Activity Relationship. Food Chem. Toxicol. 2008, 46, 149–156. [Google Scholar] [CrossRef]
  142. Pandi, A.; Kalappan, V.M. Pharmacological and Therapeutic Applications of Sinapic Acid—An Updated Review. Mol. Biol. Rep. 2021, 48, 3733–3745. [Google Scholar] [CrossRef]
  143. Maistro, E.L.; Angeli, J.P.; Andrade, S.F.; Mantovani, M.S. In Vitro Genotoxicity Assessment of Caffeic, Cinnamic and Ferulic Acids. Genet. Mol. Res. 2011, 10, 1130–1140. [Google Scholar] [CrossRef]
  144. Tasaki, M.; Umemura, T.; Maeda, M.; Ishii, Y.; Okamura, T.; Inoue, T.; Kuroiwa, Y.; Hirose, M.; Nishikawa, A. Safety Assessment of Ellagic Acid, a Food Additive, in a Subchronic Toxicity Study Using F344 Rats. Food Chem. Toxicol. 2008, 46, 1119–1124. [Google Scholar] [CrossRef] [PubMed]
  145. Marcarini, J.C.; Ferreira Tsuboy, M.S.; Cabral Luiz, R.; Regina Ribeiro, L.; Hoffmann-Campo, C.B.; Ségio Mantovani, M. Investigation of Cytotoxic, Apoptosis-Inducing, Genotoxic and Protective Effects of the Flavonoid Rutin in HTC Hepatic Cells. Exp. Toxicol. Pathol. 2011, 63, 459–465. [Google Scholar] [CrossRef] [PubMed]
  146. Ganeshpurkar, A.; Saluja, A.K. The Pharmacological Potential of Rutin. Saudi Pharm. J. 2017, 25, 149–164. [Google Scholar] [CrossRef] [PubMed]
  147. Rashed, K. Beta-Sitosterol Medicinal Properties: A Review Article. Int. J. Sci. Innov. Technol. 2020, 9, 208–212. [Google Scholar]
  148. Assmann, G.; Cullen, P.; Erbey, J.; Ramey, D.R.; Kannenberg, F.; Schulte, H. Plasma Sitosterol Elevations Are Associated with an Increased Incidence of Coronary Events in Men: Results of a Nested Case-Control Analysis of the Prospective Cardiovascular Münster (PROCAM) Study. Nutr. Metab. Cardiovasc. Dis. 2006, 16, 13–21. [Google Scholar] [CrossRef]
  149. Paniagua-Pérez, R.; Madrigal-Bujaidar, E.; Reyes-Cadena, S.; Molina-Jasso, D.; Pérez Gallaga, J.; Silva-Miranda, A.; Velazco, O.; Hernández, N.; Chamorro, G. Genotoxic and Cytotoxic Studies of Beta-Sitosterol and Pteropodine in Mouse. J. Biomed. Biotechnol. 2005, 2005, 242–247. [Google Scholar] [CrossRef]
  150. Nieminen, P.; Pölönen, I.; Ikonen, K.; Määttänen, M.; Mustonen, A.M. Evaluation of Reproductive Safety of β-Sitosterol on the American Mink (Neovison Vison). Chemosphere 2007, 71, 493–499. [Google Scholar] [CrossRef]
  151. Li, J.; Wang, X.; Liu, H.; Guo, H.; Zhang, M.; Mei, D.; Liu, C.; He, L.; Liu, L.; Liu, X. Impaired Hepatic and Intestinal ATP-Binding Cassette Transporter G5/8 Was Associated with High Exposure of β-Sitosterol and the Potential Risks to Blood-Brain Barrier Integrity in Diabetic Rats. J. Pharm. Pharm. Sci. 2013, 66, 428–436. [Google Scholar] [CrossRef]
  152. Rashidi, R.; Rezaee, R.; Shakeri, A.; Hayes, A.W.; Karimi, G. A Review of the Protective Effects of Chlorogenic Acid against Different Chemicals. J. Food Biochem. 2022, 46, e14254. [Google Scholar] [CrossRef]
  153. Cos, P.; Calomme, M.; Sindambiwe, J.-B.; De Bruyne, T.; Cimanga, K.; Pieters, L.; Vlietinck, A.J.; Berghe, D. Vanden Cytotoxicity and Lipid Peroxidation-Inhibiting Activity of Flavonoids. Planta Med. 2001, 67, 515–519. [Google Scholar] [CrossRef]
  154. Spagnuolo, C.; Russo, M.; Bilotto, S.; Tedesco, I.; Laratta, B.; Russo, G.L. Dietary Polyphenols in Cancer Prevention: The Example of the Flavonoid Quercetin in Leukemia. Ann. N. Y. Acad. Sci. 2012, 1259, 95–103. [Google Scholar] [CrossRef]
  155. Vanhees, K.; De Bock, L.; Godschalk, R.W.L.; Van Schooten, F.J.; Barjesteh, V.W.; Van Doorn-Khosrovani, S. Prenatal Exposure to Flavonoids: Implication for Cancer Risk. Toxicol. Sci. 2010, 120, 59–67. [Google Scholar] [CrossRef]
  156. Utesch, D.; Feige, K.; Dasenbrock, J.; Broschard, T.H.; Harwood, M.; Danielewska-Nikiel, B.; Lines, T.C. Evaluation of the Potential in Vivo Genotoxicity of Quercetin. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2008, 654, 38–44. [Google Scholar] [CrossRef]
  157. Ferry, D.R.; Smith, A.; Malkhandi, J.; Fyfe, D.W.; de Takats, P.G.; Anderson, D.; Baker, J.; Kerr, D.J. Phase I Clinical Trial of the Flavonoid Quercetin: Pharmacokinetics and Evidence for in Vivo Tyrosine Kinase Inhibition. Clin. Cancer Res. 1996, 2, 659–668. [Google Scholar]
  158. Mozumder, S.N.; Haque, M.I.; Kamal, M.M.; Akter, S.; Banik, B.R. Effect of Storage, Growth Regulator Treatment and Seed Priming on Germination of Eryngium foetidum. Int. J. Adv. Multidiscip. 2017, 4, 16–21. [Google Scholar] [CrossRef]
  159. Mozumder, S.N.; Moniruzzaman, M.; Rahman, S.M.M.; Sarker, P.C.; Faisal, S.M. Influence of Seed Rate and Method of Sowing on the Performance of Bilatidhonia (Eryngium foetidum L.). Bangladesh J. Agric. Res. 2010, 35, 227–234. [Google Scholar] [CrossRef]
  160. Mozumder, S.N.; Hossain, M.M.; Akter, S.; Goswami, B.K. Seed Germination and Viability Improvement in Eryngium foetidum through Priming and Chemicals. Int. J. Adv. Innov. Res. 2017, 6, 94–98. [Google Scholar]
  161. Mozumder, S.N.; Kabir, A.H.M.F.; Hossain, M.M. Influence of Seed Treatment and Priming on Growth Performance of Eryngium foetidum. Int. J. Adv. Multidiscip. 2017, 4, 45–55. [Google Scholar] [CrossRef]
  162. Mozumder, S.N.; Haque, M.I.; Zaman, M.M.; Shahiduzzaman, M. Effect of Shade and Sowing Method on Eryngium foetidum Production. Int. J. Res.-Granthaalayah 2020, 8, 251–261. [Google Scholar] [CrossRef]
  163. Kuttan, A.J. Performance of Spiny Coriander (Eryngium foetidum L.) under Different Shade Regimes. Master’s Thesis, Kerala Agricultural University, Thrissur, India, 2008. [Google Scholar]
  164. Carrera-Quirino, Y.G.; Colohua-Citlahua, B. Diseño Inicial de Una Propuesta Metodológica para la Reproducción de Plantas del Género Eryngium; Orizaba: Veracruz, Mexico, 2014. [Google Scholar]
  165. Martin, K.P. In Vitro Propagation of the Herbal Spice Eryngium foetidum L. on Sucrose-Added and Sucrose-Free Medium without Growth Regulators and CO2 Enrichment. Sci. Hortic. 2004, 102, 277–282. [Google Scholar] [CrossRef]
  166. Samad, A.; Singh, B.; Gajurel, P.R. Influence of Propagules and Groth Regulators on the Performance of Underutilized Spice—Spiny Coriander (Eryngium foetidum L.). J. Spices Aromat. 2021, 30, 204–209. [Google Scholar] [CrossRef]
  167. Ayuso, M.; García-Pérez, P.; Ramil-Rego, P.; Gallego, P.P.; Barreal, M.E. In Vitro Culture of the Endangered Plant Eryngium Viviparum as Dual Strategy for Its Ex Situ Conservation and Source of Bioactive Compounds. Plant. Cell Tissue Organ. Cult. 2019, 138, 427–435. [Google Scholar] [CrossRef]
  168. Martin, K.P. Organogenesis on Root, Leaf, Stem-Disc, and Scape Explants of Eryngium foetidum L., a Rare Medicinal Plant. J. Herbs Spices Med. Plants 2006, 11, 9–17. [Google Scholar] [CrossRef]
  169. Nagananda, G.S.; Rajath, S.; Mathew, R.K.; Rajan, S.S. Effect of Adjuvants and Nitrogen Sources on in Vitro Shoot Regeneration and Clonal Propagation of Medicinally Important Plant Eryngium foetidum L. Res. Biotechnol. 2012, 3, 21–25. [Google Scholar]
  170. Jena, B.; Biswal, B.; Giri, A.K.; Parida, R.; Acharya, L. Rapid In Vitro Propagation and Genetic Fidelity Evaluation of Medicinally Important Mexican Coriander (Eryngium foetidum L.). Plant Cell Biotechnol. Mol. Biol. 2020, 21, 79–88. [Google Scholar]
Table 1. Eryngium species reported in the central-western region of Mexico.
Table 1. Eryngium species reported in the central-western region of Mexico.
SpecieDistribution
E. alternatumState of Mexico, Jalisco, and Michoacán
E. beecheyanumState of Mexico, Guanajuato, Jalisco, and Michoacán
E. bonplandiiState of Mexico, Jalisco, Guanajuato, and Michoacán
E. carlinaeState of Mexico, Guanajuato, Jalisco, and Michoacán
E. cervantesiiState of Mexico, Jalisco, Guanajuato, and Michoacán
E. columnareState of Mexico, Jalisco, and Michoacán
E. comosumState of Mexico and Michoacán
E. cymosumState of Mexico, Jalisco, and Michoacán
E. ferrisiaeJalisco and Nayarit
E. fluitans (E. mexicanum)State of Mexico and Michoacán
E. ghiesbreghtiiState of Mexico, Jalisco, and Michoacán
E. gracileState of Mexico, Jalisco, and Michoacán
E. haenkei (E. spiculosum)State of Mexico, Michoacán, and Guerrero
E. heterophyllumState of Mexico, Jalisco, and Michoacán
E. jaliscienceJalisco
E. longifoliumState of Mexico, Jalisco, and Michoacán
E. mexiaeState of Mexico, Jalisco, and Michoacán
E. nasturtiifoliumState of Mexico, Jalisco, and Michoacán
E. monocephalumGuanajuato and Michoacán
E. palmeri (E. globosum)Jalisco and Nayarit
E. pectinatumState of Mexico and Michoacán
E. phyteumaeState of Mexico and Michoacán
E. proteaeflorumState of Mexico and Michoacán
E. pugaeJalisco and Aguascalientes
E. serratumState of Mexico, Guanajuato, and Michoacán
E. sparganophyllumJalisco and Michoacán
E. subacaule (E. ranunculoides)State of Mexico and Michoacán
E. yuccifoliumMichoacán (exotic)
Table 2. Location of American and Australian (New World) Species and corresponding sections and subsections [18,19].
Table 2. Location of American and Australian (New World) Species and corresponding sections and subsections [18,19].
SectionSubsectionSpecie
CarliniformiaComosaE. carlinae
E. comosum
E. beecheyanum
MadrensiaSetoso-dentataE. heterophyllum
E. fluitans
ReptantiaE. nasturtiifolium
PanniculataE. longifolium
SpinescentiaE. cymosum
Table 3. Some characteristics on the habitat, biology, and distribution of the Eryngium species found in the central-western region of Mexico reported with a medicinal use [6,23].
Table 3. Some characteristics on the habitat, biology, and distribution of the Eryngium species found in the central-western region of Mexico reported with a medicinal use [6,23].
SpecieHabitatAltitude (m·asl)PhenologyKnown Distribution
E. beecheyanumgrassland, tropical deciduous forest, and oak-pine forest1600–2550flowers from April to May and bears fruit from June to JanuaryChiapas, Chihuahua, Tepic, Sinaloa, Zacatecas, the State of Mexico, Jalisco, Colima, Michoacán, Oaxaca, and Sonora; Central America
E.carlinaeprairies, pastures, disturbed oak, pine-oak and coniferous forest1500–3500flowers from May to July and bears fruit from August to JanuaryChihuahua, Federal District, Durango, State of Mexico, Michoacán, Hidalgo, and Oaxaca; Central and South América Guatemala, and Costa Rica
E. comosumplains and grasslands from thorny scrub and oak forest clearings1900–2300flowers from June to August and bears fruit from September to NovemberFederal District, Guerrero, Hidalgo, Michoacán, and Querétaro
E. cymosumslopes and understory of oak forest and coniferous forests2000–3200flowers from July to October and bears fruit from October to JanuaryState of Mexico, Guerrero, Hidalgo, and Michoacán
E. fluitanshumid and flooded grasslands, plains and clearings within oak, pine-oak forests2250–2700flowers from July to August and bears fruit from September to NovemberChihuahua, Durango, the State of Mexico, Michoacán, and Morelos
E. heterophyllumgrassland and secondary vegetation, tropical deciduous, and oak forests1800–2000flowers from July to August and bears fruit from September to OctoberLouisiana, Texas, and Arizona in the United States of America; in Mexico it is in Chihuahua, Durango, San Luis Potosí, Jalisco, Michoacán, State of Mexico, and Oaxaca
E. longifoliumplains and slopes of oak, oak-pine forests, and grasslands1500–2400flowers from July to August and bears fruit from September to DecemberDurango, the State of Mexico, Guerrero, Hidalgo, Jalisco, Michoacán, and Oaxaca
E. nasturtiifoliumsemi-humid environments on the edges of cultivated and disturbed lands, grasslands, and scrublands1000–1800blooms from February to May and bears fruit from May to AugustSouthern Arizona and Texas in the United States of America; Mexico: Baja California, Sonora, Sinaloa, Tamaulipas, Veracruz, Nuevo León, Nayarit, Jalisco, and Michoacán to Oaxaca; Cuba, and Central America
Table 5. Confirmed biological activities in Eryngium species distributed in the central-western region of Mexico.
Table 5. Confirmed biological activities in Eryngium species distributed in the central-western region of Mexico.
Eryngium sp.Biological Activity ConfirmedType of ExtractPlant TissueModelReference
E. carlinaeHypolipidemicEthanolicPlantIn vivo[31,50]
AqueousPlantClinical trial[32]
HexanicInflorescenceIn vitro and in vivo[52]
HypocholesterolemicEthanolicPlantIn vivo[31,51]
HydroalcoholicAerial partsIn vivo[53]
HypoglycemicHexanicInflorescenceIn vivo[52]
HexanicInflorescenceI vitro and in vivo[30]
AntioxidantHydroalcoholicAerial partsIn vivo[53]
EthanolicPlantIn vivo[51]
HexanicInflorescenceIn vitro and in vivo[30]
Diuretic-RenoprotectiveDecoctionPlantIn vivo[55]
Antimicrobiane
(Pyhytophtora cinnamomi)
EthanolicAerial partsIn vitro[59]
AnticholelithiasisHydroalcoholicPlantIn vitro[56]
AntispasmodicMethanolicAerial partsIn vivo[57]
E. comosumHypocholesterolemicAqueousPlantIn vivo[13]
AntioxidantAqueous and ethanolicPlantIn vitro[60]
EthanolicPlantIn vitro[62]
Antimicrobiane
(Equine pathogens)
EthanolicPlantIn vitro[61]
CytotoxicityEthanolicPlantIn vitro[62]
Aqueous and ethanolicPlantIn vitro and in vivo[60]
E. cymosumHypoglycemicAqueousAerial partsIn vivo[8]
AntihyperglycemicInfusionPlantIn vivo[36]
E. heterophyllumTrypanocideMethanolicAerial parts [66]
HypocholesterolemicAqueousPlantIn vivo[64]
HypocholesterolemicDecoctionPlantClinical trial[65]
E. longifoliumHypoglycemicAqueous and ethanolicAerial partsIn vivo[35]
Plant—plant parts were not specified.
Table 6. Eryngium spp. distributed in the central-western region of Mexico whose phytochemistry has been analyzed.
Table 6. Eryngium spp. distributed in the central-western region of Mexico whose phytochemistry has been analyzed.
Species AnalyzedPlant Tissue Analyzed; Type of ExtractAnalysis TechniqueGroup of Metabolites DetectedCompounds DetectedQuantificationReferenceOther Eryngium Species Containing
E. carlinaeAerial parts (stem, leaves, and inflorescences); methanolicPhytochemical screeningTannins
Saponins
Flavonoids
NADetection only[11]E. pyramidale, E. foetidum, and E. creticum [74,88,89]
E. carlinaeAerial parts; aqueous (infusion)UV-VisTotal Phenol (Folin–Ciocalteu method)
Total flavonoids
Total terpenoids
NA0.0038 mg GAE/mL
3.3032 mg QE/mL
0.0424 mg LE/mL
[12]E. creticum and E. maritimum [74,90]
E. carlinaeAerial parts; aqueous (infusions at 1 and 2% m/v; decoctions at 5 and 10 min)UV-VisTotal Phenol (Folin–Ciocalteu method) Total flavonoidsNA≥2.5 mg GAE/mL
≤0.2 mg QE/mL
[32]E. pyramidale, E. creticum, and E. foetidum [74,88]
HPLC-DAD-MSHydroxybenzoic acidsGallic acid
4-Hydroxybenzoic acid
Protocateuic acid
1.3–2.6 µg/mL
212–332 µg/mL
105–210 mg/mL
[32]E. planum, E. campestre, E. maritumum, E. alpinum, E. foetidum, E. bornmuelleri, and E. caucasicum [75,91,92,93,94,95,96]
Hydroxycinnamic acidsChlorogenic acid
Rosmarinic acid
Caffeic acid
Ellagic acid
P-coumaric acid
Ferulic acid
Synapic acid
400–700 μg/mL
40–58 μg/mL
12–21 μg/mL
78–116 μg/mL
19–32 μg/mL
10–16.5 μg/mL
2–3.5 μg/mL
[32]
FlavanolsGalocatequin gallate
Catechin
Epicatechin
Epigallocatechin gallate
600–1600 μg/mL
68–97 μg/mL
11–16.5 μg/mL
6–10 μg/mL
[32]
FlavanonesQuercetin
Rutin
Kaempferol
89–179 μg/mL
9–156 μg/mL
9–15 μg/mL
[32]
HydroxybenzaldehydesEriocitrin
Hesperidin
Naringenin
Vanillin
0.2–0.5 μg/mL
0.4–0.6 μg/mL
0.2–04 μg/mL
4.7–8.4 μg/mL
[32]
E. carlinaeAerial parts; aqueous (infusion)HPLC-DADPhenolic acids
Flavonoids
Phytosterols
Saponins
Ellagic acid
Caffeic acid
Protocateuic acid
P-hydroxybenzoic acid
Rutin
Catechin
Epicatechin
α-7-stigmasterol
β-sitosterol
β-campesterol
Stigmastanol
Campsteryl β-d-glucopyranoside
Sitosteryl β-d-glucopyranoside
38.3 mA
20.3 mA
11.9 mA
9.8 mA
14.1 mA
12.1 mA
11.7 mA
18.7 mA
11.1 mA
8.7 mA
8.4 mA
28.9 mA
20.1 mA
[55]
E. carlinaeInflorescence; hexanicGC-MSTerpenes
Sesquiterpenes
(Z) β-farnesene
β-pinene
Calamenene
α-farnesene
38.79%
17.53%
13.3%
10.38%
[30]E. glaciale, E. amethystinum, E. campestre, E. thorifolium, E.
Creticum, E. pristis, E. maritimum, E. alpinum, and E. rosulatum [97,98,99,100,101,102]
E. carlinaeAerial parts; ethanolicGC-MSSesquiterpenes
Fatty acids
α-selinene
β-selinene
Palmitic acid
Stearic acid
Humulene
Stigmasterol
Elemol
Elemene
α-cedrene
17.54%
26.04%
14.43%
14.53%
≤5%
[31]
E. carlinaeAerial parts; hydroalcoholic (EtOH-H20, 7:3 v/v)GC-MSSaccharides
Polyols
Hexa-O-acetyl-d-mannitol, acetylated derivatives thereofDetection only[53]E. dichotomum [103]
E. carlinaeLeaves and stems; ethanolicGC-MSSaccharides
Polyols
Organic acids
D-(−)-fructofuranose
D-(−)-fructopyranose
D-(−)-tagatofuranosa
1,5-anhydro-d-sorbitol
Cinnamic acid
Detection only[87]E. dichotomum and E. bourgatii [103,104]
E. carlinaeAerial parts (leaves + inflorescences); ethanolicGC-MSTerpenoidsBorneol
α-pinene
Myrcene
Caryophyllene
β-pinene
367 mg/L
278 mg/L
256 mg/mL
225 mg/mL
120 mg/mL
[59]E. alpinum, E. amethystinum, and E. bungei [105,106]
E. carlinaeAerial parts; ethanolicPhytochemical screeningTriterpenoids
Sterols
Tannins
Coumarins
Carboxyls
Flavonoids
Carbohydrates
NADetection only[58]E. pyramidale, E. foetidum, and E. creticum [74,88,89]
E. comosumAerial parts; aqueous (decoction)Phytochemical screeningAlkaloids
Flavonoids (flavones and xanthones)
Triterpenoid saponins
Reducing sugars
Tannins derived from catechol
Phenolic compounds
Benzoquinones
NANA[13]
E. comosumAerial parts; aqueous, and hidroethanolic 50% and 70%UV-VisTotal Phenol
Total Phenol (Folin–Ciocalteu method)
Total saponins
NA8.0–13.3 mg/mg
0.69–4.33 mg GAE/mg
0–29.33 mg/g DW
[62]E. creticum and E. maritimum [74,90]
E. comosumAerial parts; ethanolicUV-VisTotal Phenol Total Phenol (Folin–Ciocalteu method)
Total saponins
NA22.1 mg/mg
4.33 mg GAE/g dry weight (DW)
62.2 mg/g DW
[61]
E. longifoliumAerial parts; aqueous (infusion)HPLCPhenolic acids
Isoflavones
Glycosylated flavonoids
Caffeic, chlorogenic acid, and rosmarinic acidDetection only[35]E. planum, E. campestre, E. maritumum, E. alpinum, E. foetidum, E. bornmuelleri, and E. caucasicum [43,75,91,92,93,94,95,96,107]
E. cymosumAerial parts; aqueous (infusion)HPLCPhenolic acidsCaffeic, chlorogenic acid, and rosmarinic acidDetection only[8]
E. cymosumAerial parts; aqueous (infusion), ethanolic, and organic (butanol)HPLC, NMRPhenolic acids
Flavonoids
Chlorogenic and rosmarinic acid (EA)
Caffeic and protocateuic acid
Kaempferol-3-O-(2,6-di-O-trans-p-coumaryl)-β-d-glucopyranoside
Detection only[40]
NA, not applicable; UV-Vis, ultraviolet visible spectrophotometry; HPLC-DAD-MS, high-performance liquid chromatography with a diode array detector coupled to mass spectrometry; HPLC-DAD, high-performance liquid chromatography with a diode array detector; GC-MS, gas chromatography coupled to mass spectrometry; NMR, nuclear magnetic resonance.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cárdenas-Valdovinos, J.G.; García-Ruiz, I.; Angoa-Pérez, M.V.; Mena-Violante, H.G. Ethnobotany, Biological Activities and Phytochemical Compounds of Some Species of the Genus Eryngium (Apiaceae), from the Central-Western Region of Mexico. Molecules 2023, 28, 4094. https://doi.org/10.3390/molecules28104094

AMA Style

Cárdenas-Valdovinos JG, García-Ruiz I, Angoa-Pérez MV, Mena-Violante HG. Ethnobotany, Biological Activities and Phytochemical Compounds of Some Species of the Genus Eryngium (Apiaceae), from the Central-Western Region of Mexico. Molecules. 2023; 28(10):4094. https://doi.org/10.3390/molecules28104094

Chicago/Turabian Style

Cárdenas-Valdovinos, Jeanette G., Ignacio García-Ruiz, María V. Angoa-Pérez, and Hortencia G. Mena-Violante. 2023. "Ethnobotany, Biological Activities and Phytochemical Compounds of Some Species of the Genus Eryngium (Apiaceae), from the Central-Western Region of Mexico" Molecules 28, no. 10: 4094. https://doi.org/10.3390/molecules28104094

Article Metrics

Back to TopTop