Next Article in Journal
Increasing the Contents of Paddy Soil Available Nutrients and Crop Yield via Optimization of Nitrogen Management in a Wheat–Rice Rotation System
Next Article in Special Issue
Antigiardial Activity of Foeniculum vulgare Hexane Extract and Some of Its Constituents
Previous Article in Journal
Phytochemical Diversity and Antioxidant Potential of Wild Heather (Calluna vulgaris L.) Aboveground Parts
Previous Article in Special Issue
Bio-Guided Isolation of SARS-CoV-2 Main Protease Inhibitors from Medicinal Plants: In Vitro Assay and Molecular Dynamics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Use of Agave Genus in Neuroinflammation Management

by
Maribel Herrera-Ruiz
1,
Enrique Jiménez-Ferrer
1,
Manasés González-Cortazar
1,
Alejandro Zamilpa
1,
Alexandre Cardoso-Taketa
2,
Martha Lucía Arenas-Ocampo
3,
Antonio Ruperto Jiménez-Aparicio
3 and
Nayeli Monterrosas-Brisson
4,*
1
Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Xochitepec 62740, Mexico
2
Centro de Investigación en Biotecnología, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca 62209, Mexico
3
Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional (IPN), Yautepec 62739, Mexico
4
Facultad de Ciencias Biológicas, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca 62209, Mexico
*
Author to whom correspondence should be addressed.
Plants 2022, 11(17), 2208; https://doi.org/10.3390/plants11172208
Submission received: 11 June 2022 / Revised: 12 August 2022 / Accepted: 21 August 2022 / Published: 25 August 2022
(This article belongs to the Special Issue Biological and Chemical Activity of Metabolites of Medicinal Plants)

Abstract

:
Agavaceae contains about 480 species, commonly used in the production of alcoholic beverages such as tequila and mezcal, making it a resource of economic and cultural importance. Uses of this plant rely mainly on the stem; other components such as the leaves are discarded, generating agro-industrial waste, despite being a source of bioactive and nutraceutical products. Reports show anti-inflammatory and anti-neuroinflammatory effects of these species, with flavonoids and saponins being mainly responsible. Neuroinflammation is a brain process that plays a key role in the pathogenesis of various neurodegenerative disorders and its effects contribute greatly to mortality and morbidity worldwide. This can be triggered by mechanisms such as glial reactions that lead to the release of inflammatory and oxidative molecules, causing damage to the CNS. Treatments do not cure chronic disease associated with inflammation; they only slow its progression, producing side effects that affect quality of life. Plant-based therapy is promising for treating these diseases. Pharmacological activities have been described for the Agavaceae family; however, their role in neuroinflammation has not been fully investigated, and represents an important target for study. This review synthesizes the existing literature on the biologically active compounds of Agave species that are related in some way to inflammation, which will allow us to propose a line of research with this genus on the forefront to orient experimental designs for treating neuroinflammation and associated diseases.

1. Introduction

The Agavaceae Endl. Family is distributed in the American continent, among the United States, Central America and the Antilles [1]. In Mexico, approximately 342 species are recognized, and are distributed among eight different genera: Agave L., Beschorneria Kunth, Furcraea Vent., Hesperaloe Engelm., Manfreda Salisb., Polianthes L., Prochnyanthes S. Watson and Yucca L. [2]. The Agave genus is mostly distributed in Mexico, where approximately 75% of these species are located [3].
In Mexico, the genus Agave is of great commercial and cultural importance; several species have been used for centuries as a source of fiber, food, medicine, fuel, shelter, ornament, textiles, compost and alcoholic beverages such as tequila, mezcal and bacanora [4]. More recently, the interest in these species has been growing as potential nutraceuticals, prebiotics, natural sweeteners and biofuels [5,6,7].
In traditional medicine, Agave species have been used to treat wounds, sores, trauma, fractures, rheumatoid arthritis, psoriasis, snake bites [8,9], syphilis, scurvy, cancer, limb paralysis, and postpartum abdominal inflammation, as well as being used as diuretics and laxatives [10]. They have also been employed to treat diseases of bacterial etiology such as gastrointestinal and wound infections, urologic disorders, and dysentery as well as cancer, diabetes and hypertension [11].
There are several reports indicating that Agaves have a variety of medicinal properties such as antioxidant, antibacterial [11], anticancer [12] and anti-inflammatory [13,14] activities; at least one study reports an anti-neuroinflammatory effect [15]. Since the process of neuroinflammation can be a start point of several chronic neurodegenerative diseases [16,17,18,19,20], a huge number of natural products have been investigated regarding their capacity to act as anti-inflammatory agents, especially in the brain. However, the role in neuroinflammation has not been fully investigated for these species. The objective of this review is to frame the pharmacological studies that refer to the capacity of extracts and secondary metabolites isolated from several species of the genus Agave, to act on some pathophysiological process associated with inflammation, so that the reader can visualize the potential of this genus and its unexplored applications in the field of neuroinflammation. In addition, the study of these plants could provide evidence for their employment in the treatment of this medical condition and related disorders. Therefore, studies that demonstrate various pharmacological effects are cited, highlighting antioxidant, immunomodulatory, anti-inflammatory and anti-neuroinflammatory properties. This review leads us to a description of the agaves, trying to address inflammation-related activities; this allows us to infer that this genus could become an object of study in the search of an anti-neuroinflammatory treatment, and for the development of a possible phytomedicine that could help to improve the quality of life of patients with chronic degenerative diseases with brain inflammation as a pathophysiological background. Additional value is assigned to the leaves of these species, because despite possessing a wide variety of active metabolites, they are considered as waste material in the mezcal and tequila industry, since the stem is the only material used.

2. Methodology

2.1. Search Planning

The methodology applied in this review consisted mainly in searching pharmacological information of species of the Agave genus, with reports about applications related to inflammation, in databases such as PubMed, Google scholar and Scopus. Showing the following results for the queries “anti-inflammatory Agave polysaccharide” (2); “anti-neuroinflammatory Agave polysaccharide” (0); “anti-inflammatory Agave saponins” (195); “anti-neuroinflammatory Agave saponins” (1); “anti-inflammatory Agave steroidal saponins” (141); “anti-neuroinflammatory Agave steroidal saponins” (1); “anti-inflammatory Agave terpenes” (102); “anti-neuroinflammatory Agave terpenes” (0); “anti-inflammatory Agave alkaloids” (200); “anti-neuroinflammatory Agave alkaloids” (0); “anti-inflammatory Agave flavonoids” (2); “anti-neuroinflammatory Agave flavonoids (0)”; “anti-inflammatory Agave fatty acids” (258); “anti-neuroinflammatory Agave fatty acids” (6); “anti-inflammatory Agave β-sitosterol” (1) and “anti-neuroinflammatory Agave β-sitosterol” (0); “neuroinflammation and Isorhamnetin” (7); “neuroinflammation and hesperidin” (31); “neuroinflammation and delphinidin” (0); “inflammation and delphinidin” (60); “neuroinflammation and quercetin” (115); “neuroinflammation and apigenin” (48); “neuroinflammation and catechin” (90).

2.2. Study Selection

Once the search was complete, it was apparent that studies focusing on evaluating the anti-neuroinflammatory activity of Agave compounds were scarce. However, there are studies that report anti-inflammatory activity in this genus and they could possibly act at the CNS level. Due to this, our bibliographic selection method consisted in choosing all those studies related with Agave genus pharmacology and phytochemistry, without considering the date of publication.

2.3. Data Extraction

The selected papers were divided into three groups, those related to botanical and taxonomic information of the genus, phytochemical studies and finally, those related to pharmacological reports.
The information gathered was synthesized in a table that included the following data: (a) scientific name, (b) common names, (c) medicinal uses, (d) distribution in Mexico and e) references. Biological activities reported for each Agave species mentioned in the text, as well as the compounds isolated, identified and/or evaluated, were synthesized for this work. Therefore, despite the number of studies that the query of different natural compounds accompanied by the word “Agave anti-inflammatory or anti-neuroinflammatory” showed, only those that actually addressed the natural compound type and pharmacology were analyzed. This is because most of the studies issued only appeared because of the similarity of some of the search terms. Hence, fatty acids, triterpenes and alkaloids will not be described in this review.
Finally, we selected only information useful to achieve the stated objective and decided to write the paper.

3. Pharmacological Background of Agave Species

This section includes background information related mainly to the pharmacology of the extracts of the Agave genus species, as well as some of the isolated compounds. In addition to the species mentioned here, the use of these plants in traditional medicine and their distribution in this country were also included (Table 1). This information demonstrates that the species of the Agave genus are perceived with important properties to reduce inflammation and that this attribute can be used as a point of research that analyzes in detail the anti-neuroinflammatory properties of these species that can be applied in the ailment of chronic degenerative diseases related to this process.

3.1. Antioxidant Activity

Using the in vitro methods of the free radical 2,2-diphenylpicrylhydrazyl (DPPH) and the 2,20–azinobis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), the antioxidant capacity was measured (as µM of Trolox Equivalents (TE)/g dw)., for the methanolic extract of leaves from A. rzedowskiana (27.4127,41 ± 3.35 µM TE/g dw) > A. ornithobroma > A. schidigera > A. tequilana > A. impressa > A. angustifolia [11].
The A. marmorata Roezl chemical and pharmacological activity studies, showed that the saponin smilagenin-3-O-[β-D-glucopyranosyl (1→2)-β-D-galactopyranoside] (1) inhibited the production and release of nitric oxide induced with lipopolysaccharide (LPS) (EC50 = 5.6 mg/mL, Emax = 101%) [29].
For A. salmiana, it was determined that the culture conditions IN (in vitro plants obtained from the tissue culture laboratory at the multiplication step), EN (ex vitro plants obtained after acclimatization step) and WT (wild-type plants obtained from a natural population) exerted an effect on the total concentration of saponins and phenolic acids in methanolic leaf extracts. They observed an increase of 35 and 40% in the phenolic acids content in the IN and EN plants, respectively, compared with WT. The same pattern was observed in terms of total saponin concentration; IN and EN plants showed a 36- and 29-fold increase in saponin content compared to the WT plants. Antioxidant activity was determined using the oxygen radical absorbance capacity assay. IN had the highest antioxidant activity (369 µmol TE/g dw), followed by EN and WT, (184 and 146 µmol TE/g dw). The IN activity was 2.5-fold and 2.8-fold higher than EN and WT, respectively. However, a correlation between antioxidant activity and total phenolic acids or total saponins was not found [30].
The methanolic extract of A. americana leaves showed a direct correlation between total polyphenol and total flavonoid contents on antioxidant activity. According to their capacity to neutralize the free radical 2,2-diphenylpicrylhydrazyl (DPPH), the best one was that with a high total polyphenol content [31].

3.2. Anti-Inflammatory and Immunomodulatory Activities

A. angustifolia extracts, fractions and a purified compound 3-O-[(6’-O-palmitoyl)-β-D-glucopyranosyl sitosterol (2), isolated from A. angustifolia, have demonstrated an anti-inflammatory effect against the oedema induced in mouse ears by 12-O-tetradecanoylphorbol-13-acetate (TPA) at 0.8 mg/ear. Treatment with A. angustifolia led to a decrease in the concentrations of various pro-inflammatory cytokines such as Tumor Necrosis Factor-α (TNF-α), Interleukin-1β (IL-1β) and Interleukin-6 (IL-6) as well as an increase in anti-inflammatory cytokines Interleukin-4 (IL-4) and Interleukin-10 (IL-10) [14].
A. tequilana has been shown to have an immunomodulatory effect in a model of systemic lupus erythematosus that is experimentally induced with pristane (2,6,10,14-tetramethylpentadecane). A. tequilana extract decreased articular inflammation and proteinuria, reduced ssDNA/dsDNA antinuclear antibody titers, reduced IL-1β, IL-6, TNF-α, Interferon-γ (IFN-γ) levels and increased IL-10. The phytochemical analysis of A. tequilana extracts revealed the presence of β-sitosterol glycoside, phytol, octadecadienoic acid-2,3-dihydroxypropyl ester, stigmasta-3,5-dien-7-one, cycloartenone and cycloartenol [32,33,34]. Aqueous extract and acetonic fractions of this plant, administered in mice (50 mg/kg) with hypertension induced by AGII, showed the plant’s capacity to diminish that condition, and also provoked a significant decrease in IL-1β, IL-6 and TNFα kidney levels induced by AGII. A. tequilana methanolic fraction showed TNFα levels in the kidney similar to the baseline treatment [35].
The saponin (1) isolated from A. marmorata, evaluated on a culture of RawBlue cell line, was capable of inhibition of NF-κB expression (EC50 = 0.086 mg/mL, Emax = 90%), [29].
Dry extracts from decoctions of A. intermixta leaves (300 and 500 mg/kg p.o.) showed potent in vivo anti-inflammatory activity in the carrageenan-induced rat paw edema with a 50.13 ± 3.8% edema inhibition, meanwhile in an assay of TPA-induced ear edema, the topical administration of A. intermixta leaves decoctions (3 and 5 mg/ear) significantly inhibited oedema (50%) compared to the control group [36].
The aqueous extract of A. americana was shown to contain terpenoid compounds and steroidal saponins with anti-inflammatory properties, including agavasaponin E (3) and agavasaponin H (4), hecogenin (5) and tigogenin (6) [37,38,39,40,41,42,43]. Using the carrageenan-induced oedema model, the aqueous extract (39 µg/kg) from A. americana and a sapogenin-enriched fraction extract (hecogenin and tigogenin) showed anti-inflammatory effects, with 50% and 70–100% inhibition of oedema, respectively [37]. A. americana hydroalcoholic extract (100, 200 and 400 mg/Kg) caused a significant concentration-dependent reduction in paw oedema [43].
The pharmacologic effect of cantalasaponin-1, a saponin isolated from the species A. americana, A. barbadensis [44] and Furcraea selloa var. marginata [45], was evaluated on TPA-induced auricular oedema, showing a dose-dependent inhibitory effect of up to 90% at the highest dose of 1.5 mg/ear [13].
It should be noted that in the review carried out in the literature, there is only one report about anti-neuroinflammatory activity to the genus Agave, specific to A. americana and the compound cantalasaponin-1. In this study, A. americana (125 mg/Kg) and cantalasaponin-1 (5 and 10 mg/kg) reduced brain concentration of LPS-induced proinflammatory cytokines IL-6 and TNF-α. Cantalasaponin-1 increased the brain concentration of the anti-inflammatory cytokine LI-10 [15].

3.3. Anti-Cancer and Anti-Bacterial Activities

The anticancer activity of linear inulin-type fructan (ITF) prebiotics from A. angustifolia play a significant role in the prevention of colorectal cancer. This was evaluated by using a culture of the human colon cancer cell line Caco-2 (colorectal adenocarcinoma), in the Simulator of Human Intestinal Microbial Ecosystem model. The proximal, transverse and distal vessels were used to investigate fructan fermentation throughout the colon to assess the alterations of the microbial composition and fermentation metabolites (short chain fatty acids and ammonia). The influence on bioactivity of the fermentation supernatant was assessed by MTT (viability assay), Comet (DNA damage assay) and transepithelial electrical resistance (TER), respectively. A. angustifolia fructans significantly increased the population of bifidobacteria, short-chain fatty acid levels and transepithelial electrical resistance while decreasing ammonia levels. This indicates a protective effect on the function of the intestinal barrier, which is an important aspect in the pathology of colon cancer [46].
Five saponins isolated from A. fourcroydes methanolic leaves extract were evaluated to determine cytotoxic activity by fluorometric microculture cytotoxicity assay (FMCA). A new steroidal saponin, elucidated as chlorogenin 3-O-[α-L-rhamnopyranosyl-(1→4)-β-D-glucopyranosyl-(1→3)-{β-D-glucopyranosyl-(1→3)-β-D-glucopyranosyl-(1→2)}-β-D-glucopyranosyl-(1→4)-beta-D-galactopyranoside], along with four known saponins including furcreastatin, chlorogenin 3,6,-di-O-β-D-glucopyranoside and tigogenin3-O-[α-L-rhamnopyranosyl-(1→4)-β-D-glucopyranosyl-(1→3)-{β-D-glucopyranosyl-(1→3)-β-D-glucopyranosyl-(1→2)}-β-D-glucopyranosyl-(1→4)-β-D-galactopyranoside] showed strong cytotoxicity against HeLa cells with IC50 values of 13.1, 5.2, and 4.8 µg/mL, respectively [47].
A smilagenin di-glycoside elucidated as (25R)-5β-spirostan-3β-ylO-β-D-glucopyranosyl-(1→4)-β-D-galactopyranoside and isolated from A. utahensis Engelm showed cytotoxicity against HL-60 cells with an IC50 value of 4.9 µg/mL, inducing apoptosis through a marked caspase-3 activation [48]. Steroidal saponins isolated from A. utahensis showed moderate cytotoxic activity against HL-60 cells at 20 µg/mL in contrast to the control, etoposide [49]. The spirostanol saponin AU-1 isolated from A. utahensis, caused a transient increase in cyclin-dependent kinase inhibitor (CDKI) p21/Cip1, through the upregulation of the miRNAs miR-34 and miR-21. AU-1 stimulated p21/Cip1 expression without exerting cytotoxicity against different types of carcinoma cell lines (human renal adenocarcinoma-derived ACHN cells and human hepatocellular carcinoma HepG2 cells). In renal adenocarcinoma ACHN cells, AU-1 transiently elevated the expression level of p21/Cip1 protein without marked increases in p21/Cip1 mRNA levels. Rapid and transient increases in miR-34 and miR-21, known to regulate p21/Cip1, were observed in AU-1-treated cells [50].
Concentrated Agave Sap (CAS) obtained from 18 different Mexican states, showed an antiproliferative effect on the culture of cancer cells since it significantly reduced cell viability to 80 % when tested at a concentration of 75 μg/mL on HT-29 (HTB-38), a cellular line with epithelial morphology isolated from colorectal adenocarcinoma. Agave sap also had apoptotic activity on HT-29 (IC503.8 ± 1.3 mg/mL) and NIH-3T3, which are embryonic mouse fibroblast, (IC508.4 ± 1.0 mg/mL) cell lines. The activities found were attributed to the main saponins detected in CAS, including kammogenin and manogenin [12].
The n-butanol extract from inflorescences of A. schotti Engelm was active against Walker carcinoma 256 (intramuscular) tumor system (5WM). Activity was detected in Sprague rats at a level of 7% T/C (Test Control; when the degree of screening activity is significantly greater than the minimum values: ≥125 for survival systems or ≤42 for tumor-weight inhibition) at 75 mg/kg and 28% T/C at 37.5 mg/kg, induced by the saponin-rich fraction separated from the n-butanol extract. Six saponins were isolated, the fourth being the most active against the 5WM tumor system (17% T/C at 65 mg/kg and 22% T/C at 33 mg/kg). Antitumor activity of the system was defined as a percent T/C value of less than 60 in a satisfactory dose response test [51].
Methanolic extract of A. tequilana leaves had antibacterial activity against Pseudomona aeruginosa ATCC 27853 and Escherichia coli ATCC 25922 strains (Minimal Inhibitory Concentration of 5 mg/mL) and the evaluation was carried out using the broth microdilution method. These results were associated with the presence of metabolites like tannins, alkaloids, flavonoids, and saponins [11]. A. tequilana syrup at 50% (w/v) inhibited Bacillus subtillis 168 and E. coli DH5 growth. The bacteriostatic activity was attributed to the high sugar content and high viscosity [52].
The A. americana methanolic leaf extract induced a potent cytotoxic effect against MCF-7 (breast carcinoma) and Vero (African green monkey kidney) cell line. IC50 values were found to be 546 and 1854 μg/mL respectively by the SRB (determination of total cell protein content by sulphorhodamine B) assay [53].

3.4. Other Activities of Agave Genus

It has been established that obesity is associated with chronic inflammation, and it has been demonstrated in animal models that A. tequilana fructans intake induced a decrease in body mass index (35.3 kg/m2 to 33.0 kg/m2), total body fat percentage (38 to 20%), and also in triglyceride levels (167.4 to 107.9 mg/dL p < 0.05), in obese individuals [54].
In addition, the aqueous extract of A. salmiana leaf (100 mg/Kg) has anthelmintic properties, strongly reducing H. gallinarum worm egg counts and worms [55]. Hecogenin (5) (90 mg/Kg), a steroid saponin isolated from A. salmiana leaves, showed anti-ulcer properties in ethanol- and indomethacin-induced gastric ulceration. The gastroprotection mechanism of hecogenin was K-ATP channel dependent [56]. In another study, the depressive behavior induced by reserpine (2 mg/kg, o.p.) was reversed by hecogenin (5 and 10 mg/kg), since it decreases the immobility time compared to the control group (Tween 80/4%) similarly to imipramine (10 mg/Kg) [57].
Magueyosides A, B, D and E isolated from the hydroalcoholic extract of A. offoyana flowers, showed phytotoxic activity at concentrations above 100 µM. They significantly inhibited Lactuca sativa L. root growth (IC50 88.4µM, 104.3 µM, 131.2 µM and, 101.6 µM, respectively) compared to the commercial herbicide Logran® (523.7 µM). At the same time, saponin concentrations below 33 µM enhanced root growth [1].
Two saponin-enriched fractions (SFs) obtained through the best traditional extraction method (ethanol:water 8:2) and the best new method (n-butanol:water 1:1) from A. tequilana, A. salmiana, A. angustifolia, A. furcroydes, A. hookeri, A. inaequidens, A. marmorata and A. atrovirens leaves exhibited some selective phytotoxic activity against weeds from the Standard Target Species (STS) Solanum lycopersicum (tomato), Lactuca sativa (lettuce) and Lepidium sativum (cress), as well as on two harmful weed species for agricultural crops Lolium perenne (perenne ryegrass) and Echinochloa crus-galli (barnyardgrass). The authors mentioned that the saponins from the leaves of these species could be a potential source of ecoherbicides [58].
The insecticidal and repellent activity of A. americana methanolic leaf extract was also assessed either by topical application, treated filter-paper methods or repellent bioassays against Sitophilus oryzae adults. The activity was attributed to the main identified compounds (flavonoids and saponins) permeating the insect’s cuticle [59].
The steroidal saponins Agamenoside G, degalactotigonin, cantalasaponin 1, (25R)-5α-spirost-3β-hydroxy-12-oxo-3-O-β-D-glucopyranosyl-(1-2)-[β-D-xylopyranosyl-(1-3)]-β-D-glucopyranosyl-(1-4)-β-D-galactopyranoside, deltonin, and dioscin isolated from A. americana, showed antifungal activity against Candida albicans ATCC 90028, Candida glabrata ATCC 90030, Candida krusei ATCC 6258, Cryptococcus neoformans ATCC 90113, and Aspergillus fumigatus ATCC 90906 [60].

4. Natural Products from Agave Species

In the next section, we present the names of some natural compounds that are part of the secondary metabolism of Agave species (Table 2). According to the purposes of this review, we present those that we consider the most relevant in terms of the anti-inflammatory and/or anti-neuroinflammatory activity they exert.
A number has been assigned to each compound. The structures correspond to compounds from different chemical families.

4.1. Phytosteroles

Phytosterols are bioactive compounds that are naturally present in plant cell membranes with chemical structures similar to the mammalian cell-derived cholesterol. Among various phytosterols, β-sitosterol (7) is the main compound, found in abundant quantities in plants. It has been evidenced in many in vitro and in vivo studies that SIT possesses various biological functions such as anxiolytic, sedative, analgesic, immunomodulator, antimicrobial, anticancer, anti–inflammatory, lipid lowering effect, hepatoprotective, aid in respiratory diseases, wound healing effect, antioxidant and anti-diabetic activities [61].

4.1.1. β-Sitosterol and Neuroinflammation

BV2 murine microglial cells treated with different concentrations of SIT prior to LPS stimulation, showed that (1) reduced the LPS-induced expression of inflammatory mediator like IL-6, inducible nitric oxide (iNOS), TNF-α and cyclooxygenase-2 (COX-2). SIT treatment also inhibited the LPS-induced activation of 38p, ERK and Nuclear factor kappaB (NF-κB) signaling pathways [62].

4.1.2. Agave β-Sitosterol

The acetonic extract of Agave angustifolia stem showed immunomodulatory activity and it was demonstrated that the compounds responsible for this effect were β-sitosterol β-D-glucoside (BSSG) and its free phytosterol, known as β-sitosterol [14].

4.2. Flavonoids

Flavonoids are phenolic compounds comprising fifteen carbon atom structures (C6-C3-C6). They consist of two aromatic carbon group rings: benzopyran and benzene. According to their structure, they are classified as flavones, flavonols, flavanones, flavanonols, flavanols, anthocyanidins, isoflavones, neoflavonoids and chalcones [63].

4.2.1. Flavonoids and Neuroinflammation

Recent studies demonstrate the potential role of these compounds in the treatment of neurodegenerative disorders.

4.2.2. Agave Flavonoids

HPLC-UV-MS analyses confirms the concentration of isorhamnetin (8, 1251.96 μg), flavanone (291.51 μg), hesperidin (9, 34.23 μg), delphinidin (10, 24.23 μg), quercetin (11, 15.57 μg), kaempferol (13.71 μg), cyanidin (12.32 μg), apigenin (9.70 μg) and catechin (7.91 μg) per gram of Agave lechuguilla dry residue [64]. Hamissa et al. (2012) reported that the flavonoid content in A. americana leaves ranged from 0.96 to 4.90 mg of quercetin equivalents g−1 d.w. Gallic acid was also reported in A. tequilana, A. ornithobroma, A. impressa, A. rzedowskiana, A. schidigera and A. angustifolia [31]. Tannic acid was reported from A. americana leaf extract [65]. In A. durangensis extract, several flavonoids were identified, including kaempferol glycoside, quercetin glycoside, kaempferol-3,7-O-diglucoside, kaempferol-3-O-[6-acetylglucoside]-7-O-glucoside, kaempferol-3-O-[rhamnosyl(1-6) glucoside], quercetin-3-O-arabinoside and kaempferol-3-O-rhamnoside [66].
Several flavonoids have been shown the ability to exert pharmacological effects on the CNS. Many of these have been isolated in different Agave species, but there are no data in the bibliography that indicate this effect in flavonoids of this genus. We will now review some studies directly related to neuroinflammation, carried out with different flavonoids.
Isorhamnetin (8). It was administered for 14 weeks in obese mice on a hypercaloric diet, and it was observed that it prevents cognitive deterioration and inhibits the overactivation of microglia and the high concentration of inflammatory cytokines in the serum and brain of said mice, in addition to decreasing the expression of inflammation regulators such as p-NFkB, p-JNK (c-Jun N-terminal kinase) and p-p38 (mitogen-activated protein kinase) [67]. Compound (2) exerts a concentration-dependent effect (50, 100 and 200 µM) on the effects of LPS (100 ng/mL) in BV2 microglia culture, inhibiting NO, prostaglandin E2 (PGE2), TNF-α, IL-1β concentration and inhibition of iNOS, COX 2, TNF α and IL-1β expression by ISO in BV2 microglial cells [68].
The neuroprotective effect of (8), was also evidenced in N9 cells culture, since it inhibited NO synthesis at an IC50 17.87μM [69].
For Hesperidin (9), a flavanone derived from the secondary metabolism of fruits, mainly those belonging to the citrus class, a wide range of information is available about its neuroprotective effect, which has been analyzed in detail in several review papers [70,71]. Therefore, only the most recent data directly related to neuroinflammation will be indicated here.
LPS administration for several weeks in adult mice induces glial activation, synaptic dysfunction, cognitive impairment and neuroinflammation, among other factors. In addition, it induces the alteration of some mechanisms such as increased expression of markers such as Toll-like receptor-4 (TLR4), GFAP and Iba-1 in hippocampal and cortical regions. In this study, animals treated with (9), showed significantly reduced levels of these factors. Furthermore, the expression of phosphorylated nuclear factor-κB (p-NF-κB), TNF-α and IL-1β in both cortical and hippocampal regions, as well as in BV2 cells, decreased with the administration of (9). It also decreased reactive oxygen species generation and lipid peroxidation, as well as preventing apoptosis, improves synaptic integrity, cognition and memory processes [72].
Oral administration of (9), at 50, 100 and 200 mg/kg for 25 days in mice with experimental autoimmune encephalomyelitis (EAE), causes a significant decrease in the concentration of proinflammatory cytokines, such as IL-17, TNF-α and IL -6; as well as an increase in IL-10 and Transforming Growth Factor-β (TGF-β) in splenocytes and lymph nodes, while in the brain it was observed that (3) induces a decrease in the expression of the same inflammatory cytokines and an increase in anti-inflammatory ones. In this study, the authors point out that the neuroprotective effect of this flavonoid is through the induction of the polarization of CD4+ T cells towards regulatory T cells [73]. Oral administration of (9) at 100 and 200 mg/kg also has neuroprotective effect against sodium fluoride (NaF)-induced neurotoxicity in rats by reducing lipid peroxidation and increasing superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) activities. In addition, this flavonoid reduced the levels of NF-κB, IL-1β, TNF-α, Beclin-1 (a mammalian protein that plays a central role in autophagy, a programmed cell survival process, which increases during periods of cellular stress and is extinguished during the cell cycle), LC3A and LC3B (structural proteins of autophagosomal membranes), in brain tissue [74].
Delphinidin (10). This anthocyanin has been investigated in multiple pharmacological studies; however, when the term “neuroinflammation” is included in the search together with this compound, there are no results. Nevertheless, there are data indicating, for example, that it is able to reduce inflammation induced by spinal cord injury in rats, because (10) is able to reduce the levels of TNF-α, IL-6, COX-2, PGE2 and caspase-3 and levels of NF-κB, AP-1 and p38-MAPK expression [75].
Quercetin (11). This flavonoid is one of the most studied; when its name is consulted in PUBMED, a total of 23,850 citations are generated. Delimiting it with “quercetin + neuroinflammation”, the result was 115 publications. As the aim of this paper is not to review this molecule and its anti-neuroinflammatory effect, only the three most recent ones and those in which the research model employed LPS will be mentioned.
LPS i.p. administration at 0.25mg/kg per day in C57BL/6N mice induces a wide spectrum of pathophysiological conditions, leading to neuroinflammation, oxidative stress and cognitive impairment, among others. In this scheme, intraperitoneal administration of (11) at 30 mg/kg per day for two weeks reduced gliosis and protected against neuroinflammation by causing a decrease in TNFα, COX-2 and NOS-2 expression, all in the cortex and hippocampus of adult mice compared to the control group of mice. Immunofluorescence showed an attenuation of the number of IL-1β-positive cells compared to control mice in the same regions of the brain [76]. LPS was injected into the lateral ventricle of rats at 2 μL/min during 5 min, eliciting neuroinflammation and anxiety-like behavior evaluated in the elevated maze. Under this experimental scheme, administration of (11) at 10, 50 and 100 mg/kg for 21 days produces an anxiolytic effect as well as reducing the levels of proinflammatory markers associated with NF-κB activation such as IL-6, COX-2, with a marked increase in brain-derived neurotrophic factor (BDNF) and mRNA levels and a decrease in the level of iNOS mRNA [77].
A recently published research paper shows that (11) protects against the deleterious effect of LPS by decreasing NO production and the expression of IL-6, TNF-α and IL-1β in macrophage and microglia cultures, preventing polarization towards the M1 genotype. This molecule also reduced reactive oxygen species (ROS) production and the ability of LPS to activate the phagocytic capacity of microglia. Likewise, (11) acts in a balanced way in cell cultures, since it modulates the activation towards the M2 genotype because of the increased expression of its markers such as IL-10, HO-1, GCLC (catalytic subunit of glutamate-cysteine ligase), GCLM (glutamate-cysteine ligase modifying subunit), and NQO1 (NAD(P)H quinone oxidoreductase-1) by directly activating the AMP-activated protein kinase (AMPK) and Akt signaling pathways [78].
Apigenin (12). This flavonoid is abundant in fruits and vegetables, with extensive studies that indicate potential pharmacological effects upon the inflammatory process, as well as extensive reports on anti-neuroinflammatory properties; for this reason, only some data relevant to the present review are mentioned here.
In a co-culture of neurons and glial cells stimulated with LPS (1 µg/mL) for 26 days from neonatal and embryonic rat cortex (Wistar strain) and treated with (12), it was demonstrated that this compound reduced the decline of neurons and astrocytes, as well as the reduction in the microglial activation and the CD68 marker expression of the M1 genotype. LPS or IL-1β (10 ng/mL) stimulated-cells increased IL-6, IL-1β and CCL5 mRNA expression and decreased IL-10 mRNA expression, actions that were counteracted by administering this flavonoid to the medium. Together, these activities were associated with the neuroprotective effect of this secondary metabolite [79].
After 22 days of a single injection of LPS (5 μg/5 μL) into the substantia nigra of rats, body weight and behavioral parameters were measured. Subsequently, the levels of inflammatory and oxidative markers, such as TNF-α, IL-1β, IL-6 and level of nitrite, MDA, and SOD, were evaluated in the striatum of these rats. NF-κB and Nrf-2 were measured by immunohistochemical analysis. The results indicated that these parameters were positively modulated by (12) oral administration at doses of 2.5 and 50 mg/kg [80].
Catechin (13). From the flavan-3-ol family, it is widely recognized due its high concentration in tea leaves as well as its antioxidant and anti-inflammatory properties, among others that have been widely studied. In this review, we will mention a little information about the (13) effect on neuroinflammation, just as a background to highlight that polyphenolic compounds represent natural active compounds, with neuroprotective potential.
Stimulation of microglial cells with toxic agents such as LPS and Aβ (amyloid-β) peptide triggers their activation to produce inflammatory mediators, contributing to neurodegeneration and cell death. BV-2 cell culture activated with amyloid (5 µM of Aβ (1-42) for 12 h, leads to the production and overexpression of cytokines such as TNF-α, IL-1β, and IL-6 via regulation of NF-κB signaling pathway. iNOS expression levels were also increased. These damage variables were inhibited with (13) application (10 µM) in the culture medium [81].

4.3. Terpenes

Terpenes, terpenoids or isoprenoids, are grouped into natural product categories based on their structure and pathways for their biosynthesis. All terpenes are derived from the 5-carbon precursor compounds dimethylallyl diphosphate (DMAPP) and its isomer isopentenyl diphosphate (IPP), and exist as single unit hemiterpene (5C) to mono- (C10), sesqui- (C15), di- (C20), sester- (C25), tri- (C30), tetra- (C40) to polyterpenes of > C40–C5 × 103–4 units. They are components of both the primary and secondary metabolism in cells. To date, tens of thousands of terpenes compounds have been identified, the majority encountered in organisms from the plant kingdom [82]. Many plant terpenes have found fortuitous uses in medicine, and the terpenes family of natural products has been a valuable source of medical discoveries [83].

4.3.1. Terpenes and Neuroinflammation

Fourteen terpenes isolated from Lychee seeds (Litchi chinensis Sonn.) showed inhibitory effects on LPS-induced NO production in BV-2 cells, as they reduced COX-2, iNOS and IL-6 expression [84].

4.3.2. Agave Terpenes

Several terpenes have been identified in extracts of Agave species. For example, α-linalool, α-terpinene, p-cymene, limonene, β-trans-ocimene, linalool, α-terpineol, geraniol and trans-nerolidol have been identified in A. salmiana and A. angustifolia extracts. In addition to the aforementioned terpenes, other terpenes have also been reported in A. tequilana Weber such as β-cis-ocimene, sabinene, 2,4,6-octatriene, 4-terpineol, α-terpineol, nerol, bornyl formate, α-cubebene, copaene, anastreptene, bergamotene, β-farnesene, 1,2,3,4-tetrahydronaphthalene, germacrene, α-curcumene, α-muurolene, α-bisabolene, cadinene, α-spirovetivene, cedrol, trans-nerolidol, cadalene, cadinol, patchouli alcohol and α-bisabolol [85]. There are many studies that have demonstrated various pharmacological activities such as the anti-inflammatory properties. However, when we ran the query for each one of these terpenic compounds found in Agave species, plus the word “anti-neuroinflammatory”, no results were found.
The variety of biological activities reviewed in the previous paragraphs emphasizes the potential use of terpenes and flavonoids in the treatment of disorders associated with neuroinflammation. It is important to realize that the lack of reports of anti-neuroinflammatory activity of these compounds in species of Agave genus does not detract from the novelty of the present study; on the contrary, the pharmacological background of these compounds in other species allows us to propose new research strategies focused on evaluating the biological anti-neuroinflammatory activity of the by-products of this genus species and their possible employment in the associated treatment of some disorders.

4.4. Saponins

Saponins are a heterogeneous group of glycosides that are widely distributed in plants of agricultural importance, particularly legumes [86]. They comprise a sugar moiety (glucose, arabinose, galactose, glucuronic acid, xylose, rhamnose or methyl pentose) linked to a non-polar aglycone (sapogenin) and are classified as steroids (C27) and triterpenoids (C30) [87].

4.4.1. Saponins from Agave Species

They were first reported in the Agave genus in 1932 [88]. Since then, saponin and sapogenin constituents have been identified in approximately 50 species of this genus [89,90,91] and they are one of the most abundant compounds in Agave plants [3]. Steroidal sapogenins such as spirostanol-type and cholestane-type (only Agavegenin D) have been isolated from Agave leaves, flowers, callus cultures and rhizomes. Sapogenins with furostanol and furospirostanol skeleta have not been discovered in this genus yet [92].
There are many studies in which several sapogenins have been isolated in various species of Agave; among these are Hecogenin, Sisalagenin, Neohecogenin, Gloriogenin, Manogenin, 9-Dehydrohecogenin, 9-Dehydromanogenin, Botogenin, Gentrogenin, Kammogenin, Hainangenin, Hongguanggenin, Chlorogenin, Agavegenin A, Tigogenin, Neotigogenin, Gitogenin, Neotigogenone, Smilagenin, Sarsasapogenin, Ruizgenin, Diosgenin, Yamogenin, Yuccagenin, Agavegenin B, Agavegenin C, Agavegenin D. Furthermore, there are also reports of steroidal saponis such as Agavoside A, Chlorogenin 3-O-β-D-glucopyranoside, Chlorogenin 6-O-β-D-glucopyranoside, Agamenoside C, tigogenin 3-O-β-D-glucopyranoside, Agamenoside I among others. A great variety of Spirostanol diglycosides have also been reported, among which are Agavoside B and Cantalasaponin-1 and other spirostanol triglycosides, tetraglycosides, hexaglycosides and hexaglycosides, all of them with diverse biological activities [92].
Steroidal saponins are effective therapeutic options to combat inflammatory diseases because they are able to act directly on pro-inflammatory cytokines such as TNF-α and IL-6, inhibit the action of macrophages and act on the arachidonic acid (AA) pathway reducing COX-2 and PGE2 activity. They are receiving much attention from chemists and biologists for new drug discovery because they are strongly associated with anticarcinogenic, hepatoprotective, immunomodulatory properties, but especially anti-inflammatory and anti-neuroinflammatory activities in animal and in vitro models, since they are found in abundance in plants, which means that access to this type of drug would be much easier for the general population [93,94].

4.4.2. Saponins and Neuroinflammation

It is well documented that saponins are neuroprotective compounds [95]. Gingsenosides Rg1 (14) and Re (15) exert neuroprotective effects in scopolamine-induced cognitive impairment, increasing choline acetyltransferase activity in rats [96], mice [97] and in vitro [98]. Compound (14) reduced ROS and cytotoxicity by downregulating the pro-apoptotic protein Bad, which counteracts oxidative stress, resulting in neuroprotection of LPS-treated cells [99]. Compound (14) also alleviates oxidative stress after ischemia/reperfusion (I/R)-induced neuronal injury by inhibiting miR-144 activity and blocking the Nrf2/ARE pathway at the post-translational level [100], promoting cerebral angiogenesis by increasing VEGF expression through the PI3K/Akt/mTOR signaling pathway after ischemic stroke. [101]. In addition, BV2 microglial cells pretreated with (15) exerted neuroprotective effects against LPS-induced neuroinflammation [102]. Compound (15) exhibited a potent neuroprotective effect against neuroinflammation in a murine model by reducing motor neuron death and decreasing pro-inflammatory cytokine TNF-α expression [103].
Pseudoginsenoside PF11 inhibited LPS-induced neuroinflammation in N9 microglia through the inhibition of TLR4 activation, NF-κB, iNOS, COX-2 expression and PGE2, IL-1β, IL-6 and TNF-α release [104].
Aster saponin B obtained from Aster tataricus L. root methanolic extract showed more than 50% inhibition of LPS-induced NO release in RAW264.7 cells and suppressed LPS-enhanced iNOS and COX-2 enzyme expression, resulting in the inhibition of pro-inflammatory mediators such as NO and PGE2. It also suppressed the phosphorylation and degradation of IκB in the cytoplasm. Therefore, the anti-inflammatory activity was exerted by inhibiting NF-κB activity in LPS-treated RAW264.7 cells [105].
Based on the review of the literature, many saponins have been isolated from the species of the Agave genus, however there is only one study that directly assayed a saponin extracted from Agave against neuroinflammation. In that study, the steroidal saponin cantalasaponin-1, isolated from the leaves of Agave americana, decreased the LPS-induced proinflammatory cytokines TNF-α and IL-6 and increased the anti-inflammatory cytokine IL-10 [15]. Considering that saponins are natural compounds with significant anti-neuroinflammatory activity [106], it is important to carry out more chemical and pharmacological studies in order to propose Agaves as medicinal plants that may be useful in the treatment of neuroinflammatory disorders.
In recent years, scientists have evaluated and discovered many medicinal plants and natural products. Many of them have been shown to be able to improve dementia treatment with fewer side effects than conventional drugs and are considered potential anti-neuroinflammatory drugs.the . Since the process of neuroinflammation may be the starting point of several neurodegenerative pathologies, a large number of natural products have been investigated for their ability to act as anti-inflammatories, especially in brain. In order to provide a complete perspective, in the following chapter, we will review some general aspects of some processes involved in neuroinflammation.

5. Neuroinflammation

Inflammation of the central nervous system (CNS), known as neuroinflammation, is a brain process that involves the blood–brain barrier, glia, and neurons and plays a key role in the pathogenesis of various neurodegenerative disorders such as Parkinson’s and Alzheimer’s diseases (AD) [106,107]. Collectively, these neuroinflammatory disorders and their sequelae contribute extensively to mortality and morbidity in the world with a significant healthcare cost [108]. Neuroinflammation can be triggered by various biological mechanisms including infection, trauma, ischemia, toxins, oxidative stress and glial reactions [109,110] and could be initiated in the periphery or within the brain [111].
The process is marked by the production of pro-inflammatory cytokines, including IL-1β, IL-6, IL-18 TNF-α, chemokines such as C-C motif chemokine ligand 1 (CCL1), CCL5 and C-X-C motif chemokine ligand 1 (CXCL1), small-molecule messengers, including prostaglandins, nitric oxide (NO), reactive oxygen species by innate immune cells in the CNS as well as increased levels of these molecules modulating the blood–brain barrier (BBB) [107].

Neuroinflammatory Molecules

Cytokines are large proteins (15–25 kDa) that are mainly released from immune cells such as monocytes, macrophages and lymphocytes, in addition to microglia and astrocytes. They are activated during situations in which inflammation, infection and/or immunological alterations occur [112]. They are classified into two groups—pro-inflammatory and anti-inflammatory cytokines—which promote and inhibit inflammatory responses, respectively [113].
Among the pro-inflammatory cytokines, IL-1β, IL-6 and TNF-α are the most studied. IL-4 and IL-10 are well known anti-inflammatory cytokines. Under normal physiological conditions, pro-inflammatory cytokines are present at low levels [114], but in pathological conditions like infection or trauma, they can increase by up to 100-fold. Neuroinflammatory mediators such as cytokines and prostaglandins are important in the development of neurodegenerative diseases [115].
Initial release of cytokines can trigger the subsequent production of signaling molecules, as evidenced by IL-6, which activates T cells and stimulates the production of other inflammatory markers, such as C-reactive protein (CRP) and fibrinogen. Upon binding to the extracellular TNF-1 receptor, TNF-α sets in motion a series of signaling cascades that affect the transcription of genes related to apoptosis and neurodegeneration. IL-1β, when bound to the IL-1 receptor complex, is also a critical initiator of a number of signal transduction cascades, specifically the mitogen-activated protein kinase (MAPK) pathways, which result in many proinflammatory responses and the production of IL-8 and IL-6 [116].
Chemokines are small chemotactic cytokines that also play a role in neuroinflammation. Although they have very low physiological concentrations in the CNS, certain chemokines, such as monocyte chemoattractant protein-1, are strongly up-regulated in chronic neuroinflammation. These molecules are involved in the regulation and chemotaxis of some CNS cells such as astrocytes and microglia in response to an inflammatory stimulus and may also alter neuronal function and adversely affect neurogenesis [117].
The cyclooxygenase enzyme (COX) converts arachidonic acid into eicosanoid groups such as prostaglandins and thromboxane, and has various inflammatory functions. The pathways of its two common isoforms COX-1 and COX-2 are increasingly associated with neuroinflammation and neurodegeneration and COX inhibitors, such as nonsteroidal anti-inflammatory drugs (NSAIDs), have several therapeutic potentials [118].

6. CNS Cells

The CNS is populated by two broad categories of cells, neurons and neuroglial cells. The neuroglial cells include microglia, astrocytes, oligodendrocytes, and ependymal cells. The innate immune cells involved in neuroinflammation are primarily microglia and astrocytes but capillary endothelial cells and infiltrating blood cells also contribute to neuroinflammation, especially when the blood–brain barrier sustains biochemical or mechanical damage [119].

6.1. Blood–Brain Barrier (BBB)

The BBB is part of the neurovascular unit, which is essential for brain health and function. It is a dynamic interface between the peripheral circulation and the central nervous system, constituting a semi-permeable and highly selective structural and chemical barrier. Endothelial cells are the main structure of this barrier. Unlike other vascular endothelial cells lining peripheral blood vessels, cerebral microvascular endothelial cells exhibit distinctive morphological, structural, and functional characteristics that differentiate them from other vascular endothelia. These include the following: (1) the expression of tight junctions (TJs) that seal paracellular pathways between adjacent endothelial cells, thereby preventing the unregulated passage of polar (water-soluble) molecules between the blood and brain; (2) the absence of fenestrations; (3) the lack of pinocyticsynapto- activity and expression of active transport mechanisms to regulate the passage of essential molecules (including nutrients and essential amino acids), while blocking the passage of potentially unwanted substances (both endogenous and xenobiotic) [120].
Under optimal physiological conditions, the BBB ensures a highly stable internal brain environment and prevents foreign objects (such as microorganisms, toxins, etc.) from invading the brain tissue. However, under certain conditions such as brain edema, brain tumor, ionizing radiation injury, inflammation and other pathological conditions, BBB permeability increases, triggering acute or chronic neuroinflammatory processes that lead to neurodegenerative disorders [121].

6.2. Microglia

Microglia cells are the resident immune (macrophages) cells of the brain, and play a crucial role in the neuroinflammation process. Unlike neurons and other neuroglial cells which are derived from the neuroectoderm, microglia originate as primitive macrophages from the embryonic yolk sac and reach the CNS via the circulatory system [122]. Microglial cells are responsible for maintaining the tissue homeostasis of neuronal cells, which are sensitive to cell environment changes. In a non-stimulated state, the resting microglia feature a highly ramified appearance, which is in sharp contrast to the amoeboid reactive cells. They are characterized by rapid proliferation, the production and secretion of a wide spectrum of cytokines, chemokines and other immune mediators in response to insult [123].
In response to cytokines and other signaling molecules from acute inflammation, microglia transform from a ramified, inactivated state to an activated phagocytic one, releasing pro-inflammatory mediators in the process. They are activated in two ways: firstly, as a response to neural death caused by inflammatory neural injury, and secondly in response to viral or bacterial toxins. Microglial activation under homeostatic physiological conditions aims to constantly monitor the environment through long cytoplasmic extensions to activate the immune system and tissue repair in the CNS, as intrinsic and extrinsic factors can lead to disastrous dysregulation of microglial cells, resulting in severe CNS pathologies [124].
In terms of chronic neuroinflammation, microglia can remain activated for long periods releasing cytokines and neurotoxic molecules that contribute to long-term neurodegeneration by promoting their migration during the initial phase of neurodegeneration, attracted by factors secreted by cells that have already been injured. They are recruited to the damaged or infected area in order to participate in degenerative or regenerative processes, to engulf microorganisms and cell debris, and promote production of neuroinflammatory and neurotoxic molecules such as IL-1β, IL-6, TNF-α, prostaglandins, and nitric oxide (NO) [124]. Moreover, they activate FN-ĸB transcription factor to induce the expression of cytokine and chemokine genes, leading to neural cell death mediated by inflammatory processes [125].

6.3. Astrocytes

Astrocytes are the other glial cells within the CNS, which provide physiological functions related to CNS homeostasis such as regulation of blood flow, energy metabolism, immune defense, neurotransmission and neurogenesis. They also are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters [126]. When any CNS damage occurs, such as neurodegeneration, trauma or infection, these cells are activated by a process called reactive astrogliosis, which express membrane receptors to recognize pathogens, depending on the intensity of the inflammatory process and damage [127].
An activated astrocyte secretes and/or responds to several cytokines, altering the neural cell state, microglia neighboring cells, and other astrocytes. Activation of these cells results in the synthesis and segregation of cytokines such as IL-1β, IL-6, TNF-α and transforming growth factor-β1 (TGF-β1). They promote or inhibit proinflammatory as well as anti-inflammatory gene expression [128].
With the term “neuroinflammation”, we refer to the inflammatory response in the CNS, as a result of neuronal injury and in related disorders that share a common neuroinflammatory pathology. BBB endothelium, microglia and astrocytes are key cellular drivers and regulators of neuroinflammation. Under physiological conditions, they are important for neurotransmission and synaptic homeostasis. However, an overexpressed inflammatory response mediated by these cells results in further cytokines production and a wide variety of inflammatory molecules that trigger synapto-toxicity and neurodegeneration in a self-reinforcing manner. Suppression of neuroinflammation with NSAIDs was one of the earliest approaches to treat various related disorders. However, despite its strong therapeutic rationale, previous clinical trials investigating compounds with anti-inflammatory properties, including NSAIDs, failed to achieve primary efficacy endpoints. In fact, there are bibliographic reports mentioning that NSAIDs are not the best pharmacological option [129], due to the absence of clear effects on the inflammatory response inhibition, which leads to neurodegeneration, as demonstrated by the lack of significant results of these drugs on cognitive impairment and the severity of AD in long term.
Current treatments can address individual symptoms for some disorders, but there is no known cure for any neuroinflammatory disorder. While the cause, pathology and symptoms of these disorders are extremely diverse, they all share a core inflammatory component [130].
Recent drug development projects were based on the emergence of new potential targets in different genomic and proteomic studies. Despite all of the drug development efforts undertaken, the number of successful drugs and novel targets have been lower than expected during the past few decades [130,131]. Therefore, it is necessary to develop other anti-inflammatory drugs, such as those derived from medicinal plants, efficient in combating neuroinflammatory processes.

7. Conclusions

The main goal of neuroinflammatory disease therapies is to stop or slow down the progression of the pathology. Current treatments include antioxidants and NSAIDs, among other substances; however, they have a modest clinical effect in the treatment of symptoms and do not prevent the progression of dementia, even though they may improve some cognitive functions.
Currently, there are data that species of the Agave genus have pharmacological properties as immunomodulators in different biological systems; however, only one study has demonstrated their ability to reduce neuroinflammation, which is complicated by the presence of the blood–brain barrier.
More pharmacological and chemical studies are needed to evaluate the possible therapeutic effects of the compounds present in the leaves of Agave species, specifically on neurodegenerative conditions, in which they exert beneficial functions in biological phenomena linked to inflammation and oxidative stress, among other factors.
Agave species, besides being an important source of pharmacologically active products and a cultural staple food in Mexico, have the potential to become a sustainable resource for the production of phytopharmaceuticals. Through research, mainly using the leaves, which are usually discarded and considered as by-products, despite possessing a wide variety of components such as saponins, their immense potential as a raw material for the development of new therapeutic resources can finally be implemented to elucidate possible applications in neuroinflammatory conditions.

Author Contributions

All authors contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are grateful for the support of the Health Research Fund (FIS/IMSS), for the project number FIS/IMSS/PROT/G8/1820 and registration number: R-2015-1702-4.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank the participating institutions for their support in the development of our scientific work (IMSS, UAEM, IPN).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pérez, A.J.; Calle, J.M.S.; Simonet, A.M.; Guerra, J.O.; Stochmal, A.; Macías, F.A. Bioactive steroidal saponins from Agave offoyana flowers. Phytochemistry 2013, 95, 298–307. [Google Scholar] [CrossRef] [PubMed]
  2. García-Mendoza, A.; Galván, R. Riqueza de las familias Agavaceae y Nolinaceae en México. Bol. Soc. Bot. México. 1995, 56, 7–24. [Google Scholar] [CrossRef]
  3. López-Romero, J.C.; Ayala-Zavala, J.F.; Peña-Ramos, E.A.; Hernández, J.; González-Ríos, H. Antioxidant and antimicrobial activity of Agave angustifolia extract on overall quality and shelf life of pork patties stored under refrigeration. J. Food Sci. Technol. 2018, 55, 4413–4423. [Google Scholar] [CrossRef] [PubMed]
  4. Statista. Mexico: Distilled Agave-Based Beverages Production Value 2013–2020. 2022. Available online: https://www.statista.com/statistics/717349/distilled-Agave-based-beverage-production-value-in-mexico/ (accessed on 16 May 2022).
  5. Xiong, L.; Maki, M.; Guo, Z.; Mao, C.; Qin, W. Agave biomass is excellent for production of bioethanol and xylitol using Bacillus strain 65S3 and Pseudomonas strain CDS3. J. Biobased Mater. Bioenergy 2014, 8, 422–428. [Google Scholar] [CrossRef]
  6. Bermúdez-Bazán, M.; Castillo-Herrera, G.A.; Urias-Silvas, J.E.; Escobedo-Reyes, A.; Estarrón-Espinosa, M. Hunting bioactive molecules from de Agave genus: An update on extraction and biological potential. Molecules 2021, 26, 6789. [Google Scholar] [CrossRef]
  7. Wang, J.; Chio, C.; Chen, X.; Su, E.; Cao, F.; Jin, Y.; Qin, W. Efficient saccharification of Agave biomass using Aspergillus niger produced low-cost enzyme cocktail with hyperactive pectinase activity. Bioresour. Technol. 2018, 272, 26–33. [Google Scholar] [CrossRef]
  8. Blomberg, L. Tequila, Mezcal y Pulque: Lo Auténtico Mexicano, 1st ed.; Planeta Mexicana, S.A. de C.V. of México: Mexico City, Mexico, 2000; pp. 23–57. ISBN 9681332695. [Google Scholar]
  9. Monroy, C.; Castillo, P. Plantas Medicinales Utilizadas en el Estado de Morelos, 1st ed.; Universidad Autónoma del Estado de Morelos: Cuernavaca, Mexico, 2007; pp. 265–266. ISBN 968-878-277-7. [Google Scholar]
  10. Lozoya, X.; Lozoya, M. Flora Medicinal de Mexico. Volume 1: Plantas Indígenas; Instituto Mexicano del Seguro Social Mexico, D.F., Ed.; Instituto Mexicano del Seguro Social Mexico D.F.: Torreon, Coahuila, Mexico, 1982; pp. 32–48. [Google Scholar]
  11. Ahumada-Santos, Y.P.; Montes-Avila, J.; Uribe-Beltrán, M.J.; Díaz-Camacho, S.P.; López-Angulo, G.; Vega-Aviña, R.; Delgado-Vargas, F. Chemical characterization, antioxidant and antibacterial activities of six Agave species from Sinaloa, Mexico. Ind. Crops Prod. 2013, 49, 143–149. [Google Scholar] [CrossRef]
  12. Santos-Zea, L.; Rosas-Pérez, A.M.; Leal-Díaz, A.M.; Gutiérrez-Uribe, J.A. Variability in Saponin Content, Cancer Antiproliferative Activity and Physicochemical Properties of Concentrated Agave Sap. J. Food Sci. 2016, 81, H2069–H2075. [Google Scholar] [CrossRef]
  13. Monterrosas-Brisson, N.; Ocampo, M.L.; Jiménez-Ferrer, E.; Jiménez-Aparicio, A.R.; Zamilpa, A.; González-Cortazar, M.; Tortoriello, J.; Herrera-Ruiz, M. Anti-inflammatory activity of different Agave plants and the compound cantalasaponin-1. Molecules 2013, 18, 8136–8146. [Google Scholar] [CrossRef]
  14. Hernández-Valle, E.; Herrera-Ruiz, M.; Salgado, G.R.; Zamilpa, A.; Ocampo, M.L.; Aparicio, A.R.; Tortoriello, J.; Jiménez-Ferrer, E. Anti-inflammatory effect of 3-O-[(6’-O-Palmitoyl)-β-Dglucopyranosyl Sitosterol]), from Agave angustifolia on ear edema in mice. Molecules 2014, 19, 15624–15637. [Google Scholar] [CrossRef] [Green Version]
  15. Herrera-Ruiz, M.; Jiménez-Ferrer, E.; Tortoriello, J.; Zamilpa, A.; Alegría-Herrera, E.; Jiménez-Aparicio, A.R.; Arenas-Ocampo, M.L.; Martínez-Duncker, I.; Monterrosas-Brisson, N. Anti-neuroinflammatory effect of Agaves and cantalasaponin-1 in a model of LPS-induced damage. Nat. Prod. Res. 2019, 14, 1–4. [Google Scholar] [CrossRef] [PubMed]
  16. Ransohoff, R.M. How neuroinflammation contributes to neurodegeneration. Science 2016, 353, 777–783. [Google Scholar] [CrossRef]
  17. Calsolaro, V.; Edison, P. Neuroinflammation in Alzheimer’s disease: Current evidence and future directions. Alzheimer Dement. 2016, 12, 719–732. [Google Scholar] [CrossRef]
  18. Chen, W.W.; Zhang, X.; Huang, W.J. Role of neuroinflammation in neurodegenerative diseases (Review). Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef] [PubMed]
  19. Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 1–12. [Google Scholar] [CrossRef] [PubMed]
  20. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [PubMed]
  21. García Mendoza, A.J. Ficha técnica de Agave ornithobroma. In Revisión de las Agavaceae (sensu stricto), Crassulaceae y Liliaceae Incluidas en el PROY-NOM-059-ECOL-2000; Bases de datos SNIB-CONABIO. Proyecto No. W020; Jardín Botánico, Instituto de Biología, Universidad Nacional Autónoma de México: Mexico City, Mexico, 2003; pp. 1–4. [Google Scholar]
  22. González-Elizondo, M.; Galván-Villanueva, R.; López-Enríquez, I.; Reséndiz-Rojas, L.; González-Elizondo, M.S. Agaves Magueyes, Lechuguillas y Notas del Estado de Durango y sus Alrededores, 1st ed.; CIIDIR Unidad Durango, Instituto Politécnico Nacional: Mexico City, Mexico, 2009; pp. 1–159. ISBN 978-970-95117-1-8. [Google Scholar]
  23. Argueta, V.A.; Cano, L.M.; Rodarte, M.E. Atlas de las Plantas de la Medicina Tradicional Mexicana, Volume I, II y III; Instituto Nacional Indigenista: Mexico City, Mexico, 1994; p. 1786. ISBN 9682961327. [Google Scholar]
  24. García-Regalado, G. Plantas Medicinales de Aguascalientes, 2nd ed.; Universidad Autónoma de Aguascalientes Mexico: Aguascalientes, Mexico, 2015; pp. 266–276. ISBN 978-607-8359-83-7. [Google Scholar]
  25. Pérez-Zavala, M.L.; Hernández-Arzaba, J.C.; Bideshi, D.K.; Barboza-Corona, J.E. Agave: A natural renewable resource with multiple applications. J Sci. Food Agric. 2020, 100, 5324–5333. [Google Scholar] [CrossRef]
  26. Rendón-Salcido, L.A.; Colunga-GarcíaMarín, P.; Barahona-Pérez, L.F.; Pimienta-Barrios, E.; Magdub-Méndez, A.; Larqué-Saavedra, A. Sugars and alcoholic by products from henequén (Agave fourcroydes) as influenced by plant age and climate. Rev. Fitotec. Mex. 2009, 32, 39–44. [Google Scholar]
  27. United States Department of Agriculture. Plants Database. Available online: https://plants.usda.gov/home/plantProfile?symbol=AGUT (accessed on 3 June 2022).
  28. Southwest Desert Flora. “Plants Profile: Agave Schottii Engelm. Schott’s Century Plant”. Available online: http://southwestdesertflora.com/WebsiteFolders/All_Species/Agavaceae/Agave%20schottii,%20Schott’s%20Century%20Plant.html (accessed on 4 May 2022).
  29. Cortés, A.J.; Sánchez-Mendoza, E.; Zamilpa, A.; González-Cortazar, M.; Herrera-Ruiz, M.; Almanza-Pérez, J.C.; Terán-Cabanillas, E.; Renaud, C.; Domínguez-Ramírez, L.; Montiel-Arcos, E.; et al. Steroidal saponin from Agave marmorata Roezl modulates inflammatory response by inhibiting NF-κB and AP-1. Nat. Prod. Res. 2020, 36, 1–6. [Google Scholar] [CrossRef]
  30. Puente-Garza, C.A.; Gutiérrez-Mora, A.; García-Lara, S. Micropropagation of Agave salmiana: Means to production of antioxidant and bioactive principles. Front. Plant Sci. 2015, 6, 1–9. [Google Scholar] [CrossRef]
  31. Ben-Hamissa, A.M.; Seffen, M.; Aliakbarian, B.; Casazza, A.A.; Perego, P.; Converti, A. Phenolics extraction from Agave americana (L.) leaves using high-temperature, high-pressure reactor. Food Bioprod. Process. 2012, 90, 17–21. [Google Scholar] [CrossRef]
  32. López, M.; Mancilla, N.; Mendoza, G. Molecular structures of fructans from Agave tequilana Weber var azul. J. Agric. Food Chem. 2003, 51, 7835–7840. [Google Scholar] [CrossRef] [PubMed]
  33. Arreola-Vargas, J.; Ojeda-Castillo, V.; Snell-Castro, R.; Corona-González, R.I.; Alatriste-Mondragón, F.; Méndez-Acosta, H.O. Methane production from acid hydrolysates of Agave tequilana bagasse: Evaluation of hydrolysis conditions and methane yield. Bioresour. Technol. 2015, 181, 191–199. [Google Scholar] [CrossRef] [PubMed]
  34. Gutiérez-Nava, Z.J.; Jiménez-Aparicio, A.R.; Herrera-Ruiz, M.L.; Jiménez-Ferrer, E. Immunomodulatory effect of Agave tequilana evaluated on an autoimmunity like-SLE model induced in Balb/c mice with pristane. Molecules 2017, 22, 848. [Google Scholar] [CrossRef] [PubMed]
  35. Herrera-Ruiz, M.; Gutiérrez-Nava, Z.J.; Trejo-Moreno, C.; Zamilpa, A.; González-Cortazar, M.; Jiménez-Aparicio, A.; Jiménez-Ferrer, E. Agave tequilana counteracts chronic hypertension and associated vascular damage. J. Med. Food. 2021, 25, 443–455. [Google Scholar] [CrossRef]
  36. García, M.D.; Quílez, A.M.; Sáenz, M.T.; Martínez-Domínguez, M.E.; de la Puerta, R. Anti-inflammatory activity of Agave intermixta Trel. and Cissus sicyoides L., species used in the Caribbean traditional medicine. J. Ethnopharmacol. 2000, 71, 395–400. [Google Scholar] [CrossRef]
  37. Peana, A.T.; Moretti, M.D.; Manconi, V.; Desole, G.; Pippia, P. Anti-inflammatory activity of aqueous extracts and steroidal sapogenins of Agave americana. Planta Med. 1997, 63, 199–202. [Google Scholar] [CrossRef]
  38. Yokosuka, A.; Mimaki, Y.; Kuroda, M.; Sashida, Y. A new steroidal saponin from the leaves of Agave americana. Planta Med. 2000, 66, 393–396. [Google Scholar] [CrossRef]
  39. Jin, J.M.; Liu, X.K.; Yang, C.R. A new C-27 steroidal saponins from fermented leaves of Agave Americana. Zhongguo Zhong Yao Za Zhi 2002, 27, 431–434. [Google Scholar]
  40. Jin, J.M.; Zhang, Y.J.; Yang, C.R. Four new steroid constituents from the waste residue of fibre separation from Agave americana leaves. Chem. Pharm. Bull. 2004, 52, 654–658. [Google Scholar] [CrossRef]
  41. Nasri, S.; Ben-Salem, H. Effect of oral administration of Agave americana or Quillaja saponaria extracts on digestion and growth of Barbarine female lamb. Livest. Sci. 2012, 147, 59–65. [Google Scholar] [CrossRef]
  42. Quintans, J.S.; Barreto, R.S.; de Luca, W., Jr.; Villarreal, C.F.; Kaneto, C.M.; Soares, M.B.; Branco, A.; Almeida, J.R.; Taranto, A.G.; Antoniolli, A.R.; et al. Evidence for the involvement of spinal cord-inhibitory and cytokines-modulatory mechanisms in the anti-hyperalgesic effect of hecogenin acetate, a steroidal sapogenin-acetylated, in mice. Molecules 2014, 19, 8303–8316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Misra, A.K.; Varma, S.K.; Kumar, R. Anti-inflammatory effect of an extract of Agave americana on experimental animals. Pharmacogn. Res. 2018, 10, 104–108. [Google Scholar]
  44. Tinto, W.F.; Simmons-Boyce, J.L.; McLean, S.; Reynolds, W.F. Constituents of Agave americana and Agave barbadensis. Fitoterapia 2005, 76, 594–597. [Google Scholar] [CrossRef]
  45. Simmons-Boyce, J.L.; Tinto, W.F.; McLean, S.; Reynolds, W.F. Saponins from Furcraea selloa var. marginata. Fitoterapia 2004, 75, 634–638. [Google Scholar] [CrossRef]
  46. Allsopp, P.; Possemiers, S.; Campbell, D.; Oyarzábal, I.S.; Gill, C.; Rowland, I. An exploratory study into the putative prebiotic activity of fructans isolated from Agave angustifolia and the associated anticancer activity. Anaerobe 2013, 22, 38–44. [Google Scholar] [CrossRef]
  47. Ohtsuki, T.; Koyano, T.; Kowithayakorn, T.; Sakai, S.; Kawahara, N.; Goda, Y.; Yamaguchi, N.; Ishibashi, M. New chlorogenin hexasaccharide isolated from Agave fourcroydes with cytotoxic and cell cycle inhibitory activities. Bioorg. Med. Chem. 2004, 12, 3841–3845. [Google Scholar] [CrossRef]
  48. Yokosuka, A.; Jitsuno, M.; Yui, S.; Yamazaki, M.; Mimaki, Y. Steroidal glycosides from Agave utahensis and their cytotoxic activity. J. Nat. Prod. 2009, 72, 1399–1404. [Google Scholar] [CrossRef]
  49. Yokosuka, A.; Mimaki, Y. Steroidal saponins from the whole plants of Agave utahensis and their cytotoxic activity. Phytochemistry 2009, 70, 807–815. [Google Scholar] [CrossRef]
  50. Fujino, T.; Yokosuka, A.; Higurashi, H.; Yokokawa, R.; Sakurai, R.; Harashima, W.; Miki, Y.; Fujiwara, Y.; Mimaki, Y.; Hayakawa, M. AU-1 from Agavaceae plants causes transient increase in p21/Cip1 expression in renal adenocarcinoma ACHN cells in an miR-34-dependent manner. J. Nat. Med. 2016, 71, 36–43. [Google Scholar] [CrossRef]
  51. Bianchi, E.; Cole, J.R. Antitumor Agents from Agave schottii (Amaryllidaceae). J. Pharm. Sci. 1969, 58, 589–591. [Google Scholar] [CrossRef] [PubMed]
  52. Corrales-Escobosa, A.R.; Gomez-Ojeda, A.; Wrobel, K.; Alcazar-Magana, A.; Wrobel, K. Methylglyoxal is associated with bacteriostatic activity of high fructose Agave syrups. Food Chem. 2014, 165, 444–450. [Google Scholar] [CrossRef] [PubMed]
  53. Anajwala, C.C.; Patel, R.M.; Dakhara, S.L.; Jariwala, J.K. In vitro cytotoxicity study of Agave americana, Strychnos nuxvomica and Areca catechu extracts using mcf-7 cell line. J. Adv. Pharm. Technol. Res. 2010, 1, 245–252. [Google Scholar] [PubMed]
  54. Padilla-Camberos, E.; Barragán-Álvarez, C.P.; Díaz-Martínez, N.E.; Rathod, V.; Flores-Fernández, J.M. Effects of Agave fructans (Agave tequilana Weber var. azul) on Body Fat and Serum Lipids in Obesity. Plant Foods Hum. Nutr. 2018, 73, 34–39. [Google Scholar] [CrossRef]
  55. Mwale, M.; Masika, P.J. In vivo anthelmintic efficacy of Aloe ferox, Agave sisalana, and Gunnera perpensa in village chickens naturally infected with Heterakis gallinarum. Trop. Anim. Health Prod. 2015, 47, 131–138. [Google Scholar] [CrossRef]
  56. Santos-Cerqueira, G.; dos Santos-e Silva, G.; Rios-Vasconcelos, E.; Fragoso-de Freitas, A.P.; Arcanjo-Moura, B.; Silveira-Macedo, D.; Lopes-Souto, A.; Barbosa-Filho, J.M.; deAlmeida-Leal, L.K.; de Castro-Brito, G.A.; et al. Effects of hecogenine and its possible mechanism of action on experimental models of gastric ulcer in mice. Eur. J. Pharmacol. 2012, 683, 260–269. [Google Scholar] [CrossRef]
  57. BritoCortez-Lima, N.; DantasCavalcante-Abreu, R.N.; Barbosa-Filho, J.M.; Leal, L.K.A.M.; Ribeiro-Honório-Júnior, J.E.; Camelo-Chaves, E.M.; Magalhaes-de Brito, E.; Martins-Dias, L.; Miranda-Sousa, A.; Mendes-Vasconcelos, S.M. P.2.d.013. Evaluation of the action mechanism of hecogenin’s antidepressant effect from Agave sisalana Perrine in mice. Eur. Neuropsychopharmacol. 2012, 22, S275. [Google Scholar] [CrossRef]
  58. Durán, A.G.; Benito, J.; Macías, F.A.; Simonet, A.M. Agave steroidal saponins as potential bioherbicides. Agronomy 2021, 11, 2404. [Google Scholar] [CrossRef]
  59. Maazoun, A.M.; Hamdi, S.H.; Belhadj, F.; Jemâa, J.M.B.; Messaoud, C.; Marzouki, M.N. Phytochemical profile and insecticidal activity of Agave americana leaf extract towards Sitophilus oryzae (L.) (Coleoptera: Curculionidae). Environ. Sci. Poll. Res. Int. 2019, 26, 19468–19480. [Google Scholar] [CrossRef]
  60. Yang, C.R.; Zhang, Y.; Jacob, M.R.; Khan, S.I.; Zhang, Y.J.; Li, X.C. Antifungal Activity of C-27 Steroidal Saponins. Antimicrob. Agents Chemother. 2006, 50, 1710–1714. [Google Scholar] [CrossRef]
  61. Babu, S.; Jayaraman, S. An update on β-sitosterol: A potential herbal nutraceutical for diabetic management. Biomed. Pharmacother. 2020, 131, 110702. [Google Scholar] [CrossRef] [PubMed]
  62. Sun, Y.; Gao, L.; Hou, W.; Wu, J. Β-Sitosterol Alleviates Inflammatory Response via Inhibiting the Activation of ERK/p38 and NF-κB Pathways in LPS-Exposed BV2 Cells. BioMed Res. Int. 2020, 2020, 1–10. [Google Scholar] [CrossRef] [PubMed]
  63. López-Romero, J.C.; Ayala-Zavala, J.F.; González-Aguilar, G.A.; Peña-Ramos, E.A.; González-Ríos, H. Biological activities of Agave by-products and their possible applications in food and pharmaceuticals. J. Sci. Food Agric. 2017, 98, 2461–2474. [Google Scholar] [CrossRef] [PubMed]
  64. Morreeuw, Z.P.; Escobedo-Fregoso, C.; Ríos-González, L.J.; Castillo-Quiroz, D.; Reyes, A.G. Transcriptome-based metabolic profiling of flavonoids in Agave lechuguilla waste biomass. Plant Sci. 2021, 305, 110748. [Google Scholar] [CrossRef]
  65. Shegute, T.; Wasihun, Y. Antibacterial Activity and Phytochemical Components of Leaf Extracts of Agave americana. J. Ex. Pharmacol. 2020, 12, 447–454. [Google Scholar] [CrossRef]
  66. Almaraz-Abarca, N.; Delgado-Alvarado, E.A.; Hernández-Vargas, V.; Ortega-Chávez, M.; Orea-Lara, G.; Leon, A.C.; Avila-Reyes, J.A.; Muniz-Martinez, R. Profiling of phenolic compounds of somatic and reproductive tissues of Agave durangensis Gentry (Agavaceae). Am. J. Appl. Sci. 2009, 6, 1076–1085. [Google Scholar]
  67. Mulati, A.; Zhang, X.; Zhao, T.; Ren, B.; Wang, L.; Liu, X.; Lan, Y.; Liu, X. Isorhamnetin attenuates high-fat and high-fructose diet induced cognitive impairments and neuroinflammation by mediating MAPK and NFκB signalling pathways. Food Funct. 2021, 12, 9261–9272. [Google Scholar] [CrossRef]
  68. Kim, S.Y.; Jin, C.Y.; Kim, C.H.; Yoo, Y.H.; Choi, S.H.; Kim, G.Y.; Yoon, H.M.; Park, H.T.; Choi, Y.H. Isorhamnetin alleviates lipopolysaccharide-induced inflammatory responses in BV2 microglia by inactivating NF-κB, blocking the TLR4 pathway and reducing ROS generation. Int. J Mol. Med. 2019, 43, 682–692. [Google Scholar] [CrossRef]
  69. Zhou, D.; Wei, H.; Jiang, Z.; Li, X.; Jiao, K.; Jia, X.; Hou, Y.; Li, N. Natural potential neuroinflammatory inhibitors from Alhagi sparsifolia Shap. Bioorg. Med. Chem. Lett. 2017, 27, 973–978. [Google Scholar] [CrossRef]
  70. Hajialyani, M.; Hosein-Farzaei, M.; Echeverría, J.; Nabavi, S.M.; Uriarte, E.; Sobarzo-Sánchez, E. Hesperidin as a Neuroprotective Agent: A Review of Animal and Clinical Evidence. Molecules 2019, 24, 648. [Google Scholar] [CrossRef]
  71. Kim, J.; Wie, M.B.; Ahn, M.; Tanaka, A.; Matsuda, H.; Shin, T. Benefits of hesperidin in central nervous system disorders: A review. Anat. Cell Biol. 2019, 52, 369–377. [Google Scholar] [CrossRef]
  72. Muhammad, T.; Ikram, M.; Ullah, R.; Rehman, S.U.; Kim, M.O. Hesperetin, a Citrus Flavonoid, Attenuates LPS-Induced Neuroinflammation, Apoptosis and Memory Impairments by Modulating TLR4/NF-κB Signaling. Nutrients 2019, 11, 648. [Google Scholar] [CrossRef] [PubMed]
  73. Haghmorad, D.; Mahmoudi, M.B.; Salehipour, Z.; Jalayer, Z.; Momtazi-Brojeni, A.A.; Rastin, M.; Kokhaei, P.; Mahmoudi, M. Hesperidin ameliorates immunological outcome and reduces neuroinflammation in the mouse model of multiple sclerosis. J Neuroimmunol. 2017, 302, 23–33. [Google Scholar] [CrossRef] [PubMed]
  74. Yıldız, M.O.; Çelik, H.; Caglayan, C.; Kandemir, F.M.; Gür, C.; Bayav, İ.; Genç, A.; Kandemir, Ö. Neuromodulatory effects of hesperidin against sodium fluoride-induced neurotoxicity in rats: Involvement of neuroinflammation, endoplasmic reticulum stress, apoptosis and autophagy. Neurotoxicology 2022, 90, 197–204. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, C.H.; Zhu, L.L.; Ju, K.F.; Liu, J.L.; Li, K.P. Anti-inflammatory effect of delphinidin on intramedullary spinal pressure in a spinal cord injury rat model. Exp. Ther. Med. 2017, 14, 5583–5588. [Google Scholar] [CrossRef] [PubMed]
  76. Khan, A.; Ali, T.; Rehman, S.U.; Khan, M.S.; Alam, S.I.; Ikram, M.; Muhammad, T.; Saeed, K.; Badshah, H.; Kim, M.O. Neuroprotective Effect of Quercetin Against the Detrimental Effects of LPS in the Adult Mouse Brain. Front. Pharmacol. 2018, 9, 1383. [Google Scholar] [CrossRef]
  77. Lee, B.; Yeom, M.; Shim, I.; Lee, H.; Hahm, D.H. Protective Effects of Quercetin on Anxiety-Like Symptoms and Neuroinflammation Induced by Lipopolysaccharide in Rats. Evid. Based Complement Alternat. Med. 2020, 2020, 1–10. [Google Scholar] [CrossRef]
  78. Tsai, C.F.; Chen, G.W.; Chen, Y.C.; Shen, C.K.; Lu, D.Y.; Yang, L.Y.; Chen, J.H.; Yeh, W.L. Regulatory Effects of Quercetin on M1/M2 Macrophage Polarization and Oxidative/Antioxidative Balance. Nutrients 2021, 14, 67. [Google Scholar] [CrossRef]
  79. Dourado, N.S.; Souza, C.D.S.; de Almeida, M.M.A.; Bispo-da Silva, A.; Dos Santos, B.L.; Silva, V.D.A.; De Assis, A.M.; da Silva, J.S.; Souza, D.O.; Costa, M.F.D.; et al. Neuroimmunomodulatory and Neuroprotective Effects of the Flavonoid Apigenin in in vitro Models of Neuroinflammation Associated With Alzheimer’s Disease. Front Aging Neurosci. 2020, 12, 119–133. [Google Scholar] [CrossRef]
  80. Patel, M.; Singh, S. Apigenin Attenuates Functional and Structural Alterations via Targeting NF-kB/Nrf2 Signaling Pathway in LPS-Induced Parkinsonism in Experimental Rats: Apigenin Attenuates LPS-Induced Parkinsonism in Experimental Rats. Neurotox. Res. 2022, 40, 941–960. [Google Scholar] [CrossRef]
  81. Tang, Y.; Xiong, R.; Wu, A.G.; Yu, C.L.; Zhao, Y.; Qiu, W.Q.; Wang, X.L.; Teng, J.F.; Liu, J.; Chen, H.X.; et al. Polyphenols Derived from Lychee Seed Suppress Aβ (1-42)-Induced Neuroinflammation. Int. J Mol. Sci. 2018, 19, 2109. [Google Scholar] [CrossRef] [PubMed]
  82. Tetali, S.D. Terpenes and isoprenoids: A wealth of compounds for global use. Planta 2018, 249, 1–8. [Google Scholar] [CrossRef] [PubMed]
  83. Bergman, M.E.; Davis, B.; Phillips, M.A. Medically Useful Plant Terpenoids: Biosynthesis, Occurrence, and Mechanism of Action. Molecules 2019, 24, 3961. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, X.; Wang, Y.; Qin, Q.; Wang, Y.; Xu, J.; He, X. Pronounced anti-neuroinflammatory jasmonates and terpenes isolated from lychee seeds. Fitoterapia 2021, 152, 104924. [Google Scholar] [CrossRef] [PubMed]
  85. Peña-Alvarez, A.; Díaz, L.; Medina, A.; Labastida, C.; Capella, S.; Vera, L.E. Characterization of three Agave species by gas chromatography and solid-phase microextraction–gas chromatography–mass spectrometry. J Chromatogr. A 2004, 1027, 131–136. [Google Scholar] [CrossRef]
  86. Savage, G.P. Saponins. In Encyclopedia of Food Sciences and Nutrition, 2nd ed.; Caballero, B., Trugo, L., Finglas, P.M., Eds.; Academic Press: London, UK, 2003; pp. 5095–5097. [Google Scholar]
  87. Juang, Y.P.; Liang, P.H. Biological and Pharmacological Effects of Synthetic Saponins. Molecules 2020, 25, 4974. [Google Scholar] [CrossRef]
  88. Jones, H.D.; Furbeck, G.N.; Colorado, N. Isolation and study of the saponin content of the juice and leaf of the Agave plant, Maguey, manso fino. J. Am. Pharm. Assoc. 1932, 21, 787–793. [Google Scholar]
  89. Marker, R.E.; Wagner, R.B.; Ulshafer, P.R.; Wittbecker, E.L.; Goldsmith, D.P.J.; Ruo, C.H. Isolation and structures of thirteen new steroidal sapogenins. New sources for known sapogenins. J. Am. Chem. Soc. 1943, 65, 1199–1209. [Google Scholar] [CrossRef]
  90. Vázquez-Rodríguez, B.; Gutiérrez-Uribe, J.A.; Guajardo-Flores, D.; Santos-Zea, L. Microencapsulation of steroidal saponins from Agave sap concentrate using different carriers in spray drying. Food sci. Technol. Int. 2021, 10820132211049949, Advance online publication. [Google Scholar] [CrossRef]
  91. Simonet, A.M.; Durán, A.G.; Pérez, A.J.; Macías, F.A. Features in the NMR spectra of the aglycones of Agave spp. saponins. HMBC method for aglycone identification (HMAI). Phytochem. Anal. 2021, 32, 38–61. [Google Scholar] [CrossRef]
  92. Sidana, J.; Singh, B.; Sharma, O.P. Saponins of Agave: Chemistry and bioactivity. Phytochemistry 2016, 130, 22–46. [Google Scholar] [CrossRef] [PubMed]
  93. Sparg, S.G.; Light, M.E.; van Staden, J. Biological activities and distribution of plant saponins. J. Ethnopharmacol. 2004, 94, 219–243. [Google Scholar] [CrossRef] [PubMed]
  94. Passos, F.; Araújo-Filho, H.G.; Monteiro, B.S.; Shanmugam, S.; Araújo, A.; Almeida, J.; Thangaraj, P.; Júnior, L.; Quintans, J. Anti-inflammatory and modulatory effects of steroidal saponins and sapogenins on cytokines: A review of pre-clinical research. Phytomedicine 2022, 96, 153842. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, A.; Xu, X.; Lin, J.; Cui, X.; Xu, R. Neuroprotection by Saponins. Phytother. Res. 2014, 29, 187–200. [Google Scholar] [CrossRef]
  96. Yamaguchi, Y.; Higashi, M.; Kobayashi, H. Effects of ginsenosides on maze performance and brain choline acetyltransferase activity in scopolamine treated young rats and aged rats. Eur. J. Pharmacol. 1997, 329, 37–41. [Google Scholar] [CrossRef]
  97. Jin, S.H.; Park, J.K.; Nam, K.Y.; Park, S.N.; Jung, N.P. Korean red ginseng saponins with low ratios of protopanaxadiol and protopanaxatriol saponin improve scopolamine induced learning disability and spatial working memory in mice. J. Ethnopharmacol. 1999, 66, 123–129. [Google Scholar] [CrossRef]
  98. Benishin, C.G.; Lee, R.; Wang, L.C.; Liu, H.J. Effects of ginsenoside Rbl on centralcholinergic metabolism. Pharmacology 1991, 42, 223–229. [Google Scholar] [CrossRef]
  99. Wu, C.F.; Bi, X.L.; Yang, J.Y.; Zhan, J.Y.; Dong, Y.X.; Wang, J.H.; Wang, J.M.; Zhang, R.; Li, X. Differential effects of ginsenosides on NO and TNF–alpha production by LPS-activated N9 microglia. Int. J. Immunopharmacol. 2007, 7, 313–320. [Google Scholar] [CrossRef]
  100. Chu, S.; Zhang, Z.; Zhou, X.; He, W.; Chen, C.; Luo, P.; Liu, D.D.; Ai, Q.D.; Gong, H.F.; Wang, Z.Z.; et al. Ginsenoside Rg1 protects against ischemic/reperfusion-induced neuronal injury through miR-144/Nrf2/ARE pathway. Acta Pharmacol. Sin. 2018, 40, 13–25. [Google Scholar] [CrossRef]
  101. Chen, J.; Zhang, X.; Liu, X.; Zhang, C.; Shang, W.; Xue, J.; Chen, R.; Xing, Y.; Song, D.; Xu, R. Ginsenoside Rg1 promotes cerebral angiogenesis via the PI3K/Akt/mTOR signaling pathway in ischemic mice. Eur. J. Pharmacol. 2019, 856, 172418. [Google Scholar] [CrossRef]
  102. Lee, K.W.; Jung, S.Y.; Choi, S.M.; Yang, E.J. Effects of ginsenoside Re on LPS-induced inflammatory mediators in BV2 microglial cells. BMC Complement. Altern. Med. 2012, 12, 196–203. [Google Scholar] [CrossRef]
  103. Cai, M.; Yang, E.J. Ginsenoside Re Attenuates Neuroinflammation in a Symptomatic ALS Animal Model. Am. J. Chin. Med. 2016, 44, 401–413. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, X.; Wang, C.; Wang, J.; Zhao, S.; Zhang, K.; Wang, J.; Zhang, W.; Wu, C.; Yang, J. Pseudoginsenoside-F11 (PF11) exerts anti-neuroinflammatory effects on LPS-activated microglial cells by inhibiting TLR4mediated TAK1IKKNF-κB, MAPKs and Akt signaling pathways. Neuropharmacology 2014, 79, 642–656. [Google Scholar] [CrossRef]
  105. Su, X.D.; Jang, H.J.; Wang, C.Y.; Lee, S.W.; Rho, M.C.; Kim, Y.H.; Yang, S.Y. Anti-inflammatory Potential of Saponins from Aster tataricus via NF-κBMAPK. J. Nat. Prod. 2019, 82, 1139–1148. [Google Scholar]
  106. Shal, B.; Ding, W.; Ali, H.; Kim, Y.S.; Khan, S. Anti-neuroinflammatory Potential of Natural Products in Attenuation of Alzheimer’s Disease. Front. Pharmacol. 2018, 9, 548–552. [Google Scholar] [CrossRef]
  107. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
  108. Shubhakaran, K.P.; Chin, J.H. The global burden of neurologic diseases. Neurology. 2015, 84, 758. [Google Scholar] [CrossRef] [PubMed]
  109. Niranjan, R. Molecular Basis of Etiological Implications in Alzheimer’s Disease: Focus on Neuroinflammation. Mol. Neurobiol. 2013, 48, 412–428. [Google Scholar] [CrossRef]
  110. Niranjan, R.; Nagarajan, R.; Hanif, K.; Nath, C.; Shukla, R. LPS induces mediators of neuroinflammation, cell proliferation, and GFAP expression in human astrocytoma cells U373MG: The anti-inflammatory and anti-proliferative effect of guggulipid. Neurol. Sci. 2014, 35, 409–414. [Google Scholar] [CrossRef]
  111. Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: A general review. Int. J. Neurosci. 2016, 127, 624–633. [Google Scholar] [CrossRef]
  112. Woodroofe, M.N. Cytokine production in the central nervous system. Neurology 1995, 45, S6–S10. [Google Scholar] [CrossRef] [PubMed]
  113. Kim, Y.K.; Na, K.S.; Myint, A.M.; Leonard, B.E. The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry. 2016, 64, 277–284. [Google Scholar] [CrossRef] [PubMed]
  114. Pitossi, F.; del Rey, A.; Kabiersch, A.; Besedovsky, H. Induction of cytokine transcripts in the central nervous system and pituitary following peripheral administration of endotoxin to mice. J. Neurosci. Res. 1997, 48, 287–298. [Google Scholar] [CrossRef]
  115. Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59. [Google Scholar] [CrossRef]
  116. Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2014, 79, 1–12. [Google Scholar] [CrossRef]
  117. Koper, O.M.; Kamińska, J.; Sawicki, K.; Kemona, H. CXCL9, CXCL10, CXCL11, and their receptor (CXCR3) in neuroinflammation and neurodegeneration. Adv. Clin. Exp. Med. 2018, 27, 849–856. [Google Scholar] [CrossRef]
  118. Miguel-Álvarez, M.; Santos-Lozano, A.; Sanchis-Gomar, F.; Fiuza-Luces, C.; Pareja-Galeano, H.; Garatachea, N.; Lucia, A. Non-Steroidal Anti-Inflammatory Drugs as a Treatment for Alzheimer’s Disease: A Systematic Review and Meta-Analysis of Treatment Effect. Drugs Aging 2015, 32, 139–147. [Google Scholar] [CrossRef] [PubMed]
  119. Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef]
  120. Kadry, H.; Noorani, B.; Cucullo, L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  121. Cai, Z.; Qiao, P.F.; Wan, C.Q.; Cai, M.; Zhou, N.K.; Li, Q. Role of Blood-Brain Barrier in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 63, 1223–1234. [Google Scholar] [CrossRef]
  122. Zhang, L.; Cao, Y.; Zhang, X.; Gu, X.; Mao, Y.; Peng, B. The origin and repopulation of microglia. Dev. Neurobiol. 2022, 82, 112–124. [Google Scholar] [CrossRef] [PubMed]
  123. Prinz, M.; Masuda, T.; Wheeler, M.A.; Quintana, F.J. Microglia and Central Nervous System-Associated Macrophages-From Origin to Disease Modulation. Annu. Rev. Immunol. 2021, 39, 251–277. [Google Scholar] [CrossRef] [PubMed]
  124. Woodburn, S.C.; Bollinger, J.L.; Wohleb, E.S. The semantics of microglia activation: Neuroinflammation, homeostasis, and stress. J. Neuroinflamm. 2021, 18, 258. [Google Scholar] [CrossRef]
  125. Matsumoto, J.; Dohgu, S.; Takata, F.; Machida, T.; Bölükbaşi-Hatip, F.F.; Hatip-Al-Khatib, I.; Yamauchi, A.; Kataoka, Y. TNF-α-sensitive brain pericytes activate microglia by releasing IL-6 through cooperation between IκB-NFκB and JAK-STAT3 pathways. Brain Res. 2018, 1692, 34–44. [Google Scholar] [CrossRef] [PubMed]
  126. Joshi, Y.; Giannopoulos, P.; Chu, J.; Praticò, D. Modulation of lipopolysaccharide-induced memory insult γ-secretase, and neuroinflammation in triple transgenic mice by 5-lipoxygenase. Neurobiol. Aging 2014, 35, 1024–1031. [Google Scholar] [CrossRef]
  127. Verkhratsky, A.; Nedergaard, M. Physiology of astroglia. Physiol. Rev. 2018, 98, 239–389. [Google Scholar] [CrossRef]
  128. Taipa, R.; Ferreira, V.; Brochado, P.; Robinson, A.; Reis, I.; Marques, F.; Mann, D.M.; Melo-Pires, M.; Sousa, N. Inflammatory pathology markers (activated microglia and reactive astrocytes) in early and late onset Alzheimer disease: A post-mortem study. Neuropathol. Appl. Neurobiol. 2018, 44, 298–313. [Google Scholar] [CrossRef]
  129. Villarejo-Galende, A.; González-Sánchez, M.; Blanco-Palmero, V.A.; Llamas-Velasco, S.; Benito-León, J. Non-steroidal Anti-inflammatory Drugs as Candidates for the Prevention or Treatment of Alzheimer’s Disease: Do they Still Have a Role? Curr. Alzheimer Res. 2020, 17, 1013–1022. [Google Scholar] [CrossRef]
  130. Granger, K.T.; Barnett, J.H. Postoperative cognitive dysfunction: An acute approach for the development of novel treatments for neuroinflammation. Drug Discov. 2021, 26, 1111–1114. [Google Scholar] [CrossRef]
  131. Ozben, T.; Ozben, S. Neuro-inflammation and anti-inflammatory treatment options for Alzheimer’s disease. Clin. Biochem. 2019, 72, 87–89. [Google Scholar] [CrossRef]
Table 1. Traditional uses, common names and distribution of the Agave species mentioned in the text.
Table 1. Traditional uses, common names and distribution of the Agave species mentioned in the text.
Scientific
Name
Common
Name (s)
UsesDistribution in MexicoReferences
A. offoyana Jacobi., 1864No data foundNo data foundNo data found[1]
A. angustifolia Haw“Maguey espadín”
“Maguey de monte”
“Bacanora”
Caribbean Agave
Urticaria
Dysentery
pain of wounds
relieve rheumatic pain
inflammation.
Mezcal production
Oaxaca
Durango
[9]
A. rzedowskiana P. Carrillo, Vega & R. Delgad.No data foundNo data foundNayarit/Sinaloa/Jalisco[21]
A. ornithobroma
Gentry, 1982
“Maguey Pajarito”,
“Amole”, “Lechuguilla”.
Fiber for the manufacture of backpacks and matsNayarit/Sinaloa[21]
A. salmiana
Otto ex Salm-Dyck
“Maguey pinto”,
En Michoacán:
“Akamha”
(purépecha *)
“Maguey cimarrón”
“Maguey pulquero”
“Pulque Agave
Inflammation, gastritis, diabetes, wounds, blows and coughTlaxcala, Hidalgo, Querétaro, San
Luis Potosí y
Zacatecas.
[22,23]
A. barbadensis TrelNo data foundNo data foundNo data found[21]
A. impressa Gentry“Maguey masparillo”
“Lechugilla”
OrnamentalSinaloa
Durango
Nayarit
[22]
A. marmorata RoezlMaguey de caballo
Pitzomel
Cough
Asthma
internal bumps
Ornamental.
Puebla
Oaxaca
[22]
A. intermixta TrelNo data foundIt is used as a decoction of the plant to treat cancer problemsNo data found[22]
A. schidigera
Lem.
“Lechugilla mansa”
Thread-leaf Agave
To relieve gastric problems in childrenSonora y Chihuahua a
Michoacán, Jalisco, Aguascalientes, San Luis Potosí
[22,24]
A. americana L., 1753“Maguey pinto”, “Mezcal”, American AgaveGastric ulcers, eye inflammation, diuretic, wounds, diarrhea. OrnamentalTropical America, widely distributed in Mexican territory[24]
A. tequilana F.A.C. Weber“Maguey azul”
Blue Agave
Tequila productionJalisco, Michoacán, Guanajuato y Nayarit. Tamaulipas[25]
A. fourcroydes LEM.“Henequen”It is a natural fiber used to make hammocks and mats.
It is fermented to produce alcohol (similar to mezcal)
Yucatán[26]
A. utahensis
Engelm
Utah AgaveNursery Stock ProductNo data found[27]
A. schotti Engelm“Maguey”
Arizona shin-dagger
“Amol” (Náhuatl) **
No data foundChihuahua, Sonora, and Baja California.[28]
* Purépecha: a national indigenous language of the Tarascan linguistic family, which is spoken in Michoacan, Mexico; ** Náhuatl: an indigenous language of the Uto-Aztecan linguistic family, which is spoken in Mexico and Central America.
Table 2. Chemical structures of compounds with anti-inflammatory activity contained in Agave species.
Table 2. Chemical structures of compounds with anti-inflammatory activity contained in Agave species.
Natural Compound NameNumberChemical Structure
smilagenin-3-O-[β-D-glucopyranosyl (1→2)-β-D-galactopyranoside]1Plants 11 02208 i001
3-O-[(6’-O-palmitoyl)-β-D-glucopyranosyl sitosterol 2Plants 11 02208 i002
Agavesaponin E3Plants 11 02208 i003
Agavasaponin H4Plants 11 02208 i004
Hecogenin5Plants 11 02208 i005
Tigogenin6Plants 11 02208 i006
β-sitosterol7Plants 11 02208 i007
Isorhamnetin8Plants 11 02208 i008
Hesperidin9Plants 11 02208 i009
Delphinidin10Plants 11 02208 i010
Quercetin11Plants 11 02208 i011
Apigenin12Plants 11 02208 i012
Catechin13Plants 11 02208 i013
Ginsenoside Rg114Plants 11 02208 i014
Ginsenoside Re15Plants 11 02208 i015
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Herrera-Ruiz, M.; Jiménez-Ferrer, E.; González-Cortazar, M.; Zamilpa, A.; Cardoso-Taketa, A.; Arenas-Ocampo, M.L.; Jiménez-Aparicio, A.R.; Monterrosas-Brisson, N. Potential Use of Agave Genus in Neuroinflammation Management. Plants 2022, 11, 2208. https://doi.org/10.3390/plants11172208

AMA Style

Herrera-Ruiz M, Jiménez-Ferrer E, González-Cortazar M, Zamilpa A, Cardoso-Taketa A, Arenas-Ocampo ML, Jiménez-Aparicio AR, Monterrosas-Brisson N. Potential Use of Agave Genus in Neuroinflammation Management. Plants. 2022; 11(17):2208. https://doi.org/10.3390/plants11172208

Chicago/Turabian Style

Herrera-Ruiz, Maribel, Enrique Jiménez-Ferrer, Manasés González-Cortazar, Alejandro Zamilpa, Alexandre Cardoso-Taketa, Martha Lucía Arenas-Ocampo, Antonio Ruperto Jiménez-Aparicio, and Nayeli Monterrosas-Brisson. 2022. "Potential Use of Agave Genus in Neuroinflammation Management" Plants 11, no. 17: 2208. https://doi.org/10.3390/plants11172208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop