Next Article in Journal
Microbial Diversity and Bioactive Compounds in Dried Lycium barbarum Fruits (Goji): A Comparative Study
Previous Article in Journal
Antihyperglycemic Properties of Extracts and Isolated Compounds from Australian Acacia saligna on 3T3-L1 Adipocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances on the Amaryllidacea Alkaloids Collected in South Africa, Andean South America and the Mediterranean Basin

by
Antonio Evidente
1,2
1
Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy
2
Institute of Sciences of Food Production, National Research Council, Via Amendola 122/O, 70185 Bari, Italy
Molecules 2023, 28(10), 4055; https://doi.org/10.3390/molecules28104055
Submission received: 3 April 2023 / Revised: 4 May 2023 / Accepted: 8 May 2023 / Published: 12 May 2023
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
The alkaloids are one of the most represented family of natural occurring biological active compounds. Amaryllidaceae are also very well known for their beautiful flower and are thus used as ornamental plants in historic and public gardens. The Amaryllidacea alkaloids constitute an important group that is subdivided into different subfamilies with different carbon skeletons. They are well known from ancient times for their long application in folk medicine, and in particular, Narcissus poeticus L. was known to Hippocrates of Cos (ca. B.C. 460–370), who treated uterine tumors with a formulate prepared from narcissus oil. To date, more than 600 alkaloids of 15 chemical groups exhibiting various biological activities have been isolated from the Amaryllidaceae plants. This plant genus is diffused in regions of Southern Africa, Andean South America and the Mediterranean basin. Thus, this review describes the chemical and biological activity of the alkaloids collected in these regions in the last two decades as weel those of isocarbostyls isolated from Amaryllidaceae in the same regions and same period.

1. Introduction

Amaryllidaceae plants grow as wild species in several countries and are also cultivated for their beautiful flowers and for the production of volatile oils (Figure 1). They are classified into 60 genera [1] that are diffused in different world regions but that are dominant within three distinct geographical locations, i.e., Andean South America, Southern Africa and the Mediterranean basin [2,3]. About one third of one thousand Amaryllidaceae species grow in South Africa and are commonly used in folk medicine [2]. They include well-known ornamental varieties such as daffodils (Narcissus), snowdrops (Galanthus) and snowflakes (Leucojum) [3]. Consequently, they have a high commercial value and are important for the floriculture industry [4,5]. The use of Narcissus in the Mediterranean basin begins at the time of Hippocrates and Pliny [6], while in South America in the archeological Inca ruins, there were found floral depictions of Ismene, Pyolirion and Stenomesson [7]. Paintings of Brunsvigia species were found in Lesotho [8].
Studies on Amaryllidaceae alkaloids (AA) began in 1877 with lycorine (1, Figure 2), which is the main Amaryllidaceae alkaloid that was isolated from Narcissus pseudonarcissus [9]. From that time, investigations of this group of alkaloids increased considering their wide range of biological activities. These include antitumor, antiviral, antibacterial, antifungal, antimalarial, analgesic, and cytotoxic activities [10]. The most important application in medicine of Amaryllidaceae alkaloids is represented by the use of galanthamine (2, Figure 2) to treat Alzheimer’s disease and is already commercialized as a drug. Galanthamine (2) is able to selectively inhibit the enzyme acetylcholinesterase (AChE), which plays a fundamental role in the disease [1,11]. Amaryllidaceae plants also synthesize poisons such as lycorine and galanthamine, and this toxicity should be always considered [12].
Hundreds of scientific articles have reported on the biosynthesis, source, isolation and biological activities of AA, which are grouped into more than 12 subfamilies considering their carbon skeleton, including norbelladine, rystilline, α-crinanes, β-crinanes, lycorane, galanthamine, pretazettine, homolycorine, montanine, cherylline, crinasiadine, clivimine, ismine analogues and miscellaneous alkaloids [1,6].
Considering the number of scientific articles describing the different aspects of AA, several articles were published on this topic regarding the phytochemical, biological and pharmacological properties of Crinum bulbispermum [13] Nerine [14], Haemantheae [15] and Crinum, Ammocharis, Amaryllis and Cyrtanthus [16]. The relation between the biological activity of AA and their absolute configuration was also extensively described [17] as well as the acetylcholinesterase inhibition of extracts of Amaryllidaceae collected in South Africa with potential for Alzheimer’s disease treatment [18]. Recently, only AA isolated in the decade 2009–2020 were reported [19].
This review reports on the plant sources, isolation, and chemical and biological characterization of alkaloids produced by Amaryllideceae that are native to Andean South America, South Africa and the Mediterranean basin, that were collected in the last two decades (2009–2023), and that are of the findings that resulted from SCI-FINDER. The results are chronologically reported in three different sections, namely Section 2, Section 3 and Section 4. Similarly, Section 5 reports the chemical and biological properties of isocarbostyls isolated from Amaryllidaceae in the same regions and same period.

2. Alkaloids Isolated from Amaryllidacae Plants Collected in Andean South America

In Argentina, about 61 species of the Amaryllidaceae family grow, covering a wide variety of genera (Chlidanthus, Crinum, Habranthus, Haylockia, Hieronymiella, Hippeastrum, Phycella, Rhodophiala, Stenomesson and Zephyranthes) [20]. In particular, almost all the wild Habranthus jamesonii, Phycella herbertiana, Rhodophiala mendocina and Zephyranthes filifolia species collected in the Argentinian Andean region contain lycorine and galanthamine (1 and 2). The organic extracts of all four species showed strong AChE inhibitory activity with IC50 values between 1.2 and 2 µg/mL. The main alkaloids found in these species were lycorine, tazettine, vittatine, haemanthamine and lycoramine (1, and 36, Figure 2), which are not produced by Z. filifolia, while galanthine and 11-hydroxyvittatine (7 and 8, Figure 2) are only produced by H. jamesonii and P. herbertiana [21]. These alkaloids show other interesting biological properties besides their AChE inhibitory activity. Haemanthamine (5) strongly induces apoptosis [22] and exhibits antimalarial activity [23]. Vittatine (4) shows cytotoxicity and antibacterial activity [23], as does 11-hydroxyvittatine (8) [6]. Lycorine (1) exhibits cytotoxic, apoptotic, antiviral, antifungal, antiprotozoan [6,22], and anti-inflammatory activities [24]. Furthermore, alkaloid 2 could be a promising therapeutic agent against leukemia [25]. Galanthine (7) possesses analgesic and hypotensive effects [23], while tazettine (3) shows moderate cytotoxic activity [26].
Lycorine, galanthamine, haemantamine (1, 2, and 5), maritidine, homolycorine and hamayne (911, Figure 2) were isolated from the ethanolic extract of Caliphruria subedentata, which showed moderate cytotoxic activity [27,28]. This Amaryllidaceae as well as Caliphruria hartwegiana and Caliphruria tenera are considered endemic in Colombia [29]. They belong to the genus Caliphruria that is not much diffused in the world, as only four species were found in some tropical regions of South America [29], but particularly, they are localized in restricted parts of the Central and Occidental Cordilleras in the departments of Valle del Cauca, Cundinamarca and Huila [30]. The successive investigation allowed for the identification of tazzetine (3, Figure 2), ismine, trisphaeridine, narwedine, kirkine, galanthindole, 6α-deoxytazettine, 6-methoxypretazettine, and 3-epi-macronine, and anydrolycorine and dehyroanhydrolycorine, (1216 and 18, 19 and 21, Figure 2, and 17 and 20, Figure 3) in the organic extract of C. subedentata using the GC-MS technique [31].
Hippapiline, papiline, and 3-O-demethyl-3-O-(3-hydroxybutanoyl)- haemanthamine (22 and 23, Figure 3, and 24, Figure 2) were isolated together with haemanthamine (5), galanthamine (2), narwedine (14), 11β-hydroxygalanthamine (25, Figure 2) apogalanthamine, and 9-O-demethyllycosinine B (26 and 27, Figure 3) from an indigenous Brazilian species of Hippeastrum papilio. This Amaryllidaceae was collected during the flowering period in the south of Brazil (Caxias do Sul—RS) [32].
Galanthamine (2) was isolated from the bulb extract of Rhodolirium andicola, which is a native Chilean Amaryllidaceae species that was collected during the flowering season in December 2016 from National Park Conguillio, Araucanía Region, Chile [33]. This organic extract, which contained 2.3 ± 0.18 g/mL of alkaloid 2, showed inhibitory of AchE with IC50 values between 11.25 ± 0.04 and 57.78 ± 1.92 g/mL. Twelve other alkaloids: lycoramine (6), galanthaminone (28, Figure 2) 6α-deoxytazettine (18), norpluviine diacetate, 3-O-acetyl-1,2-dihydrogalanthamine (29 and 30, Figure 2), heamanthamine (5), undulatine diol (31, Figure 3), tazettine (3), acetylnatalensine (=acetylhaemanthamine) (32, Figure 2), undulatine (33, Figure 3) 3-epi-macronine (21) and crinan-3-one (34, Figure 2), were isolated from the same organic extract [33]. All these alkaloids were tested for AchE inhibition. The results of the bioassay showed that lycoramine, norpluvine diacetate and 6α-deoxytazettine contributed to potential acetylcholinesterase inhibition of the plant extract [33].
Lycorine (1), which is the main alkaloid, as well as dehydroanhydrolycorine (20) and 1-O-acetyllycorine (35, Figure 2) were identified in the leaves and bulbs of Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata, which are all Amaryllidaceae species collected in different regions of Merida State-Venezuela [34]. In addition, buphanisine (36, Figure 3) in the C. amabile and C. moorei bulb extracts and undulatine (33) in the bulb extract of A. belladonna were found [34]. The extract of C. amabile leaves showed the strongest inhibition of AChE and BuChE followed by C. erubescens leaves [34].
Lycorine, galanthamine, tazettine (13), and chlidanthine (37, Figure 3) were identified in the leaf extract of Pyrolirion albicans, which grows in the coastal region of southern Peru [35]. From the bulb extract of the same plant, lycorine (1), galanthamine (2), vittatine (4), N-demethylgalanthamine (38, Figure 2), crinine, montanine and pancracine (3941, Figure 3) and sternbergine and hippeastrine (42 and 43, Figure 2) were isolated and identified [35]. Montanine (40), which is the predominant alkaloid in the bulb extract, and its derivative pancracine (41) showed anti-inflammatory and immunomodulatory [36], antioxidant and antimicrobial [37] properties. They also showed significant anxiolytic, antidepressant, anticonvulsant [38] and antirheumatic activities [39].
A. belladonna has a geographical distribution covering mainly southern Africa [40], where it has significant usage in the traditional medicine of the native people but which was also isolated in Brazil [40]. The chemical analysis of the bulb organic extract of the samples collected in Brazil helped to identify twenty-six different AA, and three of them, namely, 1-O-acetylcaranine (44, Figure 2), buphanamine, and 3-O-acetylhamayne (45 and 46, Figure 3) were isolated [40]. The AA and the crude bulb organic extracts were tested against four different parasitic protozoa (Trypanosoma cruzi, Trypanosoma brucei rhodesiense, Leishmania donovani, and Plasmodium falciparum [40]). The crude organic extract and 3-O-acetylhamayne exhibited good antiprotozoal activity in vitro, although both had a high cytotoxic index [40].
Lycorine (1), 7-demethoxy-9-O-methylhostasine (47, Figure 3), and rutin were identified in the Hippeastrum stapfianum leaf extract [41]. This Amaryllidacea is an endemic species from the Brazilian savannah with biological and pharmacological activities, including AChE inhibition showing IC50 values ranging between 386.00 ± 0.97 and 114.80 ± 0.95 µg/mL [42]. Their potential for the treatment of Alzheimer’s disease is based on the ability of the plant extract to activate PPAR-α (peroxisome proliferator-activated receptor alpha) and PPAR-γ (peroxisome proliferator-activated receptor gamma) selectively [41].
Lycorine, galanthamine, lycoramine, galanthine, anhydrolycorine, didehydroanhydrolycorine and hippeastrine, (1, 2, 6, 7, 17, 20 and 43,) norlycoramine, assoanine, and 9-O-demethylholycorine (48 and 50, Figure 2) and pancratinine C (51, Figure 3) were isolated from the bulb organic extract of Ismene amancaes, an endemic Peruvian Amaryllidaceae, which was collected during the flowering period in the surroundings of the town Pagash Alto, Salpo District, Otuzco Province, La Libertad Region (Peru) [43]. The crude plant extract also showed low inhibition of the enzymes AChE and BuChE, with IC50 values of 14.6 ± 0.6 and 37.6 ± 1.4 µg/mL, respectively, and good to moderate inhibitory activity against Plasmodium falciparum strain FCR-3 (chloroquine-resistant), the protozoan responsible of malaria disease, with IC50 value of 3.78 ± 0.3 µg/mL [43].

3. Alkaloids Isolated from Amaryllidacae Plants Collected in South Africa

Lycorine, galanthamine, tazettine haemanthamine, homolycorine, ismine, trisphaeridine, undulatine, buphanisine, and crinine montanine (13, 5, 10, 12, 13, 33, 36, 39 and 40) and buphanidrine, ambelline, norbelladine, augustine, disticahamine, distichaminol, crinamine, haemanthidine, and buphanamine (5260, Figure 4) were isolated together with pancratistatine, an isocarbostyryl (see below Section 5), from Boophone haemanthoides [44]. The bulbs of this Amaryllidacea were collected during the flowering season in the Nieuwoudtville area of the Northern Cape Province of South Africa [44]. B. haemanthoides belong to the African genus Boophone Herb, which also includes Boophone disticha. B. disticha is widely distributed in Africa, ranging from Sudan to the Western Cape Province, while B. haemanthoides is a rare and endangered species found in a more limited range in the winter rainy region of South Africa, which is confined to a relatively small area in the southwest, the Western Cape area, where gentle rain falls from May to August, but the summers are dry, and in parts of southern Namibia [45]. Both Amaryllidaceae are widely used in folk medicine [46,47]. Among the alkaloids identified, lycorine (1) and distichamine (56) showed cytotoxic activity demonstrated in acute lymphoblastic leukemia (CEM), breast adenocarcinoma (MCF7) and cervical adenocarcinoma (HeLa) cells with IC50s in the range from 1.8 to 9.2 μM [44]. Previously from B. heamanthoides, collected in Saldhana Bay area in South Africa, also buphanisine crinine and buaphanidrine (36, 38 and 52) were isolated together with distichamine (56) [48].
Crinsarnine and sarniensinol (61 and 62, Figure 4), belonging, respectively, to the crinine and mesembrine-types subgroups, were isolated together with bowdensine, sarniensine (63 and 64, Figure 4), and 1-O-acetyl-lycorine (35), from the dried bulbs of Nerine sarniensis [49], a species restricted to the Western Cape of the South Africa region [2]. All the alkaloids were assayed together with tazettine and 3-epi-macronine (3 and 21) against the Orlando reference strain of Aedes aegypti, which is the primary vector of dengue and yellow fever and Zika viruses [49]. The latter causes microcephaly and other serious brain anomalies during pregnancy, and it could easily become a potential threat to international public health safety [50]. Mosquito control is one of the main methods used to reduce the spread of these viruses. None of the compounds tested showed mortality against the first instar Ae. aegypti larvae at the concentrations tested. In adult topical bioassays, only crinsarnine (61) exhibited adulticidal activity with an LD50 value of 2.29 ± 0.049 µg/mosquito [49]. Regarding the structure–activity relationship, the pretazettine and crinine scaffold in alkaloids 62 and 64 and in 61 and 63, respectively, proved to be important for their activity, while the pyrrole[de]phenanthridine scaffold present in alkaloid 35 appeared to not be important for toxicity [49]. Among the pretazettine group compounds, the opening of the B ring or the presence of a B ring lactone as well as the trans-stereochemistry of the A/B ring junction are important features for activity, while in crinine-type alkaloids, the substituent at C-2 seems to play a role in their toxicity [49].
Buphanidrine (52) is the main alkaloids isolated from the organic extract of Boophone disticha [51,52,53,54,55,56,57,58,59]. B. disticha is one of the most popular bulbous plants widely used in traditional medicine in South Africa [52]. Other known alkaloids isolated from the same organic extract were buphanisine, buphanamine, (35 and 45) 3-O-methylcrinamidine, and acetyl-3-nerbowdine (65 and 66, Figure 4) [60]. Later, 1-O-acetylbuphanamine (67, Figure 4) was also isolated from the same organic extract [61]
Haemanthamine and haemanthidine (5 and 59), together with metolachlor, which is an unusual chlorinated amide, were isolated from Scadoxus puniceus [62], an Amaryllidaceae species used in folk medicine as an herbal tonic prescribed to treat several ailments [62]. It is used as a detoxifying and energizing agent as well as to clear skin conditions, treat kidney and urinary infections, cure tonsillitis, and treat pneumonia in South Africa. Haemanthamine, haemanthidine, and metolachlor showed strong acetylcholinesterase inhibition with IC50 values of 23.1, 23.7, and 11.5 μM, respectively [62].
The alkaloids produced from Crinum buphanoides, Crinum graminicola, Cyrtanthus mackenii, and Brunsvigia grandiflora, which are all indigenous to South Africa, were isolated and identified [63]. Lycorine (1) appeared to be the main alkaloid produced by all four species, but C. graminicola was the highest alkaloid-producer plant (2 g/kg) [63]. Furthermore, C. buphanoides produced tazettine, 1-O-acetyllycorine (3 and 35), and 2-O-acetyllycorine (68, Figure 4), while haemanthamine, haemanthidine (5 and 59), and criwelline (69, Figure 4) were isolated from C. graminicola, and tazettine and 11-hydroxyvittatine (3 and 8) were produced by C. mackenii [63]. The latter alkaloid (8) and crinamine (58) were produced by B. grandiflora [63].
Channaine (70, Figure 4), which is an AA with an unusual cage-like ring structure at the interface of two aryl-hydroindole subunits, was isolated from Sceletium tortuosum [64]. This species was collected from St. Helena in the Western Cape Province of South Africa and belongs to the Sceletium genus, which is endemic to South Africa and which is a well-known producer of alkaloids [65,66]. Alkaloid 70 was previously isolated from the same Amaryllidaceae [67], but only its empirical formula and some functional groups were assigned. Later, Popelak and Lettenbauer [68] described that channaine (66) contained two veratrole rings, being a dimer of two subunits and racemic of both channaine compounds.
Albomaculine (71, Figure 4), coccinine and incartine (72 and 73, Figure 5), and montanine (40) were isolated from Haemanthus humilis, which is indigenous to South Africa [69]. H. humilis does not synthesize lycorine, the main and most common alkaloid produced by this plant genus. Coccinine (72) appeared to be the main metabolite (1.49 g/kg). All alkaloids tested for their anticancer activity against a panel of six human cancer cell lines (the human breast MCF7 (HTB-22™), Hs578T (HTB-126™), and MDA-MB-231 (ATCC®HTB-26™), colon HCT-15 (CCL-225™), and lung A549 (CCL-185™) cancer cell lines as well as SK-MEL-28 (HTB-72™) melanoma cells, and coccinine and montanine (72 and 40) showed significant activity at low micromolar concentrations [69].
The ‘lily borer’ moth Brithys crini is a very dangerous parasite of Amaryllidaceae plants, having a great effect during the larval stage. The organic extract of Crinum moorei, collected in the botanical garden of the University of KwaZulu-Natal, was analyzed, and alkaloids belonging to different subgroups of AA were identified as ambelline (53) and cherylline (74, Figure 5). The presence of ambelline represents a surprise, as it was not previously isolated in the organic extract obtained from this Amaryllidaceae [70].
In the organic extract of Ammocharis coranica were identified lycorine, 1-O-acetylcaranine, crinamine (1, 44, 58) and caranine (75, Figure 4) [71]. Later, hamayne and 1-O-acetyllycorine (11 and 35) and 1-O-acetyl-9-O-demethylpluviine (76, Figure 5) were also isolated [72], as well as buphanisine, buphanidrine and ambelline (36, 52 and 53), and epi-buphanisine and 6α-hydroxycrinamine (77 and 78, Figure 4) [73]. A. coranica (Ker Gawl.) Herb. is the second most widely distributed Amaryllidaceae of the Ammocharis Herb. genus and is found within all southern African countries, as in Little Karoo of South Africa, northward to Zimbabwe, and the southern Angola regions [74]. More recently, 6α-hydroxybuphanidrine and golceptine (79, Figure 4 and 80, Figure 5) were isolated together with an unusual crinine type alkaloid such as charisine (81, Figure 4), from A. coronica [75].
Nineteen AA belonging to different subgroups were isolated from Clivia miniata, which is an herbaceous evergreen plant endemic to South Africa and Swaziland [76]. The alkaloids were identified as lycorine, galanthamine, tazettine, vittatine, haemanthamine 11-hydroxyvittatine, sternbergine, hippeastrine, 1-O-acetylcaranine, haemanthidine, caranine, (15, 8, 4244, 59 and 75), clivimine, cliniatine C, clivonine, nobilisitine B, 4′,O-demethylbelladine, 3-O-acetyl-8-O-demethylmaritidine, 8-O-demehylmaritidine, and clivimine B (8285, Figure 5, 86, Figure 4, and 89, Figure 4) [76]. The main alkaloids were lycorine, haemanthamine, and clivimine (1, 5 and 82). C. miniata is the most used Amaryllidacea species in folk medicine in South Africa [76]. All the isolated alkaloids were tested for their AChE/BuChE inhibition, using galanthamine and eserine as reference compounds. Alkaloids belong to the homolycorine structure type as clivimine, cliniatine C, clivonine, nobilisitine B and clivimine B, which show low AChE/BUChE inhibitory activity. Among the lycorine-type alkaloids such as lycorine, sternbergine, 1-O-acetylcaranine, and caranine was noted that the activity could be due to the presence of free hydroxyl groups in positions C1 and C2, which are not present in those of the homolycorine type. This diol system is probably a functional group that improves binding in the active site of AChE/BuChE. However, lycorine and several of its analogues did not show significant activity against AChE and BuChE [76].

4. Alkaloids Isolated from Amaryllidacae Plants Collected in Mediterranean Basin

Ungeremine and zefbetaine (90 and 91, Figure 6), which are two 2-oxyphenanthridinium alkaloids, were isolated from the bulb organic extract of Egyptian Pancratium maritimum collected from sandy hills on the northern coast during the flowering and fruit-producing stages [77]. From the same organic extract, lycorine, galanthamine, tazettine, haemanthamine, 11-hydroxyvittatine, homolycorine, trispheridine, pancracine, 9-O-demethylhomolycorine and haemanthidine, (13, 5, 8, 10, 13, 41, 50 and 59), and lycorenine (92, Figure 6) were isolated and identified [78,79]. Ungeremine showed toxicity against Flavobacterium columnare and Edwardsiella ictaluri, respectively, with IC50 and MIC values of 58 ± 0 and 3 ± 0 and (0.8 ± 0 and 0.9 ± 0.2 mg/L [80]. This activity was compared to that of lycorine (1) and pseudolycorine (93, Figure 6), isolated from Narcisuss tazetta subsp. tazetta [81], ungeremine isomer, zefbetaine (90), and anhdrolycorine (17). The aromatization of the C ring and the oxidation to an azomethine group of C-7 of the B ring are structural features important for antibacterial activity [82]. Furthermore, the presence of the 1,3-dioxole ring joined to the A ring and oxygen location of the C ring of the pyrrolo[de]phenanthridine skeleton also play significant roles on the antibacterial activity [82]. Ungeremine (90) showed to be a promising biofungicide against Penicillium roqueforti and Aspergillus niger with MIC90 values of 0.003 and 0.2 mg/mL [83]. These two fungi are very dangerous food contaminants and can cause bakery product deterioration. They show significant potential to be included as an appropriate biofilm that can be used in intelligent food packaging [83]. Alkaloid 90 was incorporated in chitosan-based microbeads, which were prepared by external gelation by using sodium tripolyphosphate (TPP) as a crosslinking agent. All the microbeads evidenced antimicrobial activity against P. roqueforti [84]. These microbeads were included in a thermoplastic starch-based polymer Mater-Bi (MBi), and MBi/CTUn and bioactive biocomposites were obtained. The films showed bioactivity against P. roqueforti [85].
1-O-(3’-acetoxybutanoyl)lycorine and narseronine (94 and 95, Figure 6) were isolated from Narcissus serotinus, an autumn flowering Amaryllidaceae collected in Ben Slimane, near Casablanca (Morocco) [86]. Successively from the same Amaryllidaceae but collected near Vinarós, Castellón Province (Spain), 3-O-methylnarcissidine and 1-O-acetyl-3-O-methylnarcissidine, 1-O-Acetyl-3-O-methyl-6-oxonarcissidine, 11-hydroxygalanthine and 2-O-methylclivonine (96100, Figure 6), and the isocarbostyryl 2-methoxypratosine (see below Section 5) were isolated together, with incartine (73) masonine (101, Figure 6), galanthine and hippeastrine (7 and 42) [87].
2-Oxomesembrenone, 7,7a-dehydromesembrenone and 2-oxo-epi-mesembrano (102104, Figure 6) together with mesembrenone (105, Figure 6), which is the main alkaloid, 4-O-demethylmesembrenone and mesembrine (106 and 107, Figure 6), and 6-epi-mesembrenol and 6-epi-mesembranol (108 and 109, Figure 7) were isolated during the flowering period from Narcissus triandus, a wild daffodil of the Ganymedes section from the Iberian Peninsula [88]. The isolated alkaloid profile confirms the presence of mesembrane alkaloids in all the Narcissus taxa, as well as the absence of trace of typical alkaloids. Furthermore, the founding of mesembrane alkaloids in Narcissus pallidulus and some hybrids [89] showed an important phylogenetic aspect of Narcissus: the biosynthetic pathway of alkaloids by species belonging to the Ganymedes section is atypical among other Amaryllidaceae. In addition, mesembrane alkaloids produced by this group of Mediterranean daffodils share chemical features with the very distantly related dicotyledonous plants of the genus Sceletium from South Africa, whose potential therapeutic applications have already been recognized [88].
Lapiedra martinezii is an Amaryllidaceae species diffused in the Mediterranean basin, including essentially the Iberian Peninsula and the north of Africa [90], together with Narcissus, Hannonia, and Vagaria gebìnera [90,91]. Lapiedra is the oldest genus and is only diffused in the Mediterranean coastal side of the Baetic mountain chain (from Malaga to Valencia), with a few populations in the nearby Spanish city of Melilla (North Africa). The ability of L. martinezii to produce alkaloids is strictly related to its distribution area. Several alkaloids belonging to different subgroups were identified: as lycorine, tazettine, 11-hydroxyvittatine, homolycorine, ismine, trisphaeridine, kirkine, anhydrolycorine, didehydroanhydrolycorine, sternbergine, hippeastrine, assoanine, demethylhomolycorine, and lycorenine (1, 3, 8, 10, 12, 13, 15, 17, 20, 42, 43, 48, 50 and 92), as well as tyramine, 5,6-dihydrobicolorine, deoxylycorenine 6-O-methyllycorenine, 1-O-acetylnorpluviine 6-deoxypretazettine, lycosenine, norpluviine, ungiminorine acetate, ungiminorine, narcissidine, and narcissidine acetate (110121, Figure 7) [92].
11α-Hydroxy-O-methylleucotamine (122, Figure 8) was isolated together with 2-hydroxyhomolycorine, sanguinine, habranthine, leucotamine, and O-methylleucotamine (123127, Figure 8) as well as lycorine, galanthamine, and vittatine (1, 2, 4), from fresh bulb extracts of Pancratium illyricum L. [93], a species endemic to Sardinia (Italy), which was collected during the flowering period in the South of the island of Punta San Michele, Cagliari, Italy [93]. Considering that the galanthamine type inhibits AChE [11], all the isolated alkaloids of the galanthamine type (1, 123127) showed good inhibitory activity against AChE, comparable to that of the positive control galanthamine hydrobromide [93]. The strongest inhibition was exhibited from 11α-hydroxy-O-methylleucotamine (122) and galanthamine hydrobromide, with IC50 values of 3.5 ± 1.1 and 1.5 ± 0.2 μM, respectively [93]. Alkaloids 123-127 showed a marked decrease in enzyme inhibition, while sanguine (124) showed the strongest activity (ca. 10 times higher than galanthamine [93]). Alkaloid 124, compared with galanthamine, has a hydroxyl group at C-9 instead of a methoxyl group, which seems to be important for the inhibition of AChE contributing to its effective binding to the enzyme [94]. In addition, the spatial orientation of the hydrophilic group can affect this binding interaction. The β-configuration of the hydroxyl group at C-11 of 11β-hydroxygalanthamine decreases, by 10 times, the activity compared to its α-epimer habranthine (125) [95]. The low decrease in 11α-hydroxy-O-methylleucotamine (122), which showed an IC50 value of 1.61 ± 0.2, is probably due to the presence of the bulky butyryl group at C-3 [93].
Jonquailine (128, Figure 8), an alkaloid belonging to the pretazettine group, was isolated from dried bulbs of Narcissus jonquilla quail, widespread in Spain and Portugal [96]. Alkaloid 128 showed significant antiproliferative effects against glioblastoma, melanoma, uterine sarcoma and non-small-cell lung cancer cells, which exhibit various forms of drug resistance, including resistance to apoptosis and multi-drug resistance [97]. Jonquailine (128) was able to synergize, with paclitaxel, its antiproliferative action against drug-resistant lung cancer cells [97]. The hydroxylation at C-8 is an important feature for anticancer activity, but this seems to be affected by both the stereochemistry and acetalization of lactol [97].
Lycorine and 8-O-demethylmaritidine (1 and 89) were isolated together with clivatine and nobilisine (129 and 130, Figure 8) from the flower extract of Clivia nobilis, cultivated in Egypt [98]. The crude flower organic extract as well as all the alkaloids isolated were tested for their antibiotic activity against Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeroginosea bacteria. The crude flower extract showed antibacterial activity against both microorganisms, while nobilisine (130) showed very good activity against the Gram-negative P. aeroginosea [98].
Lycorine, galanthamine, tazettine, haemanthamine, galanthine, 11-hydroxyvittatine, ismine, trispheridine, narwedine, galanthindole, anhydrolycorine 3-epi-macronime, crinan-3-one, crinine, hippeastrine, assoanine, 9-O-demethylhomolycorine, incartine, 8-O-demethylmaritidine, 5,6-dihydrobicolorine, saguinine, and O-methyleucotamine (1, 2, 3, 5, 7, 8, 12, 13, 14, 16, 17, 21, 34, 38, 43, 48, 50, 73, 88, 111, 124 and 127) and anhydrogalanthamine, 11,12-dehydrolycorene, 2,11-didehydro-2-dehydroxylycorine, 6-O-methoxylpretazettine, galwesine, galasine, oxoincartine (131138, Figure 8), and hippamine (139, Figure 9) were isolated from the aerial parts of Galanthus elwesii, collected from three different regions of Turkey [99].
Hippamine (139,) had been also previously extracted as a minor alkaloid from Sternbergia lutea [100]. The organic extract of samples of G. elwessii collected in Karaburun-Izmir showed to be the most active. A molecular docking study was carried out to determine the binding of alkaloids in the gorge of the active site of acetylcholinesterase (AChE) of Electrophorus electricus, and equine butyrylcholinesterase (BuChE) and O-methylleucotamine (127) showed the most interesting results [99].
Galanthamine (2) was produced by Narcissus poeticus collected in Abruzzo (Italy). Alkaloid 2 was found in all organs of the plant (flower, stem, bulb and root), but it accumulates in the bulbs [101]. The organic extract of N. poeticus obtained from the flowers is also used as an important fragrance in perfumery, while that of the petals and coronas show allergenic properties [101].
Lycorine (1) was isolated from the bulb extract of Pancratium foetidum, collected in Saïdia, Oujda region, Morocco [102]. Lycorine showed moderate antibacterial activity and had more efficacy than streptomycin and ampicillin against P. aeruginosa. A virtual docking ligand-lycorine protein screening showed that compound 1 can interact with target amino residues studied by hydrogen and metal-ion bonds [102].
Galanthamine, lycorine pseudolycorine and 11-hydroxygalanthine (1, 2, 93 and 99) were isolated from the bulb organic extract of Narsissus tazetta subsp. tazetta, collected during the flowering period in Akçapınar/Muğla, Turkey [81]. Other alkaloids were isolated and identified as narwedine, anhydrolycorine, 9-O-demethylhomolycorine, 4′,O-methylnorbelladine, 8-O-demethylmaritidine, pseudolycorine, 1-O-acetyl-3-O-methylnarcissidine, 11-hydroxygalanthine, narcissidine, saguinine (14, 17, 50, 86, 88, 93, 97, 99, 120, 124) and 9-O-methylpseudolycorine (139, Figure 9) and pancratinine C (51) [81]. All the alkaloids isolated were assayed for their ability to inhibit AChE and BuAchE, and 11-hydroxygalanthine and narcissidine (99 and 120) showed significant activity on acetylcholinesterase (AChE) [81].
From the bulbs of Pancratium maritimum, which were always collected in Turkey, Pamucak/Aydın), several alkaloids such as lycorine, galanthamine, tazettine, 11-hydroxyvittatine, homolycorine, ismine, trispheridine, galantindole, 6-O-methyoxypretazzettine, dehydroanhydrolycorine, buphanisine, N-demethylgalanthamine crinine, pancracine, hippeastrine, assoanine, 8-O-demethylmaritidine, 9-O-demethylhomolycorine 5,6-dihydrobicolorine, and 2,11-didehydro-2-dehydroxylycorine (1, 2, 3, 8, 10, 12, 13, 16, 19, 20, 36, 38, 39 41, 43, 48, 88, 90, 111 and 133) and galanthane, pancratinine, and obliquine (140, 141 and 142, Figure 9) were isolated [103]. The crude bulb organic extract showed AChE and BuChE inhibition activity with IC50 values of 3.49 and 28.96 µg/mL, respectively [103]. From the same Amaryllidaceae, but collected in Squillace, Calabria region, Italy, ten Amaryllidaceae alkaloids were isolated and identified as tazettine, vittatine, haemanthamine, 11-hydroxyvittatine, homolycorine, pancracine, haemanthidine, and 9-O-demethyllycorine (3, 4, 5, 8, 10, 41, 59 and 90), and obliquine (141) [104]. Haemanthidine (59) was isolated as a scalar mixture of two 6-epimers, as already known for other 6-hydroxycrinine alkaloids. Lycorine and haemanthidine (1 and 59) showed cytotoxic activity on Hacat cells and A431 and AGS cancer cells, while pancracine (41) exhibited selective cytotoxicity against A431 cells [104]. Alkaloids 2, 5, 41 and 59 also showed antiretroviral activity, inhibiting pseudotyped human immunodeficiency virus (HIV)−1 with EC50 values of 25.3 and 18.5 µM, respectively [104]. In addition, all the alkaloids were able to avoid dengue virus (DENV) replication with EC50 values ranging from 0.34 to73.59 µM at low non-cytotoxic concentrations (CC50 ranged from 6.25 µM to >100 µM [104]). Haemanthamine, pancracine, and haemanthidine (5, 41 and 59) appeared to be the most potent anti-DENV inhibitors with EC50 values of 337, 357 and 476 nM, respectively [104].
Several alkaloids were isolated from Galanthus fosteri, diffused mainly in south- and north-central Turkey [105], such as lycorine, galanthamine, galanthine, 11-hydroxyvittatine, ismine, trispheridine, anhydrolycorine, didehydroanhydrolycorine crinine, assoanine, incartine, 5,6-dihydrobicolorine, galwesine, and oxoincartine 9-O-demethylpseudolycorine, (1, 2, 7, 8, 12, 13, 17, 20, 38, 48, 73, 111, 136, and 139), and O,N-dimethylnorbelladine, 9-O-demethylmaritidine, 11-O-acetyl-9-demethylmaritidine, and 3,11-O-diacetyl-9-O-demethylmarititidine (143, 144, 145 and 146, Figure 9) [106].
Leucojum aestivum, commonly named summer snowflake, is a bulbous plant in the Euro-Mediterranean region and is a well-known source of pharmacologically important alkaloids. Among all the alkaloids produced, galanthamine (2) is the major bioactive compound, as well as lycorine (1). L. aestivum is a salt-tolerant plant, and treatment with 4 g/L of CaCl2 increased the amount of galanthamine and the antioxidant activities [107]. Bulbs collected in six different locations in Turkey (Gölcük-Bolu, Yeniçağa-Bolu, Kaynarca-Sakarya, Delmece-Yalova, Uluabat-Bursa and Terkos-Istanbul) at three different growing periods showed that genetic factor is important in alkaloid biosynthesis in the bulbs collected in Gölcük-Bolu, which showed to be the most productive in both alkaloids 1 and 2. The same alkaloids were obtained in abundant amounts from both the bulbs and leaves collected in Delmece-Yalova. The vegetative period followed by ripening are the two periods in which the amount of both alkaloids is high [108].

5. Isocarbostyryls Close to Some Amaryllidaceae Alkaloids

Hippadine (147, Figure 10) is an isocarbostyryl analogue of lycorine isolated together with the alkaloids ungeremine and zefbetaine (89 and 90) from Egyptian P. maritimum, as described in Section 4 [77]. Isocarbostyryl 147 was isolated together with 1-O-acetyl-lycorine, crinsarnine, sarniensinol, bowdensine, and sarniensine (35, 6164) from bulbs of N. sarniensis, as described in Section 2 [49]. In this work, compound 147 and the AA were tested for their mosquitocidal activity against Ae egypti. In adult mosquitos, it showed 23% of mortality [49]. Compound 147 was isolated together with lycorine, crinine, cherylline, and sanguinine (1, 38, 74 and 124), and flexinine, gigantelline, gigantellinine and gigancrinine (148151, Figure 10) from the bulbs of Crinum jagus (syn. = Crinum giganteum) collected in Saint Louis, Senegal [109]. Cherylline, gigantellinine, crinine, flexinine and sanguinine showed inhibitor activity against AChE in a dose-dependent manner, and sanguinine had strong efficacy with an IC50 value of 1.83 ± 0.01 μM, while cherylline and hippadine showed weak cytotoxicity at 100 μM [109].
Among isocarbostyryls, narciclasine and pancratistatin (152 and 153, Figure 10) are amide analogues of lycorine that are very well known for they anticancer activity [6,110,111,112]. Narciclasine (152) was isolated together with haemamthamine (5) from the bulbs of Narcissus pseudonarcissus [113]. Previously, isocarbostyryl 152 was also isolated from Sternbergia lutea, its new process of extraction and purification was optimized, and its NMR spectroscopic full data were assigned [114]. Pancratistatin was extracted for the first time from Hymenocallis littoralis bulbs [115], and it was also isolated from B. haemanthoides bulbs, as reported above in Section 2 [1].
2-Methoxypratosine (154, Figure 10) was isolated, together with AA galanthine, hipeastrine, incartine, narseronine, 3-O-methylnarcissidine, 1-O-acetyl-3-O-methyl, masonine, and narcissidine (7, 43, 73, 95, 97, 120, and 101) from the organic extract of fresh whole N. serotinus, as described in Section 4 [87].
Pratorimine (155, Figure 10) was isolated together with hippadine (147) and AA tazettine, 1-O-acetyl, and 2-O-acetyl-lycorine (3, 35 and 68) from the bulb organic extract of C. buphanoides [63]. Isocarbostyryl 155 was previously isolated together with the two analogues pratosine and pratorinine (156 and 157, Figure 10) as well as hippadine and the AA lycorine and ambelline (1 and 53) from Crinum latifolium [116]. The isocarbostyryls 155 had been firstly isolated from C. latifolium and Crinum pratense Herb. [117].

6. Conclusions

This review describes the chemical and biological properties of alkaloids isolated in the last two decades from different Amaryllidaceae species. The different sections (Section 2, Section 3 and Section 4) report, in detail, the characterization of alkaloids isolated from Amaryllidaceae species diffused in regions of Southern Africa, Andean South America and the Mediterranean basin. The use of plant/alkaloids in folk medicine was also reported in the discussion, and in some cases, the relationships between structure and biological activity as well as their mode of action and biosynthetic pathway were also discussed. This review is divided into four sections, with the first three (Section 2, Section 3 and Section 4) chronologically reporting the alkaloids isolated in the three different cited areas. The results and corresponding studies are summarized in Table 1. The fourth section (Section 5) describes the chemical and the biological characterization of isocarbostyryl isolated from some Amaryllidaceae species in the last two decades in the same world regions, and the results and studies are summarized in Table 1. The content of review is an overview that could be very useful for scientists and readers interested in new pro-drug compounds with potential application in medicine.

Funding

This research received no external funding.

Data Availability Statement

All the data reported in this review were based on SciFinder research using appropriate key words.

Conflicts of Interest

The author declares no conflict of interest.

Sample Availability

Not applicable.

References

  1. Nair, J.J.; Bastida, J.; Codina, C.; Viladomat, F.; van Staden, J. Alkaloids of the South African Amaryllidaceae: A review. Nat. Prod. Commun. 2013, 8, 1335–1350. [Google Scholar] [CrossRef]
  2. Meerow, A.W.; Snijman, D.A. Amaryllidaceae. In The Families and Genera of Vascular Plants; Kubitzki, K., Ed.; Springer: Berlin/Heidelberg, Germany, 1998; Volume 3, pp. 83–110. [Google Scholar]
  3. Heywood, V.H. Flowering Plants of the World; Oxford University Press: New York, NY, USA, 1993. [Google Scholar]
  4. Brochure by Washington State Tourism and Washington State Department of Agriculture. Available online: http://agr.wa.gov/Marketing/SmallFarm/docs/PierceCounty.pdf (accessed on 24 January 2013).
  5. Brochure by the Canadian Cancer Society. Available online: http://www.cancer.ca/ontario/about%20us/media%20centre/od-media%20releases/backgrounder%20about%20daffodil%20days.aspx (accessed on 24 January 2013).
  6. Kornienko, A.; Evidente, A. Chemistry, biology and medicinal potential of narciclasine and its congeners. Chem. Rev. 2008, 108, 1982–2014. [Google Scholar] [CrossRef]
  7. Vargas, F.C. Plant motifs on Inca ceremonial drinking vessels from Peru. Bot. J. Linn. 1981, 82, 313–325. [Google Scholar] [CrossRef]
  8. Dyer, R.A. A review of the genus Brunsvigia. Plant Life 1950, 6, 63–83. [Google Scholar]
  9. Hartwell, J.L. Plants used against cancer. A survey. Lloydia 1967, 30, 379–436. [Google Scholar]
  10. He, M.; Qu, C.; Gao, O.; Hu, X.; Hong, X. Biological and pharmacological activities of Amaryllidaceae alkaloids. RSC Adv. 2015, 5, 16562–16574. [Google Scholar] [CrossRef]
  11. Houghton, P.; Ren, Y.; Howes, M.-J. Acetylcholinesterase inhibitors from plants and fungi. Nat. Prod. Rep. 2006, 23, 181–199. [Google Scholar] [CrossRef]
  12. Nair, J.J.; van Staden, J. Pharmacological and toxicological insights to the South African Amaryllidaceae. Food Chem. Toxicol. 2013, 62, 262–275. [Google Scholar] [CrossRef] [PubMed]
  13. Maroyi, A. Ethnobotanical, phytochemical and pharmacological properties of Crinum bulbispermum (Burm f) Milne-Redh and Schweick (Amaryllidaceae). Trop. J. Pharm. Res. 2016, 15, 2497–2506. [Google Scholar] [CrossRef]
  14. Cahlíková, L.; Vaněčková, N.; Šafratová, M.; Breiterová, K.; Blunden, G.; Hulcová, D.; Opletal, L. The genus Nerine Herb. (Amaryllidaceae): Ethnobotany, phytochemistry, and biological activity. Molecules 2019, 24, 4238. [Google Scholar] [CrossRef]
  15. Nair, J.J.; van Staden, J. Insights to the tribe Haemantheae of the South African Amaryllidaceae. J. Ethnopharmacol. 2022, 292, 115177. [Google Scholar] [CrossRef]
  16. Nair, J.J.; van Staden, J. Pharmacological studies of Crinum, Ammocharis, Amaryllis and Cyrtanthus species of the South African Amaryllidaceae. S. Afr. J. Bot. 2022, 147, 238–244. [Google Scholar] [CrossRef]
  17. Cimmino, A.; Masi, M.; Evidente, M.; Superchi, S.; Evidente, A. Amaryllidaceae alkaloids: Absolute configuration and biological activity. Chirality 2017, 29, 486–499. [Google Scholar] [CrossRef]
  18. Masondo, N.A.; Stafford, G.I.; Aremu, A.O.; Makunga, N.P. Acetylcholinesterase inhibitors from southern African plants: An overview of ethnobotanical, pharmacological potential and phytochemical research including and beyond Alzheimer’s disease treatment. S. Afr. J. Bot. 2019, 120, 39–64. [Google Scholar] [CrossRef]
  19. Masi, M.; Di Lecce, R.; Cimmino, A.; Evidente, A. Advances in the chemical and biological characterization of Amaryllidaceae alkaloids and natural analogues isolated in the last decade. Molecules 2020, 25, 5621. [Google Scholar] [CrossRef] [PubMed]
  20. Arroyo-Leuenberger, S.C. Amaryllidaceae. In Catalogo de las Plantas Vasculares de la Republica Argentina I; Zuloaga, F.O., Morrone, O., Eds.; Missouri Botanical Garden: St. Louis, MO, USA, 1996; pp. 90–100. [Google Scholar]
  21. Ortiz, J.E.; Berkov, S.; Pigni, N.B.; Theoduloz, C.; Roitman, G.; Tapia, A.; Bastida, J.; Feresin, G.E. Wild Argentinian Amaryllidaceae, a new renewable source of the acetylcholinesterase inhibitor galanthamine and other alkaloids. Molecules 2012, 17, 13473–13482. [Google Scholar] [CrossRef]
  22. McNulty, J.; Nair, J.J.; Codina, C.; Bastida, J.; Pandey, S.; Gerasimoff, J.; Griffin, C. Selective apoptosis-inducing activity of crinum-type Amaryllidaceae alkaloids. Phytochemistry 2007, 68, 1068–1074. [Google Scholar] [CrossRef] [PubMed]
  23. Bastida, J.; Lavilla, R.; Viladomat, F. Chemical and biological aspects of Narcissus alkaloids. In The Alkaloids; Cordell, G.A., Ed.; Elsevier Scientific: Amsterdam, The Netherlands, 2006; Volume 63, pp. 87–179. [Google Scholar]
  24. Citoglu, G.; Tanker, M.; Gumusel, B. Antiinflammatory effects of lycorine and haemanthidine. Phytother. Res. 1998, 12, 205–206. [Google Scholar] [CrossRef]
  25. Liu, X.; Jiang, J.; Jiao, X.; Wu, Y.; Lin, J.; Cai, Y. Lycorine induces apoptosis and down-regulation of Mcl-1 in human leukemia cells. Cancer Lett. 2009, 274, 16–24. [Google Scholar] [CrossRef] [PubMed]
  26. Jitsuno, M.; Yokosuka, A.; Sakagami, H.; Mimaki, Y. Chemical constituents of the bulbs of Habranthus brachyandrus and their cytotoxic activities. Chem. Pharm. Bull. 2009, 57, 1153–1157. [Google Scholar] [CrossRef] [PubMed]
  27. Segura, C.; López, D. Contribución al Estudio Químico del Contenido Alcaloídico en Caliphruria subedentata (Amaryllidaceae). Bachelor’s Thesis, Universidad del Cauca, Cauca, Colombia, 2003. [Google Scholar]
  28. Cabezas, F.; López, D.; Segura, C.; Codina, C.; Bastida, J.; Viladomsat, F. Alcaloides de Caliphruria subedentata Baker (Amaryllidaceae). Actual. Biológicas 2005, 27 (Suppl. S1), 141. [Google Scholar]
  29. Meerow, A.W. Systematics of the Amazon Lilies: Eucharis and Caliphruria (Amaryllidaceae). Ann. Mo. Bot. Gard. 1989, 76, 136–220. [Google Scholar] [CrossRef]
  30. Cabezas, F.F. Biogénesis, Obtención y Caracterización de Alcaloides de Amaryllidaceae; Editorial Universidad del Valle: Calì, Colombia, 2012. [Google Scholar]
  31. Cabezas, F.; Pigni, N.; Bastida, J.; Codina, C.; Viladomat, F. Análisis del contenido alcaloidico de Caliphruria subdentata Baker (Amaryllidaceae) por el método CG-EM. Rev. Latinoam. Quim. 2013, 41, 68–73. [Google Scholar]
  32. Guo, Y.; de Andrade, J.P.; Pigni, N.B.; Torras-Claveria, L.; Tallini, L.R.; de S. Borges, W.; Viladomat, F.; Nair, J.J.; Zuanazz, J.A.S.; Bastida, J. New alkaloids from Hippeastrum papilio (Ravenna) van Scheepen. Helv. Chim. Acta 2016, 99, 143–147. [Google Scholar] [CrossRef]
  33. Moraga-Nicolás, F.; Jara, C.; Godoy, R.; Iturriaga-Vásquez, P.; Venthur, H.; Quiroz, A.; Becerra, J.; Mutis, A.; Hormazábal, E. Rhodolirium andicola: A new renewable source of alkaloids with acetylcholinesterase inhibitory activity, a study from nature to molecular docking. Rev. Bras. Farmacogn. 2018, 28, 34–43. [Google Scholar] [CrossRef]
  34. Rojas-Vera, J.D.C.; Buitrago-Diaz, A.A.; Possamai, L.M.; Timmers, L.F.; Tallini, L.R.; Bastida, J. Alkaloid profile and cholinesterase inhibition activity of five species of Amaryllidaceae family collected from Mérida state-Venezuela. S. Afr. J. Bot. 2021, 136, 126–136. [Google Scholar] [CrossRef]
  35. Huaylla, H.; Llalla, O.; Torras-Claveria, L.; Bastida, J. Alkaloid profile in Pyrolirion albicans Herb. (Amaryllidaceae), a Peruvian endemic species. S. Afr. J. Bot. 2021, 136, 76–80. [Google Scholar] [CrossRef]
  36. Reis, A.; Magne, K.; Massot, S.; Tallini, L.R.; Scopel, M.; Bastida, J.; Ratet, P.; Zuanazzi, J.A.S. Amaryllidaceae alkaloids: Identification and partial characterization of montanine production in Rhodophiala bifida plant. Sci. Rep. 2019, 9, 8471–8481. [Google Scholar] [CrossRef]
  37. Castilhos, T.; Girdani, R.B.; Henriques, A.T.; Menez, A.T.; Zuanazzi, J.A.S. Avaliaçäo in vitro das ativadades antiinflamatória, ntioxidante e antimicrobica do alcalóde montanina. Rev. Bras. Farmacogn. 2007, 17, 209–214. [Google Scholar] [CrossRef]
  38. Da Silva, A.F.; de Andrade, J.P.; Bevilaqua, L.R.; de Souza, M.M.; Izquierdo, I.; Henriques, A.T.; Zuanazzi, J.A.S. Anxiolytic-, antidepressant- and anticonvulsant-like effects of the alkaloid montanine isolated from Hippeastrum vittatum. Pharmacol. Biochem. Behav. 2006, 85, 148–154. [Google Scholar] [CrossRef]
  39. Farinon, M.; Clarimundo, V.S.; Pedrazza, G.P.; Gulko, P.S.; Zuanazzi, J.A.; Xavier, R.M.; de Oliveira, P.G. Disease modifying anti-rheumatic activity of the alkaloid montanine on experimental arthritis and fibroblast-like synoviocytes. Eur. J. Pharmacol. 2017, 799, 180–187. [Google Scholar] [CrossRef]
  40. Tallini, L.R.; Andrade, J.P.D.; Kaiser, M.; Viladomat, F.; Nair, J.J.; Zuanazzi, J.A.S.; Bastida, J. Alkaloid constituents of the Amaryllidaceae plant Amaryllis belladonna L. Molecules 2017, 22, 1437. [Google Scholar] [CrossRef] [PubMed]
  41. Gomes-Copeland, K.K.P.; Meireles, C.G.; Gomes, J.V.D.; Torres, A.G.; Sinoti, S.B.P.; Fonseca-Bazzo, Y.M.; de Oliveira Magalhães, P.; Fagg, C.D.; Luiz Alberto Simeoni, L.A.; Silveira, D. Hippeastrum stapfianum (Kraenzl.) RS Oliveira & Dutilh (Amaryllidaceae) Ethanol extract activity on acetylcholinesterase and PPAR-α/γ Receptors. Plants 2022, 11, 3179. [Google Scholar]
  42. Costa, G.G.P.; Silva, C.A.G.; Gomes, J.V.D.; Torres, A.G.; Santos, I.R.I.; Almeida, F.T.C.; Fagg, C.W.; Simeoni, L.A.; Silveira, D.; Gomes-Copeland, K.K.P. Influence of in vitro micropropagation on lycorine biosynthesis and anticholinesterase activity in Hippeastrum goianum. Rev. Bras. Farmacogn. 2019, 29, 262–265. [Google Scholar] [CrossRef]
  43. Soto-Vásquez, M.R.; Rodríguez-Muñoz, C.A.; Tallini, L.R.; Bastida, J. Alkaloid composition and biological activities of the Amaryllidaceae species Ismene amancaes (Ker Gawl.) Herb. Plants 2022, 11, 1906. [Google Scholar] [CrossRef]
  44. Nair, J.J.; Rárová, L.; Strnad, M.; Bastida, J.; van Staden, J. Alkaloids from Boophone haemanthoides (Amaryllidaceae). Nat. Prod. Commun. 2013, 8, 1934578X1300801211. [Google Scholar] [CrossRef]
  45. Wrinkle, G. An introduction to the genus Boophane. Herbertia 1984, 40, 77–82. [Google Scholar]
  46. Ibrakaw, A.S.; Omoruyi, S.I.; Ekpo, O.E.; Hussein, A.A. Neuroprotective activities of Boophone haemanthoides (Amaryllidaceae) extract and its chemical constituents. Molecules 2020, 25, 5376. [Google Scholar] [CrossRef] [PubMed]
  47. Hutchings, A.; Scott, A.H.; Lewis, G.; Cunningham, A.B. Zulu Medicinal Plants: An Inventory; University of Natal Press: Pietermaritzburg, South Africa, 1996. [Google Scholar]
  48. Nair, J.J.; Manning, J.C.; van Staden, J. Didticamine, a chemotaxonomic marker for the genus Boophone Herb. (Amaryllidacea). S. Afr. J. Bot. 2012, 83, 89–91. [Google Scholar] [CrossRef]
  49. Masi, M.; Cala, A.; Tabanca, N.; Cimmino, A.; Green, I.R.; Bloomquist, J.R.; van Otterlo, W.A.L.; Macias, F.A.; Evidente, A. Alkaloids with activity against the Zika virus vector Aedes aegypti (L.)—Crinsarnine and sarniensinol, two new crinine and mesembrine type alkaloids isolated from the South African plant Nerine sarniensis. Molecules 2016, 21, 1432. [Google Scholar] [CrossRef]
  50. Rasmussen, S.A.; Jamieson, D.J.; Honein, M.A.; Petersen, L.R. Zika virus and birth defects—Reviewing the evidence for causality. N. Engl. J. Med. 2016, 374, 1981–1987. [Google Scholar] [CrossRef]
  51. Neuwinger, H.D. African Ethnobotany: Poisons and Drugs; Chapman and Hall: London, UK, 1994. [Google Scholar]
  52. Nair, J.J.; van Staden, J. Traditional usage, phytochemistry and pharmacology of the South African medicinal plant Boophone disticha (L.f.) Herb. (Amaryllidaceae). J. Ethnopharmacol. 2014, 151, 12–26. [Google Scholar] [CrossRef] [PubMed]
  53. Adewusi, E.A.; Fouche, G.; Steenkamp, V. Cytotoxicity and acetylcholinesterase inhibitory activity of an isolated crinine alkaloid from Boophone disticha (Amaryllidaceae). J. Ethnopharmacol. 2012, 143, 572–578. [Google Scholar] [CrossRef]
  54. Cheesman, L.; Nair, J.J.; van Staden, J. Antibacterial activity of crinane alkaloids from Boophone disticha (Amaryllidaceae). J. Ethnopharmacol. 2012, 140, 405–408. [Google Scholar] [CrossRef] [PubMed]
  55. De Smet, P.A.G.M. Some ethnopharmacological notes on African hallucinogens. J. Ethnopharmacol. 1996, 50, 141–146. [Google Scholar] [CrossRef] [PubMed]
  56. Hauth, H.; Stauffacher, D. Die alkaloide von Buphane disticha (L.f.) Herb. Helv. Chim. Acta 1961, 44, 491–502. [Google Scholar] [CrossRef]
  57. Neergaard, J.S.; Andersen, J.; Pedersen, M.E.; Stafford, G.I.; van Staden, J.; Jäger, A.K. Alkaloids from Boophone disticha with affinity to the serotonin transporter. S. Afr. J. Bot. 2009, 75, 371–374. [Google Scholar] [CrossRef]
  58. Sandager, M.; Nielse, N.D.; Stafford, G.I.; van Staden, J.; Jäger, A.K. Alkaloids from Boophone disticha with affinity to the serotonin transporter in rat brain. J. Ethnopharmacol. 2005, 98, 367–370. [Google Scholar] [CrossRef] [PubMed]
  59. Watt, J.M.; Breyer-Brandwijk, M.G. The Medicinal and Poisonous Plants of Southern and Eastern Africa, 2nd ed.; E & S Livingstone: London, UK, 1962. [Google Scholar]
  60. Dictionary of Natural Products. CD-ROM, Version 16.2; Chapman and Hall: London, UK, 2008.
  61. Van Rensburg, E.; Zietsman, P.C.; Bonnet, S.L.; Wilhelm, A. Alkaloids from the bulbs of Boophone disticha. Nat. Prod. Commun. 2017, 12, 1934578X1701200911. [Google Scholar] [CrossRef]
  62. Naidoo, D.; Slavětínská, L.P.; Aremu, A.O.; Gruz, J.; Biba, O.; Doležal, K.; Van Staden, J.; Finnie, J.F. Metabolite profiling and isolation of biologically active compounds from Scadoxus puniceus, a highly traded South African medicinal plant. Phytother. Res. 2018, 32, 625–630. [Google Scholar] [CrossRef]
  63. Masi, M.; Mubaiwa, B.; Mabank, T.; Karakoyun, C.; Cimmino, A.; van Otterlo, W.A.L.; Green, I.R.; Evidente, A. Alkaloids isolated from indigenous South African Amaryllidaceae: Crinum buphanoides (Welw. ex Baker), Crinum graminicola (I. Verd.), Cyrtanthus mackenii (Hook. f) and Brunsvigia grandiflora (Lindl). S. Afr. J. Bot. 2018, 118, 188–191. [Google Scholar] [CrossRef]
  64. Veale, C.G.; Chen, W.; Chaudhary, S.; Kituyi, S.N.; Isaacs, M.; Hoppe, H.; Edkins, A.L.; Combrinck, S.; Mehari, B.; Viljoen, A. NMR structural elucidation of channaine, an unusual alkaloid from Sceletium tortuosum. Phytochem. Lett. 2018, 23, 189–193. [Google Scholar] [CrossRef]
  65. Gericke, N.; Viljoen, A.M. Sceletium—A review update. J. Ethnopharmacol. 2008, 119, 653–663. [Google Scholar] [CrossRef] [PubMed]
  66. Harvey, A.L.; Young, L.C.; Viljoen, A.M.; Gericke, N.P. Pharmacological actions of the South African medicinal and functional food plant Sceletium tortuosum and its principal alkaloids. J. Ethnopharmacol. 2011, 137, 1124–1129. [Google Scholar] [CrossRef] [PubMed]
  67. Bodendorf, K.; Krieger, W. Über die alkaloide von Mesembryanthemum tortuosum L. Arch. Pharm. 1957, 290, 441–448. [Google Scholar] [CrossRef] [PubMed]
  68. Popelak, A.; Lettenbauer, G. The mesembrine alkaloids. Alkaloids Chem. Physiol. 1967, 9, 467–482. [Google Scholar]
  69. Masi, M.; Gunawardana, S.; van Rensburg, M.J.; James, P.C.; Mochel, J.G.; Heliso, P.S.; Albalawi, A.S.; Cimmino, A.; van Otterlo, W.A.L.; Kornienko, A.; et al. Alkaloids isolated from Haemanthus humilis Jacq., an indigenous South African Amaryllidaceae: Anticancer activity of coccinine and montanine. S. Afr. J. Bot. 2019, 126, 277–281. [Google Scholar] [CrossRef]
  70. Nair, J.J.; Andresen, A.L.; Papenfus, H.B.; Staerk, D.; van Staden, J. Identification of Amaryllidaceae alkaloids in larvae of the ‘lily borer’moth Brithys crini. S. Afr. J. Bot. 2020, 131, 351–359. [Google Scholar] [CrossRef]
  71. Mason, L.H.; Puschett, E.R.; Wildman, W.C. Alkaloids of the Amaryllidaceae. IV. Crystalline alkaloids from Ammocharis coranica (Ker-Gawl) Herb., Brunsvigia rosea (Lam.) Hannibal and two Crinum species. J. Am. Chem. Soc. 1955, 77, 1253–1256. [Google Scholar] [CrossRef]
  72. Hauth, H.; Stauffacher, D. Die alkaloide von Ammocharis coranica (Ker-Gawl.) Herb. emend. M-Redh. 3. Mitteilung uber Amaryllidaceen-alkaloide. Helv. Chim. Acta 1962, 45, 1307–1317. [Google Scholar] [CrossRef]
  73. Koorbanally, N.A.; Mulholland, D.A.; Crouch, N.R. Alkaloids and triterpenoids from Ammocharis coranica. Phytochemistry 2000, 54, 93–97. [Google Scholar] [CrossRef] [PubMed]
  74. Duncan, G.; Jeppe, B.; Voigt, L. The Amaryllidaceae of southern Africa; Umdaus Press: Pretoria, South Africa, 2016. [Google Scholar]
  75. Raghoo, M.; Crouch, N.R.; Koorbanally, N.A. A novel and unusual tetramethoxylated crinine alkaloid from Ammocharis coranica (Amaryllidaceae). S. Afr. J. Bot. 2021, 137, 451–454. [Google Scholar] [CrossRef]
  76. Šafratová, M.; Křoustková, J.; Maafi, N.; Suchánková, D.; Vrabec, R.; Chlebek, J.; Kuneš, J.; Opletal, L.; Bucar, F.; Cahlíková, L. Amaryllidaceae alkaloids from Clivia miniata (Lindl.) Bosse (Amaryllidaceae): Isolation, structural elucidation, and biological activity. Plants 2022, 11, 3034. [Google Scholar] [CrossRef]
  77. Abou-Donia, A.H.; Abib, A.A.; El Din, A.S.; Evidente, A.; Gaber, M.; Scopa, A. Two betaine-type alkaloids from Egyptian Pancratium maritimum. Phytochemistry 1992, 31, 2139–2141. [Google Scholar] [CrossRef]
  78. Ahmed, Z.F.; Rizk, A.M.; Hamouda, F.M. Phytochemical studies on Egyptian Pancratium species. Lloydia 1964, 27, 115. [Google Scholar]
  79. Ali, A.A.; Mesbah, M.K.; Mohamed, M.H. Study of alkaloids of Panratium maritimum L. growing in Egypt. Bull. Pharm. Sci Assiut 1984, 7, 351–362. [Google Scholar] [CrossRef]
  80. Schrader, K.K.; Andolfi, A.; Cantrell, C.L.; Cimmino, A.; Duke, S.O.; Osbrink, W.; Wedge, D.E.; Evidente, A. A survey of phytotoxic microbial and plant metabolites as potential natural products for pest management. Chem. Biodivers. 2010, 7, 2261–2280. [Google Scholar] [CrossRef]
  81. Karakoyun, Ç.; Masi, M.; Cimmino, A.; Önür, M.A.; Somer, N.U.; Kornienko, A.; Evidente, A. A brief up-to-date overview of Amaryllidaceae alkaloids: Phytochemical studies of Narcissus tazetta subsp. tazetta L., collected in Turkey. Nat. Prod. Commun. 2019, 14, 1934578X19872906. [Google Scholar]
  82. Schrader, K.K.; Avolio, F.; Andolfi, A.; Cimmino, A.; Evidente, A. Ungeremine and its hemisynthesized analogues as bactericides against Flavobacterium columnare. J. Agric. Food Chem. 2013, 61, 1179–1183. [Google Scholar] [CrossRef]
  83. Valerio, F.; Masi, M.; Cimmino, A.; Moeini, S.A.; Lavermicocca, P.; Evidente, A. Antimould microbial and plant metabolites with potential use in intelligent food packaging. Nat. Prod. Res. 2018, 32, 1605–1610. [Google Scholar] [CrossRef]
  84. Moeini, A.; Cimmino, A.; Dal Poggetto, G.; Di Biase, M.; Evidente, A.; Masi, M.; Lavermicocca, P.; Valerio, F.; Leone, A.; Santagata, G.; et al. Effect of pH and TPP concentration on chemico-physical properties, release kinetics and antifungal activity of Chitosan-TPP-Ungeremine microbeads. Carbohydr. Polym. 2018, 195, 631–641. [Google Scholar] [CrossRef]
  85. Moeini, A.; Mallardo, S.; Cimmino, A.; Dal Poggetto, G.; Masi, M.; Di Biase, M.; van Reenen, A.; Lavermicocca, P.; Valerio, F.; Evidente, A.; et al. Thermoplastic starch and bioactive chitosan sub-microparticle biocomposites: Antifungal and chemico-physical properties of the films. Carbohydr. Polym. 2020, 230, 115627. [Google Scholar] [CrossRef] [PubMed]
  86. Pigni, N.B.; Berkov, S.; Elamrani, A.; Benaissa, M.; Viladomat, F.; Codina, C.; Bastida, J. Two new alkaloids from Narcissus serotinus L. Molecules 2010, 15, 7083–7089. [Google Scholar] [CrossRef] [PubMed]
  87. Pigni, N.B.; Rios-Ruiz, S.; Martinez-Frances, V.; Nair, J.J.; Viladomat, F.; Codina, C.; Bastida, J. Alkaloids from Narcissus serotinus. J. Nat. Prod. 2012, 75, 1643–1647. [Google Scholar] [CrossRef] [PubMed]
  88. Pigni, N.B.; Ríos-Ruiz, S.; Luque, F.J.; Viladomat, F.; Codina, C.; Bastida, J. Wild daffodils of the section Ganymedes from the Iberian Peninsula as a source of mesembrane alkaloids. Phytochemistry 2013, 95, 384–393. [Google Scholar] [CrossRef]
  89. Berkov, S.; Martínez-Francés, V.; Bastida, J.; Codina, C.; Ríos, S. Evolution of alkaloid biosynthesis in the genus Narcissus. Phytochemistry 2014, 99, 95–106. [Google Scholar] [CrossRef]
  90. Meerow, A.W.; Francisco-Ortega, J.; Kuhn, D.N.; Schnell, R.J. Phylogenetic relationships and biogeography within the Eurasian clade of Amaryllidaceae based on plastid ndhF and nrDNA ITS sequences: Lineage sorting in a reticulate area? Syst. Bot. 2006, 31, 42–60. [Google Scholar] [CrossRef]
  91. Fernandes, A. Sur la caryologie de Lapiedra martinezii Lag. Bol. Soc. Broterialui 1950, 24, 281–319. [Google Scholar]
  92. Ríos, S.; Berkov, S.; Martínez-Francés, V.; Bastida, J. Biogeographical patterns and phenological changes in Lapiedra martinezii Lag. related to its alkaloid diversity. Chem. Biodivers. 2013, 10, 1220–1238. [Google Scholar] [CrossRef]
  93. Iannello, C.; Pigni, N.B.; Antognoni, F.; Poli, F.; Maxia, A.; de Andrade, J.P.; Bastida, J. A potent acetylcholinesterase inhibitor from Pancratium illyricum L. Fitoterapia 2014, 92, 163–167. [Google Scholar] [CrossRef]
  94. Bores, G.M.; Huger, F.P.; Petko, W.; Mutlib, A.E.; Camacho, F.; Rush, D.K.; Selk, D.E.; Wolf, V.; Kosley, R.W., Jr.; Davis, L.; et al. Pharmacological evaluation of novel Alzheimer's disease therapeutics: Acetylcholinesterase inhibitors related to galanthamine. J. Pharmacol. Exp. Ther. 1996, 277, 728–738. [Google Scholar]
  95. de Andrade, J.P.; Berkov, S.; Viladomat, F.; Codina, C.; Zuanazzi, J.A.S.; Bastida, J. Alkaloids from Hippeastrum papilio. Molecules 2011, 16, 7097–7104. [Google Scholar] [CrossRef] [PubMed]
  96. LóPez-TIrado, J. Narcissus munozii-alvarezii (Amaryllidaceae): A new hybrid from southern Spain. Phytotaxa 2018, 364, 267–274. [Google Scholar] [CrossRef]
  97. Masi, M.; Frolova, L.V.; Yu, X.; Mathieu, V.; Cimmino, A.; De Carvalho, A.; Kiss, R.; Rogelj, S.; Pertsemlidis, A.; Kornienko, A.; et al. Jonquailine, a new pretazettine-type alkaloid isolated from Narcissus jonquilla quail, with activity against drug-resistant cancer. Fitoterapia 2015, 102, 41–48. [Google Scholar] [CrossRef]
  98. Shawky, E. Phytochemical and biological investigation of Clivia nobilis flowers cultivated in Egypt. Iran. J. Pharm. Res. 2016, 15, 531–535. [Google Scholar] [PubMed]
  99. Bozkurt, B.; Coban, G.; Kaya, G.I.; Onur, M.A.; Unver-Somer, N. Alkaloid profiling, anticholinesterase activity and molecular modeling study of Galanthus elwesii. S. Afr. J. Bot. 2017, 113, 119–127. [Google Scholar] [CrossRef]
  100. Evidente, A.; Iasiello, I.; Randazzo, G. Hippamine, a minor alkaloid from Sternbergia lutea. J. Nat. Prod. 1984, 47, 1061–1062. [Google Scholar] [CrossRef]
  101. Ferri, D.; Ubaldi, C.; Marcozzi, G.; Fasciani, P.; Bacchetta, L.; Pace, L. Chemical characterization of Narcissus poeticus from Sirente–Velino (Apennines-Italy): Galantamine accumulation and distribution of allergenic compounds in the flower. Nat. Prod. Commun. 2017, 12, 1934578X1701200105. [Google Scholar] [CrossRef]
  102. Bendaif, H.; Melhaoui, A.; Ramdani, M.; Elmsellem, H.; Douez, C.; El Ouadi, Y. Antibacterial activity and virtual screening by molecular docking of lycorine from Pancratium foetidum Pom (Moroccan endemic Amaryllidaceae). Microb. Pathog. 2018, 115, 138–145. [Google Scholar] [CrossRef]
  103. Bozkurt, B.; Kaya, G.I.; Somer, N.U. Chemical composition and enzyme inhibitory activities of Turkish Pancratium maritimum bulbs. Nat. Prod. Commun. 2019, 14, 1934578X19872905. [Google Scholar] [CrossRef]
  104. Masi, M.; Di Lecce, R.; Mérindol, N.; Girard, M.P.; Berthoux, L.; Desgagné-Penix, I.; Calabrò, V.; Evidente, A. Cytotoxicity and antiviral properties of alkaloids isolated from Pancratium maritimum. Toxins 2022, 14, 262. [Google Scholar] [CrossRef]
  105. Davis, A.P. The genus Galanthus-snowdrops in the wild. In Snowdrops, A Monograph of Cultivated Galanthus; Bishop, M., Grimshaw, J., Davis, A.P., Eds.; Griffin Press Publishing Ltd.: Cheltenham, UK, 2006; pp. 9–63. [Google Scholar]
  106. Ahmet, E.; Ceren, E.; Bozkurt, B.; Somer, N.Ü. GC/MS analysis of alkaloids in Galanthus fosteri baker and determination of its anticholinesterase activity. Turk. J. Pharm. Sci. 2020, 17, 36–42. [Google Scholar]
  107. Ates, M.T.; Yildirim, A.B.; Turker, A.U. Enhancement of alkaloid content (galanthamine and lycorine) and antioxidant activities (enzymatic and non-enzymatic) unders salt stress in summer snowflake (Leucojum aestivum L.). S. Afr. J. Bot. 2021, 140, 182–188. [Google Scholar] [CrossRef]
  108. Demir, S.C.; Yildirim, A.B.; Turker, A.U.; Eker, I. Seasonal variation in alkaloid content, phenolic constituent and biological activities of some Leucojum aestivum L. populations in Turkey. S. Afr. J. Bot. 2022, 147, 713–723. [Google Scholar] [CrossRef]
  109. Ka, S.; Mérindol, N.; Seck, I.; Ricard, S.; Diop, A.; Boye, C.S.B.; Landelouci, K.; Daoust, B.; Berthoux, L.; Pépin, G.; et al. Biological investigation of amaryllidaceae alkaloid extracts from the bulbs of Pancratium trianthum collected in the senegalese flora. Molecules 2021, 26, 7382. [Google Scholar] [CrossRef]
  110. Ingrassia, L.; Lefranc, F.; Mathieu, V.; Darro, F.; Kiss, R. Amaryllidaceae isocarbostyril alkaloids and their derivatives as promising antitumor agents. Transl. Oncol. 2008, 1, 1–13. [Google Scholar] [CrossRef] [PubMed]
  111. Van Goietsenoven, G.; Mathieu, V.; Lefranc, F.; Kornienko, A.; Evidente, A.; Kiss, R. Narciclasine as well as other Amaryllidaceae isocarbostyrils are promising GTP-ase targeting agents against brain cancers. Med. Res. Rev. 2013, 33, 439–455. [Google Scholar] [CrossRef] [PubMed]
  112. Mathieu, V.; Laguera, B.; Masi, M.; Dulanto, S.A.; Bingham, T.W.; Hernandez, L.W.; Sarlah, D.; Evidente, A.; Lafontaine, D.L.J.; Kornienko, K.; et al. Amaryllidaceae alkaloids decrease the proliferation, invasion, and secretion of clinically relevant cytokines by cultured human colon cancer cells. Biomolecules 2022, 12, 1267. [Google Scholar] [CrossRef]
  113. Kempthorne, C.J.; Borra, S.; Kumar, M.; Dokuburra, C.B.; Liscombe, D.K.; McNulty, J. Identification of haemanthamine as a phytotoxic alkaloid in Narcissus pseudonarcissus L. (Daffodil) emerging buds. Nat. Prod. Res. 2023, 1–7. [Google Scholar] [CrossRef] [PubMed]
  114. Evidente, A. Narciclasine: 1H-and 13C-NMR Data and a new improved method of preparation. Planta Med. 1991, 57, 293–295. [Google Scholar] [CrossRef] [PubMed]
  115. Pettit, G.R.; Gaddamidi, V.; Herald, D.L.; Singh, S.B.; Cragg, G.M.; Schmidt, J.M.; Boettner, F.E.; Williams, M.; Sagawa, Y. Antineoplastic agents, 120. Pancratium littorale. J. Nat. Prod. 1986, 49, 995–1002. [Google Scholar] [CrossRef] [PubMed]
  116. Ghosal, S.; Saini, K.S.; Frahm, A.W. Alkaloids of Crinum latifolium. Phytochemistry 1983, 22, 2305–2309. [Google Scholar] [CrossRef]
  117. Ghosal, S.; Rao, P.H.; Jaiswal, D.K.; Kumar, Y.; Frahm, A.W. Alkaloids of Crinum pretense. Phytochemistry 1981, 20, 2003–2007. [Google Scholar] [CrossRef]
Figure 1. Some representative Amaryllidaceae genera [2]: (A) Ammocharis coranica; (B) Boophone disticha; (C) Brunsvigia radulosa; (D) Clivia miniata; (E) Crinum delagoense; (F) Cyrtanthus contractus; (G) Gethyllis ciliaris; (H) Haemanthus albiflos; (I) Nerine filifolia; (J) Sternbergia lutea; (K) Pancratium maritimum; (L) Narcissus tazetta; (M) Amaryllis belladonna. The pictures are original photos obtained by the author.
Figure 1. Some representative Amaryllidaceae genera [2]: (A) Ammocharis coranica; (B) Boophone disticha; (C) Brunsvigia radulosa; (D) Clivia miniata; (E) Crinum delagoense; (F) Cyrtanthus contractus; (G) Gethyllis ciliaris; (H) Haemanthus albiflos; (I) Nerine filifolia; (J) Sternbergia lutea; (K) Pancratium maritimum; (L) Narcissus tazetta; (M) Amaryllis belladonna. The pictures are original photos obtained by the author.
Molecules 28 04055 g001
Figure 2. Alkaloids produced by Andean Amaryllidaceae plants: Habranthus jamesonii, Phycella herbertiana, Rhodophiala mendocina and Zephyranthes filifolia (16) H. jamesonii and P. herbertiana (7 and 8), Caliphruria subedentata (1, 2, 5, and 1016 and 18, 19 and 21), Hippeastrum papilio (2, 5, 14, 24 and 25) and Rhodolirium andicola (2, 5, 6, 18, 21, 2830 and 32, 34) Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata (1, and 35), Pyrolirion albicans (2, 4, and 38, 42 and 43), Amaryllis belladonna (44), and Ismene amancaes (1, 2, 6, 7, and 42 and 4850).
Figure 2. Alkaloids produced by Andean Amaryllidaceae plants: Habranthus jamesonii, Phycella herbertiana, Rhodophiala mendocina and Zephyranthes filifolia (16) H. jamesonii and P. herbertiana (7 and 8), Caliphruria subedentata (1, 2, 5, and 1016 and 18, 19 and 21), Hippeastrum papilio (2, 5, 14, 24 and 25) and Rhodolirium andicola (2, 5, 6, 18, 21, 2830 and 32, 34) Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata (1, and 35), Pyrolirion albicans (2, 4, and 38, 42 and 43), Amaryllis belladonna (44), and Ismene amancaes (1, 2, 6, 7, and 42 and 4850).
Molecules 28 04055 g002
Figure 3. Alkaloids produced by Caliphruria subedentata (17, 20), Hippeastrum papilio (22, 23, 26 and 27) and Rhodolirium andicola (31 and 33) Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata (20, 33, and 36), Pyrolirion albicans (37 and 39), Amaryllis belladonna (4547), Hippeastrum stapfianum (47), and Ismene amancaes (17, 20 and 51).
Figure 3. Alkaloids produced by Caliphruria subedentata (17, 20), Hippeastrum papilio (22, 23, 26 and 27) and Rhodolirium andicola (31 and 33) Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata (20, 33, and 36), Pyrolirion albicans (37 and 39), Amaryllis belladonna (4547), Hippeastrum stapfianum (47), and Ismene amancaes (17, 20 and 51).
Molecules 28 04055 g003
Figure 4. Alkaloids produced by South Africa Amaryllidaceae plants: Boophone haemanthoides (5260), Nerine sarniensis (6164), Boophone disticha (6567), Crinum buphanoides (68), Crinum graminicola (69), Sceletium tortuosum (70), Haemanthus humilis (71), Ammocharis coranica (75, 7779 and 81), and Clivia miniata (86).
Figure 4. Alkaloids produced by South Africa Amaryllidaceae plants: Boophone haemanthoides (5260), Nerine sarniensis (6164), Boophone disticha (6567), Crinum buphanoides (68), Crinum graminicola (69), Sceletium tortuosum (70), Haemanthus humilis (71), Ammocharis coranica (75, 7779 and 81), and Clivia miniata (86).
Molecules 28 04055 g004
Figure 5. Alkaloids produced by South Africa Amaryllidaceae plants: Haemanthus humilis (72 and 73), Crinum moorei (74), Ammocharis coranica (76, 80 and 81), and Clivia miniata (8285 and 8789).
Figure 5. Alkaloids produced by South Africa Amaryllidaceae plants: Haemanthus humilis (72 and 73), Crinum moorei (74), Ammocharis coranica (76, 80 and 81), and Clivia miniata (8285 and 8789).
Molecules 28 04055 g005
Figure 6. Alkaloids produced by Mediterranean Amaryllidaceae plants: Pancratium maritimum (9093), Narcissus serotinus L. (94101), and Narcissus triandrus (102107).
Figure 6. Alkaloids produced by Mediterranean Amaryllidaceae plants: Pancratium maritimum (9093), Narcissus serotinus L. (94101), and Narcissus triandrus (102107).
Molecules 28 04055 g006
Figure 7. Alkaloids produced by Mediterranean Amaryllidaceae plants: Narcissus triandrus (108 and 109) and Lapiedra martinezii (110121).
Figure 7. Alkaloids produced by Mediterranean Amaryllidaceae plants: Narcissus triandrus (108 and 109) and Lapiedra martinezii (110121).
Molecules 28 04055 g007
Figure 8. Alkaloids produced by Mediterranean Amaryllidaceae plants: Pancratium illyricum L. (122127), Narcissus jonquilla quail (128), Clivia nobilis (129 and 130), and Galanthus elwesii (131138).
Figure 8. Alkaloids produced by Mediterranean Amaryllidaceae plants: Pancratium illyricum L. (122127), Narcissus jonquilla quail (128), Clivia nobilis (129 and 130), and Galanthus elwesii (131138).
Molecules 28 04055 g008
Figure 9. Alkaloids produced by Mediterranean Amaryllidaceae plants: Galanthus elwesii (138), Narsissus tazetta a subsp. tazetta (139), Pancratium maritimum (140142), and Galanthus fosteri (143146).
Figure 9. Alkaloids produced by Mediterranean Amaryllidaceae plants: Galanthus elwesii (138), Narsissus tazetta a subsp. tazetta (139), Pancratium maritimum (140142), and Galanthus fosteri (143146).
Molecules 28 04055 g009
Figure 10. Isocarbostyryl isolated from Pancratium maritimum, Nerine sarniensis, Crinum jagus, and Crinum buphanoides (147), Narcissus pseudonarcissus and Sternbergia lutea (152), Hymenocallis littoralis and Boophane haemanthoides (153), Narcissus serotinus (154), Crinum buphanoide (155157), and Crinum latifolium and Crinum pretense (147 and 155157). Alkaloids isolated from C. jagus (148151).
Figure 10. Isocarbostyryl isolated from Pancratium maritimum, Nerine sarniensis, Crinum jagus, and Crinum buphanoides (147), Narcissus pseudonarcissus and Sternbergia lutea (152), Hymenocallis littoralis and Boophane haemanthoides (153), Narcissus serotinus (154), Crinum buphanoide (155157), and Crinum latifolium and Crinum pretense (147 and 155157). Alkaloids isolated from C. jagus (148151).
Molecules 28 04055 g010
Table 1. Alkaloids and isocarbostyrls produced by Amarylldaceae spp. of Andean South America, South Africa, and the Mediterranean basin in the last two decades.
Table 1. Alkaloids and isocarbostyrls produced by Amarylldaceae spp. of Andean South America, South Africa, and the Mediterranean basin in the last two decades.
AlkaloidSubgroupAmaryllidaceaeBiological ActivityReference
AA from Andean South America
Lycorine (1)LycoraneHabranthus jamesonii, Phycella herbertiana and Rhodophiala mendocinaNot reported[20]
Cytotoxic, apoptotic, antiviral, antifungal, and antiprotozoan[6,22]
Anti-inflammatory[24]
Antileukemia[25]
Caliphruria subedentataNot reported[26,28]
Crinum amabile, Crinum erubescens, Crinum moorei, Amaryllis belladonna and Zephyranthes carinata[34]
Pyrolirion albicans[35]
Hippeastrum stapfianum[41]
Ismene amancaes[43]
Boophone haemanthoidesCytotoxic[44]
Crinum buphanoides, Crinum graminicola, Cyrtanthus mackenii and Brunsvigia grandifloraNot reported[63]
Ammocharis coranica[71]
Clivia miniataInhibition of AChE/BUChE[76]
Pancratium maritimumNot reported[77,78]
Lapiedra martineziiNot reported[92]
Pancratium illyricum[93]
Clivia nobilis[98]
Galanthus elwesii[99]
Pancratium foetidumAntibiotic[102]
Narcissus tazetta subsp. tazettaNot reported[81]
Galanthus fosteri[106]
Leucojum aestivum[107,108]
Galanthamine (2)GalanthamineH. jamesonii, P. herbertiana, R. mendocina and Zephyranthes filifoliaInhibition of acetylcolinestarase[20]
Anticancer[25]
C. subedentataCytotoxic[27,28]
Hippeastrum papilioNot reported[32]
Rhodolirium andicolaInhibition of acetylcolinestarase[33]
P. albicansNot reported[35]
I. amancaes[43]
B. haemanthoides[44]
C. miniataInhibion of AChE and BuChE[76]
P. maritimumNot reported[78,79]
Pancratium illyricumInhibion of AChE and BuChE[93]
G. elwesii[99]
Narcissus poeticusNot reported[101]
N. tazetta subsp. tazettaInhibion of AChE and BuChE[81]
G. fosteriNot reported[106]
Leucojum aestivum[107,108]
Tazettine (3)PretazettineH. jamesonii, P. herbertiana and R. mendocinaNot reported[20]
Cytotoxic[26]
C. subedentataNot reported[27,28]
R. andicola[33]
P. albicans[35]
B. haemanthoides[44]
C. miniata[76]
P. maritimum[78,79]
L. martinezii[92]
Crinum graminicola[63]
Cyrtanthus mackenii
G. elwesii[99]
Vittatine (4)α-CrinaneH. jamesonii, P. herbertiana and R. mendocinaNot reported[20]
Cytotoxic and antibacterial[23]
P. albicansNot reported[35]
C. miniata[76]
P. illyricum[93]
Heamanthamine (5)H. jamesonii, P. herbertiana and R. mendocinaNot reported[20]
Induced apoptosis[22]
Antimalarial[23]
C. subedentataNot reported[27,28]
H. papilio[32]
B. haemanthoides[44]
R. andicola[33]
Scadoxus puniceusInhibition of AChE and BuAchE[62]
C. graminicola[63]
C. miniata[76]
P. maritimum[78,79]
G. elwesii[99]
Lycoramine (6)GalanthamineH. jamesonii, P. herbertiana, R. mendocinaNot reported[20]
R. andicolaInhibition of ACheE and BuChE[33]
I. amancaesNot reported[43]
Galanthine (7)LycoraneH. amesonii and P. herbertianaNot reported[21]
Analgesic and hypotensive[23]
N. serotinusNot reported[87]
G. elwesii[99]
G. fosteri[106]
11-Hydroxyvittatine (8)α-CrinaneH. amesonii and P. herbertianaNot reported[21]
Cytotoxicity and antibacterial[6]
Cyrtanthus mackeniiNot reported[63]
C. miniata[76]
P. maritimum[78,79]
L. martinezii[92]
G. elwesii[99]
G. fosteri[106]
Maritidine (9)α-CrinaneC. subedentataNot reported[27,28]
Homolycorine (10)Homolycorine
B. haemanthoides [44]
L. martinezii[92]
G. elwesii[99]
G. fosteri[106]
Hamayne (11)α-CrinaneC. subedentata[31]
A. coranica[56]
Ismine (12)IsmineC. subedentata[31]
B. haemanthoides[44]
L. martinezii[92]
G. elwesii[99]
G. fosteri[106]
Trisphaeridine (13)Narciclasine C. subedentata[31]
B. haemanthoides[44]
L. martinezii[92]
G. elwesii[99]
G. fosteri[106]
Narwedine (14)GalanthamineC. subedentata[31]
H. papilio[32]
G. elwesii[99]
Kirkine (15)LycorineC. subedentata[31]
L. martinezii[92]
Galanthindole (16)IsmineC. subedentata[31]
G. elwesii[99]
G. fosteri [106]
Anhydrolycorine (17)LycorineI. amancaes[43]
L. martinezii[92]
G. elwesii[99]
N. tazetta subsp. tazetta[81]
G. fosteri[106]
6α-Deoxytazettine (18)TazettineC. subedentata[31]
R. andicolaInhibition of acetylcolinestarase[33]
6-Methoxypretazettine (19)C. subedentataNot reported[31]
Dehydroanhydrolycorine (20)Lycorane
C. amabile, C. erubescens, C. moorei, A. belladonna and Z. carinata[34]
I. amancaes[43]
L. martinezii[92]
P. maritimum[99]
3-epi-Macronine (21)TazettineC. subedentata[31]
R. andicola[33]
Galanthus elwesii [99]
Hippapiline (22)HomolycorineH. papilio[32]
Papiline (23)
3-O-Demethyl-3-O-(3-hydroxybutanoyl)- haemanthamine (24)α-Crinane
11β-Hydroxygalanthamine (25) Galanthamine
Apogalanthamine (26),Miscellaneous
9-O-Demethyllycosinine B (27)Ismine
Galanthaminon (28)GalanthamineR. andicolaNot reported[33]
Norpluviine diacetate (29)LycoraneInhibition of acetylcolinestarase
Antibacterial
3-O-Acetyl-1,2-dihydrogalanthamine (30)GalanthamineNot reported
Undulatine diol (31)β-Crinane
Acetylnatalensine (32)α-Crinane
Undulatine (33)β-Crinane
B. haemanthoides[44]
A. belladonna [34]
Crinan-3-one (34)α-CrinaneR. andicola[33]
G. elwesii[99]
1-O-Acetyllycorine (35)LycorineC. amabile, C. erubescens, C. moorei, A. belladonna and Z. carinataNot reported[34]
N. sarniensis[43]
C. buphanoides[63]
Buphanisine (36)β-CrinineC. amabile and C. moorei[34]
B. haemanthoides[44]
B. disticha[61]
Chlidanthine (37)MiscellaneaPyrolirion albicansNot reported[35]
Crinine (38)β-Crinine
B. haemanthoides[44]
G. elwesii[99]
G. fosteri[106]
N-Demethyl galanthamine (39)GalanthamineP. albicansNot reported[35]
P. maritimum[99]
Montanine (40)MontanineP. albicans[35]
Anti-inflammatory and immunomodulatory[36]
Antioxidant and antimicrobial[37]
Anxiolytic, antidepressant, and anticonvulsant[38]
Antirheumatic[39]
B. haemanthoidesNot reported[44]
Haemanthus humilisAnticancer[69]
Pancracine (41)P. albicansNo reported[35]
Anti-inflammatory and immunomodulatory[36]
Antioxidant and antimicrobial[37]
Anxiolytic, antidepressant, and anticonvulsant[38]
Antirheumatic[39]
Sternbergine (42)LycoraneNot reported[35]
L. martinezii[92]
Hippeastrine (43)HomolycorineP. albicans[35]
L. martinezii[92]
Galanthus elwesii[99]
1-O-Acetylcaranine (44)LycoraneA. belladonna[40]
A. coranica[72]
Buphanamine (45)β-CrinineA. belladonna[40]
3-O-acetylhamayne (46)α-CrinineAntiprotozoal, cytotoxic““
7-Demethoxy-9-O-methylhostasine (47)Hippeastrum stapfianumInhibition of AChE[41]
Assoanine (48)LycorineI. amancaesNot reported[43]
Norlycoramine (49)Galanthamine
9-O-Demethylhomolycorine (50)HomolycorineP. maritimum[78,79]
L. martinezii[92]
G. elwesii[99]
N. tazetta subsp. tazetta[81]
Pancratinine C (51)MontanineP. maritimum[99]
AA from South Africa
Buphanidrine (52)B. haemanthoides[44]
B. disticha[61]
A. coronarica[73]
Ambelline (53)B. haemanthoides[44]
C. moorei[70]
C. latifolium[117]
Norbelladine (54)NorbelladineB. haemanthoides[44]
Augustine (55)
Distichamine (56)Cytotoxic
Distichaminol (57)Not reported
Crinamine (58)α-Crinine
B. grandiflora[63]
Haemanthidine (59)B. haemanthoides[44]
S. puniceusInhibition of acetylcholinesterase[62]
C. graminicolaNot reported[63]
Buphanamine (60)β-CrinineA. belladona[40]
B. haemanthoides[44]
B. disticha[61]
Crinsarnine (61) N. sarniensisInsecticidal[43]
Sarniensinol (62)MesembrineNot toxic
Bowdensine (63)β-Crinine
Sarniensine (64)Mesembrine
3-O-methylcrinamidine (65) β-CrinineB. distichaNot reported[61]
Acetyl-3-nerbowdine (66)
1-O-acetylbuphanamine (67)
2-O-Acetyllycorine (68)LycoraneC. buphanoides[63]
Criwelline (69)TazettineC. graminicola
Channaine (70)MscellaneaSceletium tortuosum[64]
Albomaculine B (71)HomolycorineH. humilisNo activity[69]
Coccinine (72)MontanineAnticancer
Incartine (73) LycoraneNo activity
N. serotinumNot reported[87]
G. elwesii[99]
G. fosteri[106]
Cherylline (74)CheryllineC. moorei[70]
Caranine (75)LycoraneA. coranica[71]
1-O-Acetyl-9-O-demethylpluviine (76)[72]
epi-Buphanisine (77)β-Crinine[81]
6α-Hydroxycrinamine (78)
6α-Hydroxybuphanidrine (79)[75]
Golceptine (80)Lycorane
Charisine (81)β-Crinine
Clivimine (82)ClivimineC. miniataNot reported[76]
Cliniatine C (83)HomolycorineInhibition of AChE/BuChE
Clivonine (84)
Nobilisitine B (85)
4′,O-Demethylbelladine (86)NorbelladineNot reported
3-O-Acetyl-8-O-demethylmaritidine (87)α-Crinine
8-O-Demethylmaritidine (88)
Clivia nobilis[98]
Galanthus elwesii[99]
Clivimine B (89)ClivimineClivia miniataNot reported[76]
AA fromMediterannean Basin
Ungeremine (90)LycoraneP. maritimumNot reported[71]
Antibacterial[80,82]
Antifungal[83]
Zefbetaine (91)Not reported[71]
Lycorenine (92)P. maritimum[78,79]
L. martinezii[92]
Pseudolycorine (93)LycoraneNarcisuss tazetta subsp. tazetta[81]
1-O-(3´-acetoxybutanoyl)lycorine (94)Narcissus serotinus[86]
Narseronine (95)Homolycorine
3-O-Methylnarcissidine (96) Lycorane[87]
1-O-Acetyl-3-O-methylnarcissidine (97)
1-O-Acetyl-3-O-methyl-6-oxonarcissidine (98)
11-Hydroxygalanthine (99)
N. tazetta subps. tazettaInhibition of AChEand BuAchE[81]
2-O-Methylclivonine (100)HomolycorineN. serotinusNot reported[87]
Masonine (101)Homolycorine
2-Oxomesembrenone (102)MesembraneNarcissus triandrus[88]
7,7a-Dehydromesembrenone (103)
Oxoepimesembrano (104)
Mesembrenone (105)
4-O-Demethylmesembrenone (106)
Mesembrine (107)
6-epi-Mesembrenol (108)
6-epi-Mesembranol (109)
Tyramine (110)AlkylamideL. martinezii[92]
5,6-Dihydrobicolorine (111)Miscellanea
G. elwesii [99]
P. maritimum[103]
G. fosteri[106]
Deoxylycorenine (112)HomolycorineL. martineziiNot reported[92]
6-O-Methyllycorenine (113)
1-O-Acetylnorpluviine (114)
6-Deoxypretazettine (115)
Lycosenine (116)Homolycorine
Norpluviine (117)Lycorane
Ungiminorine acetate (118)
Ungiminorine (119)
Narcissidine (120)
Narcissidine acetate (121)
11α-hydroxy-O-methylleucotamine (122)GalanthaminePancratium illyricumInhibition of acetylchlinestarase[93]
2-Hydroxyhomolycorine (123)HomolycorineNo activity
Sanguinine (124)GalanthamineInhibition of acetylchlinestarase
G. elwesiiNot reported[99]
Habranthine (125)P. illyricumInhibition of acetylchlinestarase[93]
Leucotamine (126)
O-Methylleucotamine (127)
G. elwesii[99]
Jonquailine (128)TazettineNarcissus jonquilla quailAnticancer[97]
Clivatine (129)HomolycorineC. nobilisNo activity[98]
Nobilisine (130)Antibiotic
Anhydrogalanthamine (131) GalanthamineG. elwesiiNot reported[99]
11,12-Dehydrolycorene (132)Lycorane
2,11-Didehydro-2-dehydroxylycorine (133)
6-O-Methoxylpretazettine (134)Tazettine
Galwesine (135)LycoraneG. elwesii[99]
G. fosteri[106]
Galasine (136)HomolycorineG. elwesii[99]
Oxoincartine (137)Lycorane
G. fosteri[106]
Hippamine (138)TazettineG. wlwesi[99]
S. lutea[100]
9-O-demethylpseudolycorine (139)LycoraneP. maritimum[99]
Galanthane (140) Miscellaneous
Pancratinine (141)Homolycorine
Obliquine (142)TazettineAntiviral[104]
O,N-Dimethylnorbelladine (143)NorbelladineG. fosteriNot reported[106]
9-O-Demethylmaritidine (144)α-Crinine
11-O-Acetyl-9-O-demethylmaritidine (145)
3,11-O-Diacetyl-9-O-demethylmarititidine (146)
Isocarbostyryls close to some Amaryllidaceae alkaloids
IsocarbostyrylSoubgroupAmar yllidaceaeBiological ActivityReference
Hippadine (147)LycoraneP. maritimumNot reported[71]
N. sarniensisInsecticidal[43]
C. jagusCytotoxic[109]
Flexinine (148)β-CrinineCrinum jagusInhibition of AChE[109]
Gigantelline (149)Cherylline
Gigantellinine (150)
Gigancrinine (151)β-Crinine
Narciclasine (152)LycorineN. peudonarcissusAnticancer[6,110,111,112,113]
S. lutea[114]
Pncratistatin (153)Hymenocallis littoralis[110,115]
2-Methoxypratosine (154)N. serotinus[87]
Pratorimine (155)C. buphanoidesNot reported[63]
Pratosine (156)C. latifolium[116]
Pratorinine (157)
“ means the same content.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Evidente, A. Advances on the Amaryllidacea Alkaloids Collected in South Africa, Andean South America and the Mediterranean Basin. Molecules 2023, 28, 4055. https://doi.org/10.3390/molecules28104055

AMA Style

Evidente A. Advances on the Amaryllidacea Alkaloids Collected in South Africa, Andean South America and the Mediterranean Basin. Molecules. 2023; 28(10):4055. https://doi.org/10.3390/molecules28104055

Chicago/Turabian Style

Evidente, Antonio. 2023. "Advances on the Amaryllidacea Alkaloids Collected in South Africa, Andean South America and the Mediterranean Basin" Molecules 28, no. 10: 4055. https://doi.org/10.3390/molecules28104055

Article Metrics

Back to TopTop