Next Article in Journal
Putative Biosynthesis of Talarodioxadione & Talarooxime from Talaromyces stipitatus
Previous Article in Journal
Determination of Osimertinib, Aumolertinib, and Furmonertinib in Human Plasma for Therapeutic Drug Monitoring by UPLC-MS/MS
Previous Article in Special Issue
New C15 Acetogenins from Two Species of Laurencia from the Aegean Sea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Phytochemistry and Pharmacology of Tulbaghia, Allium, Crinum and Cyrtanthus: ‘Talented’ Taxa from the Amaryllidaceae

by
Cynthia Amaning Danquah
1,*,
Prince Amankwah Baffour Minkah
1,2,
Theresa A. Agana
3,
Phanankosi Moyo
4,
Michael Ofori
1,5,
Peace Doe
6,
Sibusiso Rali
4,
Isaiah Osei Duah Junior
7,
Kofi Bonsu Amankwah
8,
Samuel Owusu Somuah
9,
Isaac Newton Nugbemado
1,
Vinesh J. Maharaj
4,
Sanjib Bhakta
10 and
Simon Gibbons
11
1
Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
2
Global Health and Infectious Disease Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
3
Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
4
Department of Chemistry, University of Pretoria, Pretoria 0028, South Africa
5
Department of Pharmaceutical Sciences, Dr. Hilla Limann Technical University, Wa P.O. Box 553, Ghana
6
Department of Pharmaceutical Sciences, School of Pharmacy, Central University, Accra, Ghana
7
Department of Optometry and Visual Science, College of Science, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
8
Department of Biomedical Sciences, University of Cape Coast, Cape Coast, Ghana
9
Department of Pharmacy Practice, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
10
Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, Malet Street, London WC1E 7HX, UK
11
The Centre for Natural Products Discovery (CNPD), Liverpool John Moores University, Liverpool L3 3AF, UK
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(14), 4475; https://doi.org/10.3390/molecules27144475
Submission received: 16 May 2022 / Revised: 24 June 2022 / Accepted: 28 June 2022 / Published: 13 July 2022

Abstract

:
Amaryllidaceae is a significant source of bioactive phytochemicals with a strong propensity to develop new drugs. The genera Allium, Tulbaghia, Cyrtanthus and Crinum biosynthesize novel alkaloids and other phytochemicals with traditional and pharmacological uses. Amaryllidaceae biomolecules exhibit multiple pharmacological activities such as antioxidant, antimicrobial, and immunomodulatory effects. Traditionally, natural products from Amaryllidaceae are utilized to treat non-communicable and infectious human diseases. Galanthamine, a drug from this family, is clinically relevant in treating the neurocognitive disorder, Alzheimer’s disease, which underscores the importance of the Amaryllidaceae alkaloids. Although Amaryllidaceae provide a plethora of biologically active compounds, there is tardiness in their development into clinically pliable medicines. Other genera, including Cyrtanthus and Tulbaghia, have received little attention as potential sources of promising drug candidates. Given the reciprocal relationship of the increasing burden of human diseases and limited availability of medicinal therapies, more rapid drug discovery and development are desirable. To expedite clinically relevant drug development, we present here evidence on bioactive compounds from the genera Allium, Tulgbaghia, Cyrtanthus and Crinum and describe their traditional and pharmacological applications.

1. Introduction

Amaryllidaceae belongs to the order Asparagales and consists of bulbous flowering plants separated into three infrageneric ranks: Agapanthoideae, Allioideae and Amaryllidoideae, as delineated by the Angiosperm Phylogeny Group [1]. The term “Amaryllidaceae” is frequently used in either phytochemical or pharmacological literature to refer to plants or alkaloids originating from the subfamily Amaryllidoideae [2,3]. Monocotyledonous plants constitute seventy-nine genera (including Allium, Crinum, Cyrtanthus, and Tulbaghia) with over 1000 species [4]. Aside from their broad pantropical distribution, Amaryllidaceae are located in Africa, the Mediterranean Coast and South America, and have high adaptation and speciation [5]. The genus Allium is distributed in temperate, arid, semi-arid and subtropical areas such as the Mediterranean region, central Asia, Africa and parts of Europe. As herbaceous geophyte perennials, Allium comprises a plethora of species with pungent linear leaves that may or may not arise from a bulb or rhizome [6,7]. The Tulbaghia genus, popularly called “sweet garlic”, “wild garlic”, or “pink agapanthus”, is crown shaped with outgrowth or appendages of the perianth and predominantly colonizes the Eastern cape belt of South Africa, and is adapted for growth in areas such as Europe and America [8,9]. The genus Crinum encompasses 104 species and appear as showy flowers on leafless stems, which thrive in the tropics and warm temperate parts, specifically Asia, Africa, America, and Australia [10]. Cyrtanthus is popularly known as “fire lily” due to its unique rapidly flowering response to natural bush fires. Most species are found in South Africa and play an important role in South African traditional medicine [11].
Amaryllidaceous plants are known for their ornamental, nutritional, and medicinal value. Given their attractive flowering plant-like features, Crinum species are prized for their umbel lily-like blossoms in China and Japan [3]. Concurrently, Amaryllidaceae are known for their longstanding exploitation in medicinal therapy owing to their inherent biosynthesis of chemically diverse bioactive compounds with peculiar biological properties. The use of proximate and mineral composition analysis enabled the identification of phytoconstituents [10,12], while in vitro, in vivo, and in silico model systems have permitted the unravelling of intrinsic pharmacological activities of the natural products and other alkaloids isolated from this source [13,14,15]. Of note, bioactive compounds from Amaryllidaceae possesses a wide range of bioactivities ranging from antioxidant [16,17], anti-inflammatory [16,18], antimicrobial [17], antifungal [19], antiviral [20,21], antiplasmodial [22,23,24], anticarcinogenic [18,25,26], antispasmodic [1,27], antiplatelet [28], antiasthmatic [29], antithrombotic [30,31], antitumor [25], antihyperlipidemic [25], antihyperglycemic [25,32,33], antiarthritic [25], antimutagenic [16], immunomodulatory [16] and several others [34].
Given the aforementioned biological activities, Allium, Tulbaghia, Cyrtanthus and Crinum are utilized in traditional medicinal therapy for varying diseases and conditions [35,36,37,38,39,40,41]. For example, Allium is used as concoctions, decoctions, extracts, and herbal preparations to treat angina, amoebic dysentery, arthritis, cardiovascular diseases, cholera, catarrh, dysmenorrhea, fever, headaches, hepatitis, stomach disorders, throat infections, and prostatic hypertrophy [30,31,35,36,37,38]. The genus Tulbaghia has unique pharmacotherapeutic properties and is utilized to manage ailments such as earache, pyrexia, tuberculosis, and rheumatism [9,42]. Crinum species are used to treat haemorrhoids, malaria, osteoarthritis, varicosities, wounds, urinary tract infections, and gynaecological remedies [40,41]. Cyrtanthus are also employed in the management of ailments associated with pregnancy, as well as cystitis, age-related dementia, leprosy, scrofula, headaches, chronic coughs, among others [43,44]. In modern clinical practice, galanthamine from Amaryllidaceae is a primary choice of drug in managing symptomatic neurological disorders such as Alzheimer’s disease due to its selective inhibitory action on the acetylcholine biosynthetic enzyme, acetylcholinesterase [45]. The pancratistatin phenanthridone class of alkaloids are also promising chemotherapeutic drug candidates with unique cell line-specific antiproliferative properties, conferring a selective advantage for clinical development [46].
Although Amaryllidaceae represents a source of valuable bioactive compounds, developing promising drug candidates into clinically relevant therapeutics has been slow. Similarly, other genera in this family, including Cyrtanthus, Crinum and Tulbaghia, are untapped reservoirs and could serve as an alternative window for novel drug targets and warrant further investigation. This review consolidates evidence on the bioactive compounds from Allium, Tulbaghia, Crinum and Cyrtanthus and ascertains their traditional and pharmacological applications. Specifically, bibliographic searches were conducted on multiple standard databases (such as, Scopus, Web of Science, MEDLINE, Sci verse, Embase, Google scholar among others) using MESH and non-MESH terms to retrieve and synthesize relevant publications over the 3-month search period. This review highlights panoply of promising biomolecules from the taxa Amaryllidaceae and their prominent medicinal values. The evidence from this study could hasten drug discovery among the pharmaceutical industries. An update on the natural products from these lesser explored genera could also augment the lean pipeline of novel therapeutics.

2. The Genus Tulbaghia

2.1. Botanical Description

Tulbaghia is made up of monocotyledonous species with herbaceous perennial bulbs covered by brown scales and are mostly found in Africa [8]. South African species possess bulb-like corms or rhizomes which are swollen, irregularly shaped and wrapped in dry, fibrous leaves [8]. Members of this genus usually possess a raised crown-like structure or ring at the center of their flower tube [8]. Their seeds are black, flat and elongated with the mature ones having embryos [8]. Examples of species of this genus are Tulbaghia violacea (T. violacea), Tulbaghia acutiloba Harv. (T. acutiloba), Tulbaghia capensis L. (T. capensis) and Tulbaghia cepacea L.f (T. cepacea) [8].

2.2. Geographical Distribution and Traditional Uses of Tulbaghia Species

With approximately 66 species (https://www.kew.org/science accessed on 22 February 2022) [47], Tulbaghia is the second-most species-rich genus within Amaryllidaceae. Tulbaghia is a monocotyledonous genus comprised morphologically of herbaceous perennial bulbous species, which produce a variety of volatile sulfur compounds, hence resulting in a distinct pungent garlic odor released by bruised plants [8,48]. The genus was named by Carl Linnaeus after Ryk Tulbagh (1699–1771), a former governor of the Cape of Good Hope in South Africa, where most of the native species are to be found, particularly in the Eastern Cape Province [49]. In addition to South Africa, the genus is widely distributed across southern African countries including Botswana, Lesotho, Swaziland, and Zimbabwe, where the plant is revered in folk medicine being used for the treatment of a plethora of infectious and non-infectious diseases [9] as highlighted in Table 1.

2.3. Phytochemistry of Tulbaghia

Tulbaghia produces many different classes of compounds with diverse chemical structures dominated by sulfur-containing natural products (Figure 1; Table S1).
Most compounds reported have a small molecular weight (<500) and are of a broad lipophilicity (Figure 2).
Tulbaghia violacea has been the most widely investigated for its phytochemistry and pharmacological properties. To date, close to 100 compounds have been tentatively identified, largely using gas chromatography techniques, from different parts of this species (Supplementary File S1) [55]. Most prominent are the sulfur compounds with reported broad-spectrum pharmacological activity. The thiosulfinate marasmicin (1) is the most prolific antimicrobial compound reported thus far from this genus [56]. This compound is formed from its precursor compound marasmin (2), by the enzyme c-lyase. Marasmicin is responsible for the characteristic garlic odor generated by damaged plants [48]. Other notable compounds produced by this species include phenols, tannins and flavonoids [55], which are also responsible for several observed biological activities. Phytochemical characterization has been carried out, albeit minimally for other Tulbaghia species particularly T. alliacea and T. acutiloba. Unlike other genera in Amaryllidaceae, Tulbaghia is so far devoid of any alkaloids [57,58]. Despite the extensive in vitro pharmacological screening of extracts of Tulbaghia, it is possible that less effort has been made to isolate and identify their active principles. Hence, the phytochemistry of the genus Tulbaghia largely remains understudied. The chemicals structurers of noteworthy compounds isolated from T. violacea have been represented in Figure 3.

2.4. Pharmacological Studies of Tulbaghia Species

Because of its perceived medicinal value, Tulbaghia has received marked interest within the scientific community which has meticulously subjected it to various in vitro and in vivo studies experimentally evaluating its pharmacological activities. The volume of published studies generated from these investigations mirror the distribution of the genus with most articles on Tulbaghia having emerged from South Africa (Table 2), a country highly rich in this genus both in terms of species diversity and abundance.
The greatest numbers of pharmacological screens have been on interrogating the antimicrobial properties of this genus. This is closely followed by cardiovascular, antioxidants and cancer investigations as shown in Table 3. T. violacea prominently features, being the most studied species, with T. alliacea and T. aticulata having received minimal attention.

2.4.1. Antimicrobial and Antiparasitic Activity

As antimicrobial resistance continues to be a global health threat, the need to find therapeutic alternatives has never been more urgent [63]. This has encouraged scientists to search for novel alternatives with natural products having drawn marked interest as a potential oasis of new antimicrobial agents [64,65,66]. Tulbaghia has received significant relevance in this regard, with multiple studies providing ample evidence substantiating its use as an antimicrobial agent. Extracts of T. violacea have potency against many microbial species including those designated as priority by the World Health Organization. These include Pseudomonas aeruginosa (P. aeruginosa), Staphylococcus aureus (S. aureus) and Klebsiella pneumoniae (K. pneumoniae) with MIC values ranging between 20 and 300 µg/mL [67]. This activity was confirmed in another study where the disc diffusion method was used [68]. In addition to bacteriostatic activity, extracts of T. violacea have shown noteworthy potency against yeasts including Candida albicans (C. albicans) and Candida parapsilosis (C. parapsilosis) with MIC and MMC values ranging between 20 and 40 µg/mL [68]. Beyond human pathogens, extracts of T. violacea have activity against microorganisms of agricultural significance, for example against the fungus Aspergillus flavus (A. flavus), which is responsible for significant agricultural produce loss at a global scale due to production of aflatoxins [69]. Extracts of T. violacea compromised cell wall synthesis by significantly reducing β-glucan and chitin synthesis in A. flavus corresponding to a dose-dependent inhibition of the enzymes β-glucan and chitin synthase, respectively [70]. Further studies suggested an alternative mode of action (MoA) via reduction of ergosterol production in fungi [71]. Interestingly, related to value in agriculture, a patent has been filed on the use of extracts of T. violacea as a plant protecting remedy as a substitute for chemical agents [72]. Some thought-provoking studies have shown that growth conditions including light intensities, watering frequency and pH, substantially impact both growth and biological potency of T. violacea extracts against Fusarium oxysporum (F. oxysporum) [73,74]. Likewise, storage conditions of dried plant material also affect the antimicrobial potency of extracts [56]. In addition to antimicrobial activity, T. violacea has shown good antiparasitic activity against the parasitic worm Meloidogyne incognita (M. incognita) on tomato roots and in soil [75]. Antiparasitic activity has also been observed against Trypanosoma brucei (T. brucei) (IC50 = 2.83 µg/mL) and Leishmania tarentolae (L. tarentolae) (IC50 = 6.29 µg/mL) [67]. Table 4 highlights the antimicrobial activity of Tulbaghia species.

2.4.2. Anticancer Activity

Owing to the need for novel anticancer agents [76] and motivated by the success of cancer drug discovery projects from natural products [77], Mthembu and Motadi in (2014), evaluated the in vitro anticancer properties of crude methanol extracts of T. violacea using an MTT assay [78]. Extracts displayed time- and concentration-dependent antiproliferative properties against cervical cancer cell lines with an IC50 of 150 µg/mL. The MoA was deciphered to be induction of apoptosis by a p53-independent pathway [78]. However, in contrast to this finding, continued work showed a proportional increase in the activity of caspase 3/7, and the expression of p53 genes strongly suggests apoptosis was triggered by a p53-dependent pathway [79]. This latter finding has been partly substantiated by data emerging from a study examining the antineoplastic properties of T. violacea against ovarian tumor cells. These extracts were shown to partially induce both apoptosis and necrosis with the most pronounced activity due to induction of autophagy [80].
Triple-negative breast cancer remains one of the most challenging cancers, being highly aggressive [81]. T. violacea extracts have demonstrated good cytotoxic activity against MDA-MB-231, with an IC50 of 300 µg/mL [82]. Additionally, extracts inhibited migration of the cancer cell lines (metastasis), an important physiological process in the progression of this cancer [83]. In addition to the gynecological cancers, antineoplastic properties of T. violacea were further observed against pancreatic cancer with 63% inhibition of cell proliferation at a concentration of 250 µg/mL [68]. Against a non-sex-specific cancer, T. violacea showed noticeable activity against oral cancer with an IC50 of 0.2 and 1 mg/mL for acetone and water-soluble extracts, respectively. Extracts activated caspase activity in a dose-dependent manner leading to induction of apoptosis in the human oral cancer cell line [84]. Using a bioassay guided approach, the active anticancer compounds in T. violacea have been identified to be glucopyranosides d-fructofuranosyl-β (2→6)-methyl-α-d-glucopyranoside and β-d-fructofuranosyl-(2→6)-α-d-glucopyranoside. Both compounds act by mediating induction of apoptosis in Chinese hamster cells by targeting the mitochondrial (intrinsic) pathway [85,86]. A summary of the anticancer activity of Tulbaghia species is shown in Table 5.

2.4.3. Antioxidant Activity

The imbalance of reactive oxygen species (ROS) and antioxidants in the body can lead to oxidative stress [87]. This physiological condition can result in cellular and tissue damage [88]. Oxidative stress is associated with pathologies including cancer, cardiovascular disease, diabetes, and neurodegenerative diseases amongst others [88,89]. To avert the development of oxidative stress, attenuation of ROS has been identified as a viable target, with natural products seen as a potential source capable of neutralizing it [88]. Tulbaghia has generated some interest on this front particularly as it is rich in compounds with proven antioxidant activity including phenols, tannins and flavonoids. Multiple studies have demonstrated that extracts of Tulbaghia have marked antioxidant activity as assessed using different assays in vitro including Trolox equivalent antioxidant capacity (TEAC; also commonly referred to as the ABTS assay), ferric-reducing antioxidant power (FRAP) and 2,2-diphenyl-1-picryl-hydrazyl-hydrate (DPPH) (Table 6) [58,80,90,91]. Furthermore, using an in vivo model of Caenorhabditis elegans, T. violacea extracts attenuated oxidative stress produced by a free radical generator, (2,2′-azobis-2-amidinopropane dihydrochloride; AAPH), in the roundworm [80]. Data from these studies strongly suggested continued investigation of other species in the search for more potent antioxidant agents from Tulbaghia. The antioxidant activity of Tulbaghia species is highlighted in Table 6.

2.4.4. Antidiabetic, Anticardiovascular and Antithrombogenic Activity

The incidence of diabetes and cardiovascular diseases continues to grow substantially across the globe, with both conditions combined accounting for the highest global morbidity and mortality [93,94]. Both of these chronic conditions are closely linked with cardiovascular disease being responsible for high morbidity and mortality in diabetic patients [95]. Tulbaghia has been documented in ethnopharmacological studies for the treatment of these ailments with emerging scientific data strongly validating its use. In streptozotocin diabetes-induced rat models, T. violacea attenuated diabetes-associated physiological conditions resulting in improved body weights, reduced fasting blood glucose levels, enhanced glucose tolerance and significantly elevated plasma insulin and liver glycogen content [96]. These data were corroborated in another study in which T. violacea noticeably reduced blood glucose and serum lipid (triglyceride (TG), total cholesterol (TC), and very low-density lipoprotein (VLDL)) levels while raising plasma insulin in a streptozotocin-induced diabetic rat model [97]. In an assessment for negating cardiovascular associated conditions, T. violacea in in vivo models markedly reduced systolic blood pressure (BP), diastolic BP, mean arterial pressure (MAP) and the heart rate in both age-induced and spontaneous hypertensive rats [98]. Furthermore, dosing rats with extracts of T. violacea led to improved kidney function [99]. This is an essential pharmacological property as kidney function is impaired in hypertension leading to high morbidity and mortality in people suffering from cardiovascular diseases [100].
One of the multiple factors strongly associated with cardiovascular disease is atherothrombotic vascular disease (AVD). Platelet aggregation plays a role in development of AVD and subsequent cardiovascular events [90,101]. Against this background, platelet aggregation has been identified as a key process to target to prevent AVD. Encouragingly, T. violacea demonstrated marked potency being able to significantly inhibit platelet adhesion 15 min post-exposure (Table 7) [90,92].

2.4.5. Miscellaneous Pharmacological Activity

In addition to diabetes and cardiovascular diseases, T. violacea has shown activity against another chronic condition, Alzheimer’s disease. In an in vivo Alzheimer’s disease transgenic C. elegans strain model, T. violacea significantly reduced 1-42 β-amyloid peptide formation (Table 8) [80]. T. violacea exhibited in vivo anticonvulsant activity by attenuating tonic convulsions induced by either pentylenetetrazole, bicuculline, picrotoxin, strychnine or NMDLA [102] and validating its traditional use for the treatment of epilepsy. T. violacea displayed marked tick repellence properties of fungus-exposed plants at low treatment concentrations (5% w/v and 10% w/v) [59], further enhancing its credentials as a potential agricultural product. Somewhat concerning is that, extracts of T. violacea also induced genotoxic effects albeit at high test concentrations (250, 500 and 1000 µg/mL) in the Allium cepa assay [103]. Furthermore, broad murine macrophage antiproliferative and cytotoxicity activity, influenced by extract test concentrations, type of solvent and plant part used, have been observed (Table 8) [104]. There is consequently a need for rigorous assessment of safety of extracts of this and other species of the genus Tulbaghia.

3. The Genus Allium

3.1. Botanical Description

Species of the genus Allium are mostly found in warm–temperate and temperate zones of northern hemisphere as well as the boreal zone [105]. They are petaloid perennial herbs with parallel narrow leaves [33] and possess true bulbs, which are sometimes found on rhizomes [106]. Allium species are also characterized by onion or garlic odor and flavor similar to Tulbaghia [106]. Well known species include Allium cepa (A. cepa), Allium sativum (A. sativum), Allium ascalonicum (A. ascalonicum,), Allium porrum (A. porrum), and Allium schoenoprasum (A. schoenoprasum) (chive) [33]
Allium has over 500 species making it the largest genus of Amaryllidaceae [6,7]. There are plethora of species, notably A. cepa and A. sativum [107,108,109]. Other examples grown for their medicinal and nutraceutical value are Allium ducissae (A. ducissae), Allium strictum (A. strictum), Allium umbilicatum (A. umbilicatum), Allium victorialis (A. victorialis), A. ascalonicum, Allium chinense (A. chinense), Allium tuberosum (A. tuberosum), Allium griffithianum (A. griffithianum), Allium oreoprasum (A. oreoprasum), and Allium oschaninii (A. oschaninii). Species tolerate varying climatic conditions, hence are geographically distributed across several continents, including Asia, Africa, the Americas, and Europe [107,110]. Fernandes et al. identified A. cepa that colonizes four different geographical regions of the Madeira island, an archipelago near the North Atlantic ocean with a hot and/or warm-summer Mediterranean climate conditions [107]. As the world’s second-most relevant and cultivated horticulture vegetable crop, the onion (A. cepa), is distributed in over 175 countries and covers approximately six million hectares of the total land size of the world. Approximately two-thirds (66%) of global onion production emanates from the Asia, with China and India being the world’s largest producers [111]. The maximal diversification of A. cepa is found in Iran and Afghanistan’s Mediterranean basin. A. cepa thrives in areas with boreal, temperate, and tropical climates [108]. Similarly, A. sativum (garlic) bears close resemblance to onions and originates from Central Asia but has spread to include regions in Europe, America, and Africa [112]. The global garlic production estimates show that out of the 28.5 million tonnes (MT) of A. sativum cultivated, the majority (91.6%; 26.1 MT) were from Asia, followed by Europe (3.0%; 0.86 MT), America (2.9%; 0.83 MT), and with the least from Africa (2.7%; 0.73 MT) [112]. Bartolucci et al. identified A. ducissae, a new breed of Allium that grows in the mountainous regions of the Central Apennines in the Abruzzo and Lazio counties of Italy [113]. Furthermore, A. strictum, a Eurasian species, is distributed across China, Europe, Russia, Kazakhstan, Kyrgyzstan, and Mongolia [114,115]. A. umbilicatum, also called gladiolus or leek is usually localized in semi-arid regions and can tolerate sub-zero freezing winters [116]. It occurs as a weed in oases and span across Afghanistan, Iran, Pakistan, Turkmenistan, Tajikistan, and central and Eastern Asian regions [116]. As a representative circumboreal plant, A. victorialis has a wide altitudinal climatic tolerance [117]. It is predominantly located in lowland deciduous forest and subalpine birch forest, but seldom found in the subalpine meadows [117]. This species is scattered distribution on the island stretches of Japan, Russia, and Northern China [117,118]. Although practically grown throughout the world, A. ascalonicum, also called shallot, is native to the Middle East, and the name is derived from the Syrian city Ascalon. These shallots are distributed on the main islands of Indonesia, in Bangladesh, Japan, Korea, Malaysia, Taiwan, and Thailand [119]. A. chinense (locally referred to as Chinese/Japan onion or scallion, Kiangski scallion, oriental onion, Rakkyo) is an uncommon Allium species found mainly in the tropical and sub-tropical regions of China, Japan, Vietnam, and eastern areas of India [111,120]. A. tuberosum is an indigenous species native to southeastern Asia and regarded as a late-seasonal bloomer. During the initial growth phases, A. tuberosum is evergreen in hot climates but succumbs to cold climatic conditions. However, the Chinese chive becomes tolerant to all seasonal variations [121,122]. A. griffithianum and A. oreoprasum are geographically skewed towards the mountainous regions of Pakistan, Afghanistan, Kyrgyzstan, Uzbekistan, and Tajikistan [123], whereas A. oschaninii are located in the Darvaz mountains of Central Tajikistan [124].

3.2. Traditional Uses of Genus Allium

Increasing scientific evidence asserts the traditional uses of plants in folklore medicine [124,125,126]. Researchers over the years have investigated various parts of local medicinal plants to identify phytoconstituents with potential bioactivity, and further develop them into new drug therapies [127,128]. Allium species contain the common phytocompounds (anthocyanins, flavonoids, organosulfur, sterols, saponins, phenolic acids, amino acids, vitamins and minerals) [129,130,131,132] with innumerable biological properties [130,131,132,133]. Owing to these biological advantages, Allium species are locally used in managing various diseases affecting human organs and organ systems such as inflammation, microbial pathologies and oxidative stress injuries [130,131,132,133]. In particular, A. cepa is used to treat alopecia, hearing impairment, menstrual disorders, erectile dysfunction and ocular and metabolic diseases [133,134,135]. Similarly, A. sativum is employed in the management of hematological disorders, carcinomas, muscle weakness and compromised airways [135,136,137,138,139]. Other varieties of Allium species also serve as appetizers, nerve soothers, and relieving agents against digestive, respiratory, and urinary system discomfort as seen in Table 9.

3.3. Phytochemistry of Allium

Owing to the numerous traditional uses of these species, it is not surprising that the genus contains several phytoconstituents which may be responsible for their observed activity. Table 10 outlines various phytochemicals isolated, their geographic location and their biological activity.
The chemical structures of compounds from the genus Allium are shown in Figure 4.

3.4. Pharmacological Effects of Allium

There are several species within Allium whose biological activities have been well established [198]. This section focuses on the pharmacological activities associated with these species.

3.4.1. Antimicrobial Activities

Garlic has shown antimicrobial effects against Gram-positive, Gram-negative and acid fast stain organisms [199,200,201]. Allicin from garlic showed effectiveness toward methicillin-resistant S. aureus (MRSA) [200]. Extracts from garlic also showed broad-spectrum fungicidal effect against several fungi including Candida, Trichophyton, Cryptococcus, Aspergillus, Trichosporon and Rhodotorula species. Garlic extract was recently found to inhibit Meyerozyma guilliermondii and Rhodotorula mucilaginosa germination and growth [202]. A study by Fufa reported the antifungal activity of various A. sativum extracts, namely aqueous, ethanol, methanol, and petroleum ether against human pathogenic fungi such are Trichophyton verrucosum, T. mentagrophytes, T. rubrum, Botrytis cinereal (B. cinerea), Candida species, Epidermophyton floccosum, A. niger, A. flavus, Rhizopus stolonifera, Microsporum gypseum, M. audouinii, Alternaria alternate, Neofabraea alba, and Penicillium expansum [203]. Essential oil from garlic showed antifungal activity against a number of fungi such as (C. albicans, C. tropicalis and Blastoschizomyces capitatus). Saponins extracted from A. sativum had antifungal activity against B. cinerea and Trichoderma harzianum [204]. Allium species from Ghana were reported by Danquah et al. to possess anti-infective and resistance modulatory effects on selected microbial strains [205]. Allium hirtifolium was found to exhibit antimicrobial activities against E. faecalis [206].
Previous studies have shown that garlic extract inhibit the growth of Blastocystis species in vivo and this effect was attributed to the several phytochemicals contained in garlic extracts. Examples of these phytochemicals are thiosulfinates and allicin which have been investigated to possess antibacterial and antiprotozoal effects [204,207]. Garlic extracts have been evaluated for antiviral effects against influenza B, human rhinovirus type 2, human cytomegalovirus (HCMV), parainfluenza virus type 3, Herpes simplex type 1 and -2, vaccinia virus, and vesicular stomatitis virus [208]. Danquah et al. again reported the antitubercular effects of analogues of disulfides from A. stipitatum as well as their anti-biofilm and anti-efflux effects [209].

3.4.2. Antioxidant Properties

It has been reported that frequent garlic intake promotes internal antioxidant activities and reduces oxidative adverse effects either by increasing the endogenous antioxidant synthesis or reducing the production of oxidizing agents such as oxygen-free radical species (ORS) [210]. It has also been demonstrated that garlic possesses protective properties against gentamycin as well as acetaminophen-induced hepatotoxicity by improving antioxidant status, and regulating oxidative stress [200]. Garlic extract was found to elevate the activities of selected antioxidant enzymes (e.g., superoxide dismutase (SOD)) and decrease glutathione peroxidase (GSH-Px) in rats’ hepatic tissues [13,118,211]. Saponins extracted from garlic were reported to scavenge intracellular ROS and protect mouse-derived C2C12 myoblasts towards growth inhibition and H2O2-induced DNA damage [13,212]. A. ursinum aqueous extract also demonstrated antioxidant effect which lasted approximately 16 h [213]. A. hirtifolium was reported to possess antioxidant capacity by neutralizing the free radical species in a system [214].

3.4.3. Anti-Inflammatory Properties

It has been reported widely that garlic extracts and its related phytochemicals possess anti-inflammatory activity. A study by Ahmad et al. revealed that garlic extracts significantly impaired liver inflammation and damage caused by Eimeria papillata infections [215]. The mechanism underlying the anti-inflammatory effects of garlic was attributed to the inhibition of emigration of neutrophilic granulocytes into epithelia as described by Hobauer et al. [216] and Gu et al. [217]. The chloroform extract of aged black garlic acts by reducing NF-κB activation in human umbilical vein endothelial cells caused by tumor necrosis factor-α (TNF-α) and the methanolic extract also reported to prevent the cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) production by NF-κB inactivation [218]. A report by Jin et al. confirmed that thiacremonone (a sulfur compound isolated from garlic) prevents neuroinflammation and amyloidogenesis by blocking the NF-κB activity, and therefore makes it an ideal remedy to manage neurodegenerative disorders (e.g., Alzheimer’s disease) related to inflammation [219].
Krejčová et al. reported that pyrithione and related sulfur-containing pyridine N-oxides from Persian shallot possessed anti-inflammatory and neurological activity [220]. The extracts of A. stipitatum were reported to exhibit antibacterial effect in vivo against methicillin-resistant S. aureus [221]. Anti-inflammatory effect of A. hookeri on carrageenan-induced air pouch mouse model was also established by Kim et al. [222].

3.4.4. Anticancer Activity

Comparison of the anticancer effect of raw garlic extracts against other extracts from different plants found garlic to be the most effective and highly specific anticancer agent [223]. The anticancer mechanisms of garlic extracts were reported to be mediated via inhibition of cell growth and proliferation, regulation of carcinogen metabolism, stimulation of apoptosis, prevention of angiogenesis, invasion, and migration; and thus affording the anticancer agent with minimal negative effects [13]. Chabria et al. reported that allicin isolated from garlic suppresses colorectal cancer metastasis through enhancing immune function and preventing the formation of tumor vessels as well as surviving gene expression to enhance the cancer cell’s apoptosis [224]. Fleischauer and Arab [225] reported that continuous garlic intake could decrease different kinds of cancer propagation such as cancer of the lung, colon, stomach, breast, and prostate. Piscitelli et al. reported that garlic reduced the plasma concentrations of saquinavir by approximately 50% in healthy participants after a 3-week garlic supplement intake. In addition to this, many researchers evaluated the antitumor and cytotoxic actions of garlic and its related constituents in vitro and in vivo [226].

3.4.5. Other Pharmacological Effects of Allium Species

Investigations on extracts of A. sativum (garlic) revealed anticholinesterase effects, which could be further developed and utilized in the management of Alzheimer’s disease [227,228,229]. Garlic is known to possess hypolipidemic effects by reducing the total glycosaminoglycans concentration in heart and aorta [230]. Garlic is also known to reduce the level of cholesterol either by acid stimulation and excretion of neutral steroids or by reducing the cholesterogenic and lipogenic effects of fatty acid synthase, 3-hydroxy-3-methyl-glutaryl-CoA reductase, malic acid, and glucose-6 phosphate dehydrogenase in hepatocytes [231]. Garlic tablets formulated by Ashraf et al. and administered at a dose of 600 mg/day for 12 weeks in diabetic patients with dyslipidemia resulted in high HDL, low LDL and TC levels [232].
Allicin, a constituent in garlic, was found to reduce diabetes mellitus in rats, which was similar to that demonstrated by glibenclamide and insulin [233]. Garlic extracts reduce body weight, adipose tissue mass and improved plasma lipid profiles in mice with high-fat diet-induced obesity [234]. The mechanism of these activities is downregulation of multiple gene expression such as adipogenesis along with upregulation of the mitochondrial inner membrane proteins expression [234]. Garlic extract is widely known to significantly control blood pressure by reducing both systolic and diastolic pressures [235]. Moreover, several reports have confirmed the antihypertensive effects of garlic [236]. Extracts of A. stipitatum were also assessed and established to possess significant wound healing properties [237].

4. The Genus Crinum

4.1. Geographical Distribution of Crinum

Crinum, which also belongs to the Amaryllidaceae family, comprises approximately 160 beautiful lilies that grow naturally in coastal areas of the tropics and subtropics. They are widely distributed in Africa, Asia, Australia and America [238,239,240,241]

4.2. Traditional Uses of Crinum

Plants of the genus Crinum have been used to treat various diseases across the world [242]. In China and Vietnam, Crinum plants in traditional medicine are believed to possess antiviral and immune-stimulatory properties. A hot aqueous extract of Crinum latifolium (C. latifolium) is used as an antitumor agent. Crinum asiaticum (C. asiaticum) is used in Malaysia to treat rheumatism and to relieve local pain [239]. Crinum amabile Donn. (C. amabile) is used in Vietnam to induce emesis, as well as for rheumatism and earache [241].
The bulbs of C. asiaticum L. are used as a tonic, laxative and expectorant in Indian traditional medicine, as well as for treating urinary tract diseases [241]. The seeds are used as purgatives, diuretics, and tonics, while the raw roots are used as an emetic. The leaves are also very useful in the management of skin problems, inflammation and cough [241]. C. latifolium L. is also used to treat rheumatism, abscesses, earaches, and as a tonic. Crinum pratense (C. pratense) and Crinum longifolium (C. longifolium) are also used as bitter tonics, laxatives and in the management of chest illnesses [243].
Crinum zeylanicum (C. zeylanicum) L. is used in Sri Lanka to treat abscesses and fevers; the bulbs are also used as rubefacient in rheumatism and against snake bites; and the juice from the leaves used to treat earaches [244].
The roots of Crinum species have been used in African traditional medicine to cure urinary infections, coughs and colds, renal and hepatic disorders, ulcers, sexually transmitted infections, and backache, as well as enhance breastfeeding in both animal and human mothers [241]. Crinum kirkii Bak. (C. kirkii), a widespread East African grassland plant, is used to heal wounds in Kenya. In Tanzania, the fruit and inner part of the bulbs are used as purgatives, and the outer scales employed as rat poison [245,246]. Extracts of Crinum delagoense (C. delagoense) Verdoorn is utilized in Zulu and Xhosa traditional medicine in South Africa to treat urinary tract infections and body oedema [247,248,249]. Rheumatism, aching joints, septic sores, varicose veins, and kidney and bladder infections have all been treated using the South African Crinum bulbispermum (C. bulbispermum) [250]. In Cameroon, Crinum pupurascens (C. pupurascens) Herb is used to treat sexual asthenia and spleen disorders. Crinum species (C. defixum Keraudren et Gawl., C. firmifolium Baker, C. modestum Baker) are as well used in Madagascar to treat abscesses, anthrax, and otitis. It is also employed as an emetic, diaphoretic, and emollient. Externally, Crinum firmifolium (C. firmifolium) is used to treat a variety of parasite skin afflictions [40,243].

4.3. Phytochemistry of Crinum

Several phytochemical and pharmacological studies have been conducted on the genus Crinum. The compounds isolated from various species of Crinum as well as their biological activities have been outlined in Table 11.
Chemical structures of common compounds from Crinum are shown in Figure 5.

4.4. Pharmacological Activities of Crinum

4.4.1. Anti-Inflammatory and Analgesic Effects

The anti-inflammatory and the analgesic properties of various Crinum species have been investigated by several authors. The anti-inflammatory effect of C. asiaticum as well as its effect on bradykinin-induced contractions on isolated uterus has been reported [288,289,290,291]. The ethanolic extract of C. asiaticum demonstrated significant analgesic effect in an acetic-acid-induced writhing test [292]. Antipyretic and anti-inflammatory properties of C. jagus were recently reported by Minkah and Danquah [291]. Leaf extract of C. bulbispermum has also been established to possess antinociceptive effects [293,294].

4.4.2. Anticancer and Cytotoxicity Effects

The cytotoxic effects of C. asiaticum extract was investigated and was shown to exert toxic effect on brine shrimps and murine P388 D1 cells [294,295,296,297,298]. Yui et al. demonstrated that hot water extracts of C. asiaticum exhibited potent inhibition of calprotectin-induced cytotoxicity in MM46 mouse mammary carcinoma cells. This activity whi1ch was later attributed to lycorine, an active compound in C. asiaticum [297]. Some alkaloids isolated from the bulbs of C. asiaticum have been reported to show remarkable inhibition against tumor cell lines A549, LOVO, HL-60, and 6T-CEM [261].
The extract of C. asiaticum exhibited antiproliferative and chemosensitizing effects against multi-drug-resistant cancer cells [298,299]. The antiangiogenic activity of the methanolic leaf extract of C. asiaticum was evaluated and established by Yusoff [300]. The cytotoxic effect of the essential oil extracted from C. asiaticum was as well established in MCF-7 cells [301]. A recent work done by Yu et al. reported the inhibition of the growth of HepG 2 cells in a dose-dependent manner by polysaccharide CAL-n, an isolate from C. asiaticum [262]. Also, the neuroprotection and anti-neuroinflammatory effects in Neuronal Cell Lines were reported by Lim et al. [279,302]. Alkaloids from C. bulbispermum have also been reported to possess cytotoxic activities [284]. Evaluation of the cytoprotective potential of C. bulbispermum, after induction of toxicity using rotenone, in SH-SY5Y neuroblastoma cells proved that, the plant has such effect as reported [303]. Aboul-Ela et al. [279] tested the cytotoxic effect of C. bulbispermum bulbs using the brine shrimp bioassay.

4.4.3. Antimicrobial Properties

The in vitro antitubercular effects of C. asiaticum on Mycobacterium tuberculosis (M. tuberculosis) surrogate, Mycobacterium smegmatis (M. smegmatis), were reported [291,304]. C. asiaticum was shown to possess a broad-spectrum antimicrobial activity against Gram-positive, Gram-negative bacteria and fungal pathogens [291,299]. Antifungal activities of the essential oil and extracts of C. asiaticum against pathogenic fungi have also been established [305,306]. It is reported that the methanolic root extract of C. asiaticum exerts significant anti-HIV-1 activity [307]. The ethanolic extract of C. asiaticum significantly inhibited selected bacteria as evaluated by Naira et al. [308]. Dichloromethane extract of C. asiaticum was found to be the most effective against selected oral and vaginal Candida species [309]. Minkah and Danquah again demonstrated the antimicrobial activity of extracts of C. jagus against clinically significant microorganisms in the High-throughput spot culture growth inhibition (HT-SPOTi) assay [291]. Water/Ethanol extract of C. jagus was observed to be active on Shigella flexneri-induced diarrhea in rats [310]. The antimicrobial and antioxidant properties of C. jagus make it suitable as a wound healing agent [311]. The crude methanolic extract of C. jagus was investigated to have effect on Mycobacterium tuberculosis [312,313]. The crude alkaloid of C. jagus inhibited Dengue virus infection [314]. C. macowanii has also been shown to possess biological effects such as antifungal, antiviral and antiplasmodial activities [315].

4.4.4. Antioxidant Properties

There antioxidant effects of C. asiaticum have been studied extensively. The ethanolic extract exhibited protective effects on human erythrocyte [316]. C. asiaticum bulbs also exerted remarkable free radical scavenging ability [317]. The antioxidant activity of the ethanolic extract of C. asiaticum leaves in alloxan-induced diabetic rats was well demonstrated [318]. More recent work on the methanolic extract of C. asiaticum showed antioxidant effects [319]. Potent DPPH radical scavenging activity was also observed for the aqueous C. asiaticum leaf extract [304]. Both the leaves and bulbs of C. jagus are important sources of antioxidant compounds [320]. A methanolic bulb extract of C. bulbispermum showed mild radical scavenging activity [321]. The leaf extracts of C. bulbispermum also showed modest antioxidant activity in a thiobarbituric acid reactive substances assay [297].

4.4.5. Other Pharmacological Properties

Kumar reported the wound healing activities of the ethanolic C. asiaticum extract. The extract was found to possess pro-healing effects when topically applied on animal models by influencing various stages of healing process [322]. C asiaticum extract and norgalanthamine potentially influenced hair growth via inhibition of 5α-reductase activity and TGF-β1-induced canonical pathway [39,314]. There is a report on the inhibitory effects of three C. asiaticum genotypes against key enzymes implicated in the pathogenesis of Alzheimer’s disease and diabetes [319].
The anti-obesity effect of the C. asiaticum extract on a high-fat diet-induced obesity in monogenic mice has been reported [323,324]. An active fraction of C. jagus was shown to possess anticonvulsant activities in experimental rats [325].
Ethyl acetate and methanol extracts of C. bulbispermum have also been shown to exhibit acetylcholinesterase inhibitory properties [321]. The alkaloid galanthamine isolated from C. bulbispermum and other genera of Amaryllidaceae, has been approved for the treatment of Alzheimer’s disease [326]. Cognitive enhancing effect of a hydroethanolic extract of C. macowanii against memory impairment induced by aluminum chloride in balb/c mice has as well been reported [327].

5. The Genus Cyrtanthus

5.1. Botanical Description

Another large genus of the family Amaryllidaceae is Cyrtanthus. Cyrtanthus is derived from a Greek word for curved flower [6]. Species of this genus have numerous, black, winged seeds and give off a strong onion smell [6]. They possess a rhizome or bulb, flowers and a loculicidal capsule fruit [6]. They have leaves that are linear to lorate [6]. Flowers are funnel shaped with their stamens fixed in the corolla tube [6]. Species that belong to this genus include Cyrtanthus elatus (C. elatus) (Jacq.) Traub, Cyrtanthus obliquus (C. obliquus) (L.f.) Aiton, and Cyrtanthus mackenii (C. mackenii) Hook [44].

5.2. Geographical Distribution

Cyrtanthus is diverse and is a large sub-Saharan Africa genus consisting of approximately 55 species found mostly in South Africa. Cyrtanthus extends from the summer-dry southwest to the summer rainfall northeast [328]. The genus displays diverse floral morphology. The three major lineages show varying biogeographic affinities.
Clade A comprises taxa located in Southern African Grassland Biome with a few outliers in the Savanna Biome to the east and north, the Indian Ocean Coastal Belt Biome to the extreme east and the Fynbos Biome to the south [328]. Hence, it falls in the Afrotemperate Phytogeographical Region [329] that encompasses Afromontane phytochorion in the north and the Cape Floristic Region in the south [328]. Most existing species in the Afrotemperate lineage (Cyrtanthus attenuatus (C. attenuatus), Cyrtanthus macowanii (C. macowanii), Cyrtanthus epiphyticus (C. epiphyticus), C. mackenii subsp. cooperi, Cyrtanthus huttonii (C. huttonii), Cyrtanthus macmasteri (C. macmasteri), Cyrtanthus suaveolens (C. suaveolens), Cyrtanthus stenanthus (C. stenanthus var. stenanthus) and Cyrtanthus flanaganii (C. flanaganii) occur currently in the south-eastern African temperate grasslands. Cyrtanthus tuckii var. transvaalensis (C. tuckii) is the only species found in the grassland of the Highveld in the northern parts of South Africa. Few species are found outside this grassland area and includes Cyrtanthus angustifolius (C. angustifolius), Cyrtanthus fergusoniae (C. fergusoniae) and Cyrtanthus aureolinus (C. aureolinus) in the Cape Region together with C. mackenii subsp. Mackenii and Cyrtanthus brachyscyphus (C. brachyscyphus) that occupies drainage lines on the subtropical Indian Ocean Coastal Belt [330]. Southern Africa is the area where Cyrtanthus breviflorus (C. breviflorus) is found extending northwards in a series of disjunct populations along mountain corridors to East Africa and Angola.
Clade B is limited to the Fynbos and Succulent Karoo Biomes which constitute the Greater Cape Region, referred to hereafter as ‘the Cape’ [331]. Cyrtanthus labiatus (C. labiatus) and Cyrtanthus montanus (C. montanus) from the Baviaansklo of Mountains and Eastern Cape are found at the interface of the Fynbos and Albany Thicket Biomes. The Richtersveld species, Cyrtanthus herrei (C. herrei) is found in the semi-arid Succulent Karoo [328]. Most species found in ‘the Cape’ lineage are located on the summer-dry, southeast coast forelands with half the number in the Fynbos of the nonseasonal rainfall Eastern Cape. Cyrtanthus carneus (C. carneus, C. elatus, Cyrtanthus guthrieae (C. guthrieae, C. labiatus, Cyrtanthus leptosiphon (C. leptosiphon), Cyrtanthus leucanthus (C. leucanthus, Cyrtanthus montanus (C. montanus), and Cyrtanthus odorus (C. odorus) are found in specific vegetation types and soils.
Only two species of this taxon, namely Cyrtanthus collinus (C. collinus) and Cyrtanthus ventricosus (C. ventricosus) are well known, inhabiting the same soils and aspect in habitats on the continuous Cape Fold mountain ranges [328]. Cyrtanthus collinus is found on the coastal and inland mountains of the southern Cape and C. ventricosus extends from the Cape Peninsula into the Eastern Cape [328].
Most species of Clade C are found in the eastern lowlands and midlands of southern Africa, where they are concentrated in the subtropical biomes, Albany Thicket and Savanna [330,332]. This lineage constitutes Cyrtanthus flammosus (C. flammosus) and Cyrtanthus spiralis (C. spiralis), which are narrowly widespread to the Albany Thicket Biome. Confined to the Savanna Biome are Cyrtanthus eucallus (C. eucallus) and Cyrtanthus galpinii (C. galpinii) in the Lowveld. Other species span the Albany Thicket and Savanna Biomes: the Eastern Cape Cyrtanthus helictus (C. helictus) and, extending northwards from the Albany region through South Africa, Zimbabwe, western Mozambique and East Africa into Sudan, is Cyrtanthus sanguineus (C. sanguineus) [328]. Cyrtanthus obliquus, adapted to nutrient-poor soils, occupies rocky habitats in east–west tending valleys. A summary of their geographic distribution is presented in Table 12.

5.3. Traditional Uses

Cyrtanthus obliquus, locally known as umathunga in South Africa, is used traditionally in the management of chronic coughs, headaches and scrofula [43,44]. C. obliquus root infusions are also employed in the management of stomach aches [333] while the crushed roots have been reported to find use in the management of leprosy [334]. Cyrtanthus species are also employed in the management of ailments associated with pregnancy, as well as cystitis, age-related dementia and leprosy [43,44]. Bulbs of C. contractus extracted in May and September is widely used locally in the management of mental illness, infections, inflammation, and cancer [335]. Infusions from species such as C. breviflorus, C. contractus, C. mackenii, C. sanguineus, C. stenanthus and C. tuckii are used by the Zulu in South Africa as protective sprinkling charms against storms and evil spirits [336]. Extracts of C. breviflorus Harv. are used as an anti-emetic agent and in the management of worm infestations such as tapeworm and roundworm. Extracts of C. elatus also finds use in the management of cough, headache and in labour induction [337].

5.4. Phytochemistry of Cyrtanthus

Species of Cyrtanthus have been identified as reservoirs for a host of chemical compounds. In a study performed by Mahlangeni et al., four homoisoflavanones, namely 5,7-dihydroxy-6-methoxy-3-(4′-methoxybenzyl)chroman-4-one, 5,7-dihydroxy-6-methoxy-3-(4′hydroxybenzyl)chroman-4-one and two 5,7-dihydroxy-6-methoxy-3-(4′-methoxybenzylidene)chroman-4-one, 5,7-dihydroxy-3-(4′hydroxybenzylidene)-chroman-4-one were isolated from the hexane, methanol and dichloromethane extracts of Cyrtanthus obliquus [338]. The bulbs of C. obliquus extracted with ethanol also revealed the presence of novel alkaloid obliquine, as well as 1α-hydroxygalanthamine, 3-epimacronine, narcissidine, tazettine and trisphaeridine [339].
The presence of lycorine, tazettine and 11-hydroxyvittatine in dried bulb ethanol extract of Cyrtanthus mackenii (Hook f.) has been demonstrated by Masi et al. [340]. Fresh bulb methanol extracts of C. contractus also contains a phenanthridone alkaloid called narciclasine [335]. Furthermore, two crinine alkaloids; haemanthamine and haemanthidine have been isolated from fresh bulb ethanol extracts of C. elatus. Further studies on the alcoholic extracts of the fresh bulbs also yielded the alkaloids zephyranthine, galanthamine and 1,2-O-diacetylzephyranthine [43,44]. Tazettine, maritidine, O-methylmaritidine, and papyramine are all phytochemicals that have been identified in fresh bulb methanol extracts of C. falcatus [337].
Chemical structure of compounds isolated from Cyrtanthus have been shown in Figure 6.

5.5. Pharmacological Activities

5.5.1. Antioxidant Activity

5,7-dihydroxy-6-methoxy-3-(4′-methoxybenzyl)chroman-4-one and 5,7-dihydroxy-6-methoxy-3-(4′-hydroxybenzyl)chroman-4-one isolated from the fresh bulbs of C. obliquus have been shown to possess significant antioxidant activity with an IC50 of 371.54 and 288.40 µg/mL, respectively [338].

5.5.2. Anti-Inflammatory Activity

The methanol extract of the bulbs of C. contractus has been investigated and shown to possess significant anti-inflammatory activity. The extract exhibited dose-dependent inhibition of E-selectin, a proinflammatory agent, when tested on endothelial cells. Further studies of the methanol extract on human umbilical vein endothelial cells revealed a concentration-dependent reduction in THP-1 adhesion via blockade of the expression of endothelial adhesion molecule ICAM-1. Narciclasine was identified as the main anti-inflammatory compound in the methanol extract of the bulbs of C. contractus [335].
The dichloromethane (DCM) extracts of C. falcatus (roots) and C. mackenii (leaves) were shown to interfere with the activity of cyclooxygenase 2 (COX-2) by at least 90%. DCM extract of C. suaveolens also blocked prostaglandin synthesis via antagonizing COX-2 activity by 81.6 %. Moderate inhibition (approximately 70%) of COX-2 activity was also observed with the methanol extracts of the roots and leaves of C. falcatus [341,342]. Selective inhibition of COX-2 by these extracts makes them suitable candidates for development for clinical use.

5.5.3. Inhibition of Acetylcholinesterase

The phenanthridone alkaloid nacriprimine, isolated from the ethanolic bulb extract of C. contractus has been shown to possess mild acetylcholinesterase inhibition property with an IC50 of 78.9 µg/mL compared to the 40-fold more potent standard galanthamine with an IC50 of 1.9 µg/mL [11].

5.5.4. Antimicrobial Activity

Cyrtanthus species and their isolated compounds have demonstrated noteworthy antimicrobial activity against a panel of microorganisms. C. suaveolens bulbs/roots and leaves isolated with DCM demonstrated broad-spectrum antimicrobial activity against B. subtilis, E. coli, K. pneumoniae, M. luteus and S. aureus with zones of inhibition ranging between 0.13–0.91 mm. DCM extracts of C. falcatus also inhibited the growth of B. subtilis S. aureus and E. coli. C. mackenii bulb/root extracts also inhibited the growth M. luteus and S. aureus [337].
Haemanthamine and haemanthidine isolated from the bulbs of C. elatus have been investigated for their activity against parasitic protozoans [43]. Haemanthamine showed activity against trophozoite stage of Entamoeba histolytica (E. histolytica) HK9 with an IC50 of 0.75 μg/mL and mild activity against Plasmodium falciparum (P. falciparum) NF54 with an IC50 of 0.67 μg/mL. The activity against E. histolytica was compared to ornidazole with an IC50 0.28 μg/mL whiles the activity against P. falciparum was compared to chloroquine with an IC50 of 0.004 μg/mL and artemisinin with an IC50 of 0.002 μg/mL [43].
Haemanthidine also showed weak activity against P. falciparum, T. brucei rhodesiense STIB 900, and T. cruzi Tulahuen C4 with an IC50 of 0.70, 1.1 and 1.38 μg/mL, respectively. Melarsoprol with an IC50 of 0.002 μg/mL and benznidazole with an IC50 of 0.56 μg/mL were used as standards for Trypanosoma brucei rhodesiense STIB 900, and Trypanosoma cruzi Tulahuen C4, respectively [43].

5.5.5. Cytotoxic Activity

Haemanthamine isolated from C. elatus was shown to possess cytotoxic activity which was mediated via the apoptotic pathway as depicted in rat hepatoma cell (5123tc). The ED50 was determined at 15 μM and this result was of particular interest due to its selectivity; haemanthamine demonstrated insignificant activity in normal human embryo kidney (293t) cells [337].
Alkaloids isolated from C. obliquus tested for cytotoxic activity against Chinese Hamster ovarian and human hepatoma (hepG2) cells showed no cytotoxic activity up to a concentration of 100 μg/mL [339].
Tazettine isolated from C. falcatus and other members of Amaryllidaceae has been reported to possess cytotoxic activity on colon cell line murine alveolar non-tumoral fibroblast [343,344]. Papyramine, also extracted from C. falcatus showed cytotoxic activity against murine alveolar non-tumoral fibroblast and human lymphoid neoplasm as well [343,344].

5.5.6. Miscellaneous Pharmacological Activities

Roots of C. falcatus and C. surveolens extracted with DCM exhibited mutagenicity in Salmonella strain TA98 which was higher than that observed in the leaves of these plants. Mutagenicity was, however, not observed in the methanol extracts of these plants [337]. The mutagenicity of C. suaveolens has been attributed to the compound captan isolated from the bulbs/roots using DCM [344].
A summary of the traditional uses, phytochemicals and pharmacological activities of Cyrtanthus species have been highlighted in Table 13.

6. Conclusions

The discovery of new drugs in response to the growing burden of infectious and non-communicable diseases is of utmost necessity in this era. The genera Tulbaghia, Allium, Crinum, and Cyrtanthus of the Amaryllidaceae family have been well presented and shown to be a source of promising medicinal compounds with varying biological properties. Further research is therefore necessary to propel these compounds through clinical trials for possible usage in therapeutics. Although natural products have been attributed with high safety profiles, the presence of mutagenic compounds in crude extracts of these plants underscores the importance of pharmacological studies prior to their use in traditional medicine. These findings are relevant in light of augmenting the lean pipeline of drug discovery.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27144475/s1, Table S1: Compounds from Tulbaghia. Reference [345] is cited in the Supplementary Materials.

Author Contributions

Conceptualization and study design, C.A.D. and P.A.B.M.; literature search and review, C.A.D., P.A.B.M., T.A.A., P.M., M.O., P.D., S.R., I.O.D.J., K.B.A., S.O.S., I.N.N., V.J.M., S.B. and S.G.; writing—original draft preparation, C.A.D., P.A.B.M., T.A.A., P.M., M.O., P.D., S.R., I.O.D.J., K.B.A., S.O.S., I.N.N., V.J.M., S.B. and S.G.; writing—review and editing, C.A.D., P.A.B.M., P.M., I.O.D.J., V.J.M., S.B. and S.G.; formatting and design—P.A.B.M. and P.M.; project administration, C.A.D. and P.A.B.M.; supervision, C.A.D. All authors have read and agreed to the published version of the manuscript.

Funding

The authors received no specific grant from the government, private or non-profit organizations.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

MIC, minimum inhibitory concentration; MBC, minimum bactericidal concentration; MTT, 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide; MoA, mode of action; ROS, reactive oxygen species; TEAC, Trolox equivalent antioxidant capacity; FRAP, ferric-reducing antioxidant power; DPPH, 2,2-diphenyl-1-picryl-hydrazyl-hydrate; AAPH, 2,2′-azobis-2-amidinopropane dihydrochloride; TG, triglyceride; TC, total cholesterol; VLDL, very low-density lipoprotein; HDL, high-density lipoprotein; BP, blood pressure; AVD, atherothrombotic vascular disease; DCM, dichloromethane.

References

  1. Barile, E.; Capasso, R.; Izzo, A.A.; Lanzotti, V.; Sajjadi, S.E.; Zolfaghari, B. Structure-activity relationships for saponins from Allium hirtifolium and Allium elburzense and their antispasmodic activity. Planta Med. 2005, 71, 1010–1018. [Google Scholar] [CrossRef] [PubMed]
  2. Chase, M.W.; Reveal, J.L.; Fay, M.F. A subfamilial classification for the expanded asparagalean families Amaryllidaceae, Asparagaceae and Xanthorrhoeaceae. Bot. J. Linn. Soc. 2009, 161, 132–136. [Google Scholar] [CrossRef] [Green Version]
  3. Takos, A.M.; Rook, F. Towards a Molecular Understanding of the Biosynthesis of Amaryllidaceae Alkaloids in Support of Their Expanding Medical Use. Int. J. Mol. Sci. 2013, 14, 11713–11741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Vosa, C.G.; Siebert, S.J.; Van Wyk, A.E.B. Micromorphology and cytology of Prototulbaghia siebertii, with notes on its taxonomic significance. Upsp. Inst. Repos. 2011, 41, 311–314. [Google Scholar]
  5. Elgorashi, E.E.; van Staden, J. Bioactivity and Bioactive Compounds of African Amaryllidaceae; ACS Publications: Washington, DC, USA, 2009; ISBN 1947-5918. [Google Scholar]
  6. Fenwick, G.R.; Hanley, A.B.; Whitaker, J.R. The genus allium—Part 1. Crit. Rev. Food Sci. Nutr. 1985, 22, 199–271. [Google Scholar] [CrossRef]
  7. Fenwick, G.R.; Hanley, A.B. The genus allium—Part 2. Crit. Rev. Food Sci. Nutr. 1985, 22, 273–377. [Google Scholar] [CrossRef]
  8. Aremu, A.O.; Van Staden, J. The genus Tulbaghia (Alliaceae)—A review of its ethnobotany, pharmacology, phytochemistry and conservation needs. J. Ethnopharmacol. 2013, 149, 387–400. [Google Scholar] [CrossRef]
  9. Styger, G.; Aboyade, O.M.; Gibson, D.; Hughes, G. Tulbaghia—A Southern African Phytomedicine. J. Altern. Complement. Med. 2016, 22, 255–261. [Google Scholar] [CrossRef]
  10. Jagtap, U.B.; Lekhak, M.M.; Fulzele, D.P.; Yadav, S.R.; Bapat, V.A. Analysis of selected Crinum species for galanthamine alkaloid: An anti-Alzheimer drug. Curr. Sci. 2014, 107, 2008–2010. [Google Scholar]
  11. Nair, J.J.; Aremu, A.O.; Van Staden, J. Isolation of narciprimine from Cyrtanthus contractus (Amaryllidaceae) and evaluation of its acetylcholinesterase inhibitory activity. J. Ethnopharmacol. 2011, 137, 1102–1106. [Google Scholar] [CrossRef]
  12. Torras-Claveria, L.; Berkov, S.; Codina, C.; Viladomat, F.; Bastida, J. Daffodils as potential crops of galanthamine. Assessment of more than 100 ornamental varieties for their alkaloid content and acetylcholinesterase inhibitory activity. Ind. Crops Prod. 2013, 43, 237–244. [Google Scholar] [CrossRef]
  13. Shang, A.; Cao, S.-Y.; Xu, X.-Y.; Gan, R.-Y.; Tang, G.-Y.; Corke, H.; Mavumengwana, V.; Li, H.-B. Bioactive Compounds and Biological Functions of Garlic (Allium sativum L.). Foods 2019, 8, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sharma, D.; Rani, R.; Chaturvedi, M.; Rohilla, P.; Yadav, J.P. In silico and in vitro approach of Allium cepa and isolated quercetin against MDR bacterial strains and Mycobacterium smegmatis. S. Afr. J. Bot. 2019, 124, 29–35. [Google Scholar] [CrossRef]
  15. Stoica, F.; Aprodu, I.; Enachi, E.; Stănciuc, N.; Condurache, N.N.; Duță, D.E.; Bahrim, G.E.; Râpeanu, G. Bioactive’s Characterization, Biological Activities, and In Silico Studies of Red Onion (Allium cepa L.) Skin Extracts. Plants 2021, 10, 2330. [Google Scholar] [CrossRef] [PubMed]
  16. Albishi, T.; John, J.A.; Al-Khalifa, A.S.; Shahidi, F. Antioxidant, anti-inflammatory and DNA scission inhibitory activities of phenolic compounds in selected onion and potato varieties. J. Funct. Foods 2013, 5, 930–939. [Google Scholar] [CrossRef]
  17. Benmalek, Y.; Yahia, O.A.; Belkebir, A.; Fardeau, M.-L. Anti-microbial and anti-oxidant activities of Illicium verum, Crataegus oxyacantha ssp monogyna and Allium cepa red and white varieties. Bioengeineered 2013, 4, 244–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Elberry, A.A.; Mufti, S.; Al-Maghrabi, J.; Abdel Sattar, E.; Ghareib, S.A.; Mosli, H.A.; Gabr, S.A. Immunomodulatory effect of red onion (Allium cepa Linn) scale extract on experimentally induced atypical prostatic hyperplasia in Wistar rats. Mediat. Inflamm. 2014, 2014, 640746. [Google Scholar] [CrossRef] [Green Version]
  19. Lanzotti, V. The analysis of onion and garlic. J. Chromatogr. 2006, 1112, 3–22. [Google Scholar] [CrossRef] [Green Version]
  20. Rouf, R.; Uddin, S.J.; Sarker, D.K.; Islam, M.T.; Ali, E.S.; Shilpi, J.A.; Nahar, L.; Tiralongo, E.; Sarker, S.D. Antiviral potential of garlic (Allium sativum) and its organosulfur compounds: A systematic update of pre-clinical and clinical data. Trends Food Sci. Technol. 2020, 104, 219–234. [Google Scholar] [CrossRef]
  21. Harazem, R.; El Rahman, S.A.; El-Kenawy, A. Evaluation of Antiviral Activity of Allium cepa and Allium sativum Extracts Against Newcastle Disease Virus. Alex. J. Vet. Sci. 2019, 61, 108–118. [Google Scholar] [CrossRef]
  22. Elmi, T.; Hajialiani, F.; Asadi, M.R.; Sadeghi, S.; Namazi, M.J.; Tabatabaie, F.; Zamani, Z. Antimalarial effects of the hydroalcoholic extract of Allium paradoxum in vitro and in vivo. J. Parasit. Dis. 2021, 45, 1055–1064. [Google Scholar] [CrossRef]
  23. Ruslan, M.S.; Baba, M.S. In vivo antimalarial assessment and toxicity evaluation of garlic (Allium sativum) in plasmodium berghei NK65-induced mice. Malays. Appl. Biol. 2018, 47, 17–24. [Google Scholar]
  24. Syaban, M.F.R.; Rachman, H.A.; Arrahman, A.D.; Hudayana, N.; Khamid, J.P.; Pratama, F.A. Allium sativum as antimalaria agent via falciapin protease-2 inhibitor mechanism: Molecular docking perspective. Clin. Res. J. Intern. Med. 2021, 2, 130–135. [Google Scholar] [CrossRef]
  25. Upadhyay, R.K. Nutritional and therapeutic potential of Allium vegetables. J. Nutr. Ther. 2017, 6, 18–37. [Google Scholar] [CrossRef]
  26. Thomson, M.; Ali, M. Garlic [Allium sativum]: A review of its potential use as an anti-cancer agent. Curr. Cancer Drug Targets 2003, 3, 67–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Corea, G.; Fattorusso, E.; Lanzotti, V.; Capasso, R.; Izzo, A.A. Antispasmodic saponins from bulbs of red onion, Allium cepa L. var. Tropea. J. Agric. Food Chem. 2005, 53, 935–940. [Google Scholar] [CrossRef] [Green Version]
  28. Galmarini, C.R.; Goldman, I.L.; Havey, M.J. Genomics Genetic analyses of correlated solids, flavor, and health-enhancing traits in onion (Allium cepa L.). Mol. Genet. Genom. 2001, 265, 543–551. [Google Scholar] [CrossRef]
  29. Takahashi, M.; Shibamoto, T. Chemical compositions and antioxidant/anti-inflammatory activities of steam distillate from freeze-dried onion (Allium cepa L.) sprout. J. Agric. Food Chem. 2008, 56, 10462–10467. [Google Scholar] [CrossRef]
  30. Nishimura, H.; Wijaya, C.H.; Mizutani, J. Volatile flavor components and antithrombotic agents: Vinyldithiins from Allium victorialis L. J. Agric. Food Chem. 1988, 36, 563–566. [Google Scholar] [CrossRef]
  31. Brace, L.D. Cardiovascular benefits of garlic (Allium sativum L.). J. Cardiovasc. Nurs. 2002, 16, 33–49. [Google Scholar] [CrossRef]
  32. Ali, M.; Thomson, M.; Afzal, M. Garlic and onions: Their effect on eicosanoid metabolism and its clinical relevance. Prostaglandins Leukot. Essent. Fat. Acids 2000, 62, 55–73. [Google Scholar] [CrossRef] [PubMed]
  33. Sabiu, S.; Madende, M.; Ajao, A.A.; Aladodo, R.A.; Nurain, I.O.; Ahmad, J.B. The Genus Allium (Amaryllidaceae: Alloideae): Features, Phytoconstituents, and Mechanisms of Antidiabetic Potential of Allium cepa and Allium sativum, 2nd ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2019; ISBN 9780128138229. [Google Scholar]
  34. Muñoz-Torrero López-Ibarra, D. Recent Advances in Pharmaceutical Sciences I; Transworld Research Network: Trivandrum, India, 2011; ISBN 8178955288. [Google Scholar]
  35. Akash, M.S.H.; Rehman, K.; Chen, S. Spice plant Allium cepa: Dietary supplement for treatment of type 2 diabetes mellitus. Nutrition 2014, 30, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  36. Corzo-Martínez, M.; Corzo, N.; Villamiel, M. Biological properties of onions and garlic. Trends Food Sci. Technol. 2007, 18, 609–625. [Google Scholar] [CrossRef]
  37. Kumar, K.P.S.; Debjit, B.; Pankaj, T. Allium cepa: A traditional medicinal herb and its health benefits. J. Chem. Pharm. Res. 2010, 2, 283–291. [Google Scholar]
  38. Shri, R.; Bora, K.S. Neuroprotective effect of methanolic extracts of Allium cepa on ischemia and reperfusion-induced cerebral injury. Fitoterapia 2008, 79, 86–96. [Google Scholar] [CrossRef]
  39. Kongkwamcharoen, C.; Itharat, A.; Pipatrattanaseree, W.; Ooraikul, B. Effects of Various Preextraction Treatments of Crinum asiaticum Leaf on Its Anti-Inflammatory Activity and Chemical Properties. Evid. Based. Complement. Alternat. Med. 2021, 2021, 8850744. [Google Scholar] [CrossRef] [PubMed]
  40. Fennell, C.W.; Van Staden, J. Crinum species in traditional and modern medicine. J. Ethnopharmacol. 2001, 78, 15–26. [Google Scholar] [CrossRef]
  41. 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] [Green Version]
  42. Takaidza, S.; Pillay, M.; Mtunzi, F.M. Biological activities of species in the genus Tulbaghia: A review. Afr. J. Biotechnol. 2015, 14, 3037–3043. [Google Scholar]
  43. Herrera, M.R.; Machocho, A.K.; Nair, J.J.; Campbell, W.E.; Brun, R.; Viladomat, F.; Codina, C.; Bastida, J. Alkaloids from Cyrtanthus elatus. Fitoterapia 2001, 72, 444–448. [Google Scholar] [CrossRef]
  44. Nair, J.J.; van Staden, J. Chemical and biological studies of the South African Amaryllidaceae genera Crinum, Ammocharis, Amaryllis, Cyrtanthus and Brunsvigia. S. Afr. J. Bot. 2021, 142, 467–476. [Google Scholar] [CrossRef]
  45. Heinrich, M.; Teoh, H.L. Galanthamine from snowdrop—the development of a modern drug against Alzheimer’s disease from local Caucasian knowledge. J. Ethnopharmacol. 2004, 92, 147–162. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. Govaerts, R. World Checklist of Selected Plant Species. Facilitated by the Royal Botanic Gardens, Kew. 2015. Available online: https://wcsp.science.kew.org/about.do (accessed on 25 March 2022).
  48. Kubec, R.; Velíšek, J.; Musah, R.A. The amino acid precursors and odor formation in society garlic (Tulbaghia violacea Harv.). Phytochemistry 2002, 60, 21–25. [Google Scholar] [CrossRef]
  49. Dillon, H.; Nelson, E.C. Tulbaghia leucantha: Alliaceae. Kew Mag. 1991, 8, 12–15. [Google Scholar] [CrossRef]
  50. Makunga, N.P. Medicinal Plants of South Africa; Briza Publications: Pretoria, South Africa, 2010; Volume 105. [Google Scholar]
  51. Van Wyk, B.E. The potential of South African plants in the development of new food and beverage products. S. Afr. J. Bot. 2011, 77, 857–868. [Google Scholar] [CrossRef] [Green Version]
  52. Raji, I.A.; Obikeze, K.; Mugabo, P.E. Potential beneficial effects of tulbaghia violacea william henry harvey (Alliaceae) on cardiovascular system—A Review. Trop. J. Pharm. Res. 2015, 14, 1111–1117. [Google Scholar] [CrossRef] [Green Version]
  53. Pooley, E. A Field Guide to the Wild Flowers of KwaZulu-Natal and the Eastern Region. Natal Flora Publ. Trust. Pg 2005, 93, 630. [Google Scholar]
  54. Sander, T.; Freyss, J.; Von Korff, M.; Rufener, C. DataWarrior: An open-source program for chemistry aware data visualization and analysis. J. Chem. Inf. Model. 2015, 55, 460–473. [Google Scholar] [CrossRef]
  55. Pino, J.A.; Quijano-Celís, C.E.; Fuentes, V. Volatile compounds of tulbaghia violacea harv. J. Essent. Oil-Bear. Plants 2008, 11, 203–207. [Google Scholar] [CrossRef]
  56. Ranglová, K.; Krejčová, P.; Kubec, R. The effect of storage and processing on antimicrobial activity of Tulbaghia violacea. S. Afr. J. Bot. 2015, 97, 159–164. [Google Scholar] [CrossRef]
  57. Smith, S.; Stansbie, J. Flora of Tropical East Africa. Crown Agents for Oversea Governments and Administration; CRC Press: Boca Raton, FL, USA, 2003; p. 230. [Google Scholar]
  58. Takaidza, S.; Mtunzi, F.; Pillay, M. Analysis of the phytochemical contents and antioxidant activities of crude extracts from Tulbaghia species. J. Tradit. Chin. Med. 2018, 38, 272–279. [Google Scholar] [CrossRef]
  59. Staffa, P.; Nyangiwe, N.; Msalya, G.; Nagagi, Y.P.; Nchu, F. The effect of Beauveria bassiana inoculation on plant growth, volatile constituents, and tick (Rhipicephalus appendiculatus) repellency of acetone extracts of Tulbaghia violacea. Vet. World 2020, 13, 1159–1166. [Google Scholar] [CrossRef] [PubMed]
  60. Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef] [PubMed]
  61. Teffo, L.S.; Aderogba, M.A.; Eloff, J.N. Antibacterial and antioxidant activities of four kaempferol methyl ethers isolated from Dodonaea viscosa Jacq. var. angustifolia leaf extracts. S. Afr. J. Bot. 2010, 76, 25–29. [Google Scholar] [CrossRef] [Green Version]
  62. Yang, C.; Yang, W.; He, Z.; Guo, J.; Yang, X.; Wang, R.; Li, H. Kaempferol Alleviates Oxidative Stress and Apoptosis Through Mitochondria-dependent Pathway During Lung Ischemia-Reperfusion Injury. Front. Pharmacol. 2021, 12, 11. [Google Scholar] [CrossRef]
  63. Murray, C.J. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 388, 629–655. [Google Scholar] [CrossRef]
  64. Anand, U.; Jacobo-Herrera, N.; Altemimi, A.; Lakhssassi, N. A comprehensive review on medicinal plants as antimicrobial therapeutics: Potential avenues of biocompatible drug discovery. Metabolites 2019, 9, 258. [Google Scholar] [CrossRef] [Green Version]
  65. Kokoska, L.; Kloucek, P.; Leuner, O.; Novy, P. Plant-Derived Products as Antibacterial and Antifungal Agents in Human Health Care. Curr. Med. Chem. 2018, 26, 5501–5541. [Google Scholar] [CrossRef]
  66. Salam, A.M.; Quave, C.L. Opportunities for plant natural products in infection control. Curr. Opin. Microbiol. 2018, 45, 189–194. [Google Scholar] [CrossRef]
  67. Krstin, S.; Sobeh, M.; Braun, M.S.; Wink, M. Tulbaghia violacea and Allium ursinum extracts exhibit anti-parasitic and antimicrobial activities. Molecules 2018, 23, 313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Eid, H.H.; Metwally, G.F. Phytochemical and biological study of callus cultures of Tulbaghia violacea Harv. Cultivated in Egypt. Nat. Prod. Res. 2017, 31, 1717–1724. [Google Scholar] [CrossRef] [PubMed]
  69. Kumar, P.; Mahato, D.K.; Kamle, M.; Mohanta, T.K.; Kang, S.G. Aflatoxins: A global concern for food safety, human health and their management. Front. Microbiol. 2017, 7, 2170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Belewa, V.; Baijnath, H.; Frost, C.; Somai, B.M. Tulbaghia violacea Harv. plant extract affects cell wall synthesis in Aspergillus flavus. J. Appl. Microbiol. 2017, 122, 921–931. [Google Scholar] [CrossRef]
  71. Somai, B.M.; Belewa, V.; Frost, C. Tulbaghia violacea (Harv) Exerts its Antifungal Activity by Reducing Ergosterol Production in Aspergillus flavus. Curr. Microbiol. 2021, 78, 2989–2997. [Google Scholar] [CrossRef]
  72. Pretorius, J.C. Extracts and Compounds from Tulbaghia Violacea and Their Use as Biological Plant Protecting Agents 2014. Google Patents US8697149B2, 15 April 2014. [Google Scholar]
  73. Ncise, W.; Daniels, C.W.; Etsassala, N.G.E.R.; Nchu, F. Interactive effects of light intensity and ph on growth parameters of a bulbous species (Tulbaghia violacea l.) in hydroponic cultivation and its antifungal activities. Med. Plants 2021, 13, 442–451. [Google Scholar] [CrossRef]
  74. Ncise, W.; Daniels, C.W.; Nchu, F. Effects of light intensities and varying watering intervals on growth, tissue nutrient content and antifungal activity of hydroponic cultivated Tulbaghia violacea L. under greenhouse conditions. Heliyon 2020, 6, 3906. [Google Scholar] [CrossRef] [PubMed]
  75. Malungane, M.M.F.; Florah, M.M. Effect of Crude Extracts of Tulbaghia violacea (Wild Garlic) on Growth of Tomato and Supression of Meloidogyne Species; University of Limpopo: Mankweng, South Africa, 2014. [Google Scholar]
  76. Kaushik, I.; Ramachandran, S.; Prasad, S.; Srivastava, S.K. Drug rechanneling: A novel paradigm for cancer treatment. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2021; Volume 68, pp. 279–290. [Google Scholar]
  77. Cragg, G.M.; Grothaus, P.G.; Newman, D.J. Impact of natural products on developing new anti-cancer agents. Chem. Rev. 2009, 109, 3012–3043. [Google Scholar] [CrossRef]
  78. Mthembu, N.N.; Motadi, L.R. Apoptotic potential role of Agave palmeri and Tulbaghia violacea extracts in cervical cancer cells. Mol. Biol. Rep. 2014, 41, 6143–6155. [Google Scholar] [CrossRef]
  79. Motadi, L.R.; Choene, M.S.; Mthembu, N.N. Anticancer properties of Tulbaghia violacea regulate the expression of p53-dependent mechanisms in cancer cell lines. Sci. Rep. 2020, 10, 12924. [Google Scholar] [CrossRef]
  80. Rivas-García, L.; Romero-Márquez, J.M.; Navarro-Hortal, M.D.; Esteban-Muñoz, A.; Giampieri, F.; Sumalla-Cano, S.; Battino, M.; Quiles, J.L.; Llopis, J.; Sánchez-González, C. Unravelling potential biomedical applications of the edible flower Tulbaghia violacea. Food Chem. 2022, 381, 132096. [Google Scholar] [CrossRef]
  81. Bianchini, G.; Balko, J.M.; Mayer, I.A.; Sanders, M.E.; Gianni, L. Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat. Rev. Clin. Oncol. 2016, 13, 674–690. [Google Scholar] [CrossRef] [PubMed]
  82. Dlamini, Z.; Alouna, M.; Hull, R.; Penny, C. Abstract 2843: The effects of extracts of the indigenous South African plant, Tulbaghia violacea, on triple negative breast cancer cells. In Proceedings of the NCRI Cancer Conference, London, UK, 8–12 November 2021. [Google Scholar]
  83. Deepak, K.G.K.; Vempati, R.; Nagaraju, G.P.; Dasari, V.R.; Nagini, S.; Rao, D.N.; Malla, R.R. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020, 153, 104683. [Google Scholar] [CrossRef] [PubMed]
  84. Takaidza, S.; Kumar, A.M.; Ssemakalu, C.C.; Natesh, N.S.; Karanam, G.; Pillay, M. Anticancer activity of crude acetone and water extracts of Tulbaghia violacea on human oral cancer cells. Asian Pac. J. Trop. Biomed. 2018, 8, 456–462. [Google Scholar] [CrossRef]
  85. Lyantagaye, S.L. Two new pro-apoptotic glucopyranosides from Tulbaghia violacea. J. Med. Plants Res. 2013, 7, 2214–2220. [Google Scholar] [CrossRef] [Green Version]
  86. Lyantagaye, S. Characterization of the Biochemical Pathway of Apoptosis Induced by D-glucopyranoside Derivatives from Tulbaghia violacea. Annu. Res. Rev. Biol. 2014, 4, 962–977. [Google Scholar] [CrossRef]
  87. Lushchak, V.I. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem. Biol. Interact. 2014, 224, 164–175. [Google Scholar] [CrossRef]
  88. Thorpe, G.W.; Fong, C.S.; Alic, N.; Higgins, V.J.; Dawes, I.W. Cells have distinct mechanisms to maintain protection against different reactive oxygen species: Oxidative-stress-response genes. Proc. Natl. Acad. Sci. USA 2004, 101, 6564–6569. [Google Scholar] [CrossRef] [Green Version]
  89. Lourenço, S.C.; Moldão-Martins, M.; Alves, V.D. Antioxidants of natural plant origins: From sources to food industry applications. Molecules 2019, 24, 4132. [Google Scholar] [CrossRef] [Green Version]
  90. Madike, L.N.; Pillay, M.; Popat, K.C. Antithrombogenic properties of Tulbaghia violacea–loaded polycaprolactone nanofibers. J. Bioact. Compat. Polym. 2020, 35, 102–116. [Google Scholar] [CrossRef]
  91. Arhin, I.; Depika, D.; Ajay, B.; Delon, N.; Irene, M. Biochemical, phytochemical profile and angiotensin-1 converting enzyme inhibitory activity of the hydro-methanolic extracts of Tulbaghia acutiloba harv. J. Nat. Remedies 2019, 19, 221–235. [Google Scholar] [CrossRef]
  92. Madike, L.N.; Pillay, M.; Popat, K.C. Antithrombogenic properties of: Tulbaghia violacea aqueous leaf extracts: Assessment of platelet activation and whole blood clotting kinetics. RSC Adv. 2021, 11, 30455–30464. [Google Scholar] [CrossRef] [PubMed]
  93. Glovaci, D.; Fan, W.; Wong, N.D. Epidemiology of Diabetes Mellitus and Cardiovascular Disease. Curr. Cardiol. Rep. 2019, 21, 1–8. [Google Scholar] [CrossRef] [PubMed]
  94. Wild, S.; Roglic, G.; Green, A.; Sicree, R.; King, H. Global Prevalence of Diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care 2004, 27, 1047–1053. [Google Scholar] [CrossRef] [Green Version]
  95. Matheus, A.S.D.M.; Tannus, L.R.M.; Cobas, R.A.; Palma, C.C.S.; Negrato, C.A.; Gomes, M.D.B. Impact of diabetes on cardiovascular disease: An update. Int. J. Hypertens. 2013, 2013, 653789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Moodley, K.; Joseph, K.; Naidoo, Y.; Islam, S.; Mackraj, I. Antioxidant, antidiabetic and hypolipidemic effects of Tulbaghia violacea Harv. (wild garlic) rhizome methanolic extract in a diabetic rat model. BMC Complement. Altern. Med. 2015, 15, 408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Moodley, K.; Mackraj, I. Metabolic effects of tulbaghia violacea harv. In a diabetic model. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 113–122. [Google Scholar] [CrossRef] [Green Version]
  98. Raji, I.; Obikeze, K.; Mugabo, P. Comparison of the acute effects of Tulbaghia violacea William Henry Harvey (Alliaceae) on blood pressure and heart rate of ageing male normotensive Wistar kyoto rats and adult male spontaneously hypertensive rats. Trop. J. Pharm. Res. 2016, 15, 2429–2434. [Google Scholar] [CrossRef] [Green Version]
  99. Moodley, K.; Naidoo, Y.; Mackraj, I. Effects of Tulbaghia violacea Harv. (Alliaceae) rhizome methanolic extract on kidney function and morphology in Dahl salt-sensitive rats. J. Ethnopharmacol. 2014, 155, 1194–1203. [Google Scholar] [CrossRef]
  100. Navar, L.G. The role of the kidneys in hypertension. J. Clin. Hypertens. 2005, 7, 542–549. [Google Scholar] [CrossRef]
  101. Davì, G.; Patrono, C. Platelet Activation and Atherothrombosis. N. Engl. J. Med. 2007, 357, 2482–2494. [Google Scholar] [CrossRef] [PubMed]
  102. Masoud, K.A.A.; Okobi, E.; Ekpo, G.J. Amabeoku Investigation of Some Possible Mechanisms Involved in the Anticonvulsant Activity of Tulbaghia violacea Harv. J. Pharm. Pharmacol. 2017, 5. [Google Scholar] [CrossRef] [Green Version]
  103. Madike, L.N.; Takaidza, S.; Ssemakalu, C.; Pillay, M. Genotoxicity of aqueous extracts of Tulbaghia violacea as determined through an Allium cepa assay. S. Afr. J. Sci. 2019, 115, 1–6. [Google Scholar] [CrossRef] [Green Version]
  104. Madike, L.N.; Takaidza, S.; Ssemakalu, C.C.; Pillay, M. The effect of extracts of Tulbaghia violacea on the proliferation of a murine macrophage cell line. S. Afr. J. Bot. 2020, 130, 185–197. [Google Scholar] [CrossRef]
  105. Stavělíková, H. Morphological characteristics of garlic (Allium sativum L.) genetic resources collection—Information. Hortic. Sci. 2008, 35, 130–135. [Google Scholar] [CrossRef] [Green Version]
  106. Wheeler, E.J.; Mashayekhi, S.; Mcneal, D.W.; Travis Columbus, J.; Chris Pires, J. Molecular systematics of Allium subgenus Amerallium (Amaryllidaceae) in North America. Am. J. Bot. 2013, 100, 701–711. [Google Scholar] [CrossRef]
  107. Fernandes, S.; Gois, A.; Mendes, F.; Perestrelo, R.; Medina, S.; Câmara, J.S. Typicality Assessment of Onions (Allium cepa) from Different Geographical Regions Based on the Volatile Signature and Chemometric Tools. Foods 2020, 9, 375. [Google Scholar] [CrossRef] [Green Version]
  108. Jo, J.; Purushotham, P.M.; Han, K.; Lee, H.-R.; Nah, G.; Kang, B.-C. Development of a Genetic Map for Onion (Allium cepa L.) Using Reference-Free Genotyping-by-Sequencing and SNP Assays. Front. Plant Sci. 2017, 8, 1342. [Google Scholar] [CrossRef]
  109. Najeebullah, S.; Shinwari, Z.K.; Jan, S.A.; Khan, I.; Ali, M. Ethno medicinal and phytochemical properties of genus Allium: A review of recent advances. Pak. J. Bot. 2021, 53, 135–144. [Google Scholar] [CrossRef]
  110. Lawande, K.E. Onion. In Handbook of Herbs and Spices; Elsevier: Amsterdam, The Netherlands, 2012; pp. 417–429. [Google Scholar]
  111. Debin, W.; Jiande, G.; Guangshu, L. General situation of Allium crops in China. In Proceedings of the IV International Symposium on Edible Alliaceae 688, Beijing, China, 21–26 April 2004; pp. 327–332. [Google Scholar]
  112. Food and Agriculture Organization of the United Nations. Available online: http://www.fao.org/faostat/en/#data (accessed on 26 March 2022).
  113. Bartolucci, F.; Iocchi, M.; De Castro, O.; Conti, F. Allium ducissae (A. subgen. Polyprason, Amaryllidaceae) a New Species from the Central Apennines (Italy). Plants 2022, 11, 426. [Google Scholar] [CrossRef]
  114. Friesen, N. The genus Allium L. in the flora of Mongolia. Feddes Repert. 1995, 106, 59–81. [Google Scholar] [CrossRef]
  115. Sinitsyna, T.A. Genus Allium L.(Alliaceae) in Siberia. Vavilovia 2020, 2, 3–22. [Google Scholar] [CrossRef]
  116. Temperate Plants Database, Ken Fern. Available online: https://temperate.theferns.info/ (accessed on 26 March 2022).
  117. Kawano, S.; Nagai, Y. Life-history monographs of Japanese plants. 4: Allium victorialis L. ssp. platyphyllum (Makino) Hultén (Alliaceae) Syn. Allium victorialis L. var. platyphyllum Makino; A. latissimum Prokh. Plant Species Biol. 2005, 20, 219–225. [Google Scholar] [CrossRef]
  118. Kitamura, S.; Murata, G.; Hori, M. Coloured Illustrations of Herbaceous Plants of Japan; Hoikusha Publishing Co.: Osaka, Japan, 1958. [Google Scholar]
  119. Arifin, N.S.; Okudo, H. Geographical distribution of allozyme patterns in shallot (Allium cepa var. ascalonicum Backer) and wakegi onion (A. × wakegi Araki). Euphytica 1996, 91, 305–313. [Google Scholar] [CrossRef]
  120. Bah, A.A.; Wang, F.; Huang, Z.; Shamsi, I.H.; Zhang, Q.; Jilani, G.; Hussain, S.; Hussain, N.; Ali, E. Biology Phyto-characteristics, Cultivation and Medicinal Prospects of Chinese Jiaotou (Allium chinense). Int. J. Agric. Biol. 2012, 14, 650–657. [Google Scholar]
  121. Blattner, F.R.; Friesen, N. Relationship between Chinese Chive (Allium tuberosum) and Its Putative Progenitor A. Ramosum as Assessed by Random Amplified Polymorphic DNA (RAPD); California University Press: Los Angeles, CA, USA, 2006; pp. 134–142. [Google Scholar]
  122. Pandey, A.; Pradheep, K.; Gaikwad, A.B.; Gupta, R.; Malav, P.K.; Rai, M. Systematics study on a morphotype of Allium tuberosum Rottler ex Spreng. (Alliaceae) from Ladakh, India. Indian J. Plant Genet. Resour. 2019, 32, 223–231. [Google Scholar] [CrossRef]
  123. Sajad, M.A.; Khan, M.S.; Bahadur, S.; Naeem, A.; Ali, H.; Batool, F.; Shuaib, M.; Khan, M.A.S.; Batool, S. Evaluation of chromium phytoremediation potential of some plant species of Dir Lower, Khyber Pakhtunkhwa, Pakistan. Acta Ecol. Sin. 2020, 40, 158–165. [Google Scholar] [CrossRef]
  124. Keusgen, M.; Fritsch, R.M.; Hisoriev, H.; Kurbonova, P.A.; Khassanov, F.O. Wild Allium species (Alliaceae) used in folk medicine of Tajikistan and Uzbekistan. J. Ethnobiol. Ethnomed. 2006, 2, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Ijaz, F.; Iqbal, Z.; Rahman, I.U.; Alam, J.; Khan, S.M.; Shah, G.M.; Khan, K.; Afzal, A. Investigation of traditional medicinal floral knowledge of Sarban Hills, Abbottabad, KP, Pakistan. J. Ethnopharmacol. 2016, 179, 208–233. [Google Scholar] [CrossRef]
  126. Ajaib, M.; Ishtiaq, M.; Bhatti, K.H.; Hussain, I.; Maqbool, M.; Hussain, T.; Mushtaq, W.; Ghani, A.; Azeem, M.; Khan, S.M.R.; et al. Inventorization of traditional ethnobotanical uses of wild plants of Dawarian and Ratti Gali areas of District Neelum, Azad Jammu and Kashmir Pakistan. PLoS ONE 2021, 16, e0255010. [Google Scholar] [CrossRef]
  127. Islam, M.; Inamullah, A.I.; Akhtar, N.; Alam, J.; Razzaq, A.; Mohammad, K.; Mahmood, T.; Khan, F.U.; Muhammad Khan, W.; Ishtiaq, A.; et al. Medicinal plants resources of Western Himalayan Palas Valley, Indus Kohistan, Pakistan: Their uses and degrees of risk of extinction. Saudi J. Biol. Sci. 2021, 28, 3076–3093. [Google Scholar] [CrossRef] [PubMed]
  128. Amjad, M.S.; Qaeem, M.F.; Ahmad, I.; Khan, S.U.; Chaudhari, S.K.; Malik, N.Z.; Shaheen, H.; Khan, A.M. Descriptive study of plant resources in the context of the ethnomedicinal relevance of indigenous flora: A case study from Toli Peer National Park, Azad Jammu and Kashmir, Pakistan. PLoS ONE 2017, 12, e0171896. [Google Scholar] [CrossRef] [PubMed]
  129. Tavares, L.; Santos, L.; Zapata Noreña, C.P. Bioactive compounds of garlic: A comprehensive review of encapsulation technologies, characterization of the encapsulated garlic compounds and their industrial applicability. Trends Food Sci. Technol. 2021, 114, 232–244. [Google Scholar] [CrossRef]
  130. Zhao, X.X.; Lin, F.J.; Li, H.; Li, H.B.; Wu, D.T.; Geng, F.; Ma, W.; Wang, Y.; Miao, B.H.; Gan, R.Y. Recent Advances in Bioactive Compounds, Health Functions, and Safety Concerns of Onion (Allium cepa L.). Front. Nutr. 2021, 8, 463. [Google Scholar] [CrossRef] [PubMed]
  131. Beretta, H.V.; Bannoud, F.; Insani, M.; Berli, F.; Hirschegger, P.; Galmarini, C.R.; Cavagnaro, P.F. Relationships Between Bioactive Compound Content and the Antiplatelet and Antioxidant Activities of Six Allium Vegetable Species. Food Technol. Biotechnol. 2017, 55, 266–275. [Google Scholar] [CrossRef]
  132. Majewski , M. Allium sativum: Facts and myths regarding human health. Rocz Panstw Zakl Hig. 2014, 65, 1–8. [Google Scholar] [PubMed]
  133. Marrelli, M.; Amodeo, V.; Statti, G.; Conforti, F. Biological Properties and Bioactive Components of Allium cepa L.: Focus on Potential Benefits in the Treatment of Obesity and Related Comorbidities. Molecules 2019, 24, 119. [Google Scholar] [CrossRef] [Green Version]
  134. Teshika, J.D.; Zakariyyah, A.M.; Zaynab, T.; Zengin, G.; Rengasamy, K.R.; Pandian, S.K.; Fawzi, M.M. Traditional and modern uses of onion bulb (Allium cepa L.): A systematic review. Crit. Rev. Food Sci. Nutr. 2019, 59, S39–S70. [Google Scholar] [CrossRef]
  135. Turati, F.; Pelucchi, C.; Guercio, V.; La Vecchia, C.; Galeone, C. Allium vegetable intake and gastric cancer: A case-control study and meta-analysis. Mol. Nutr. Food Res. 2015, 59, 171–179. [Google Scholar] [CrossRef]
  136. Zhou, X.F.; Ding, Z.S.; Liu, N.B. Allium vegetables and risk of prostate cancer: Evidence from 132,192 subjects. Asian Pac. J. Cancer Prev. 2013, 14, 4131–4134. [Google Scholar] [CrossRef] [Green Version]
  137. Turati, F.; Guercio, V.; Pelucchi, C.; La Vecchia, C.; Galeone, C. Colorectal cancer and adenomatous polyps in relation to allium vegetables intake: A meta-analysis of observational studies. Mol. Nutr. Food Res. 2014, 58, 1907–1914. [Google Scholar] [CrossRef] [PubMed]
  138. Guercio, V.; Turati, F.; La Vecchia, C.; Galeone, C.; Tavani, A. Allium vegetables and upper aerodigestive tract cancers: A meta-analysis of observational studies. Mol. Nutr. Food Res. 2016, 60, 212–222. [Google Scholar] [CrossRef] [PubMed]
  139. Kuete, V. Moringa oleifera, in Medicinal Spices and Vegetables from Africa. Med. Spices Veg. Afr. 2017, 43, 605–610. [Google Scholar]
  140. Peltola, R. Allium victoralis. Available online: https://portal.mtt.fi/portal/page/portal/mtt/hankkeet/BARENTSPEC (accessed on 20 March 2022).
  141. Shafakatullah, N.; Chandra, M. Isolation of lactic acid bacteria from Allium cepa var. aggregatum and study of their probiotic properties. Int. J. Pharma Sci. Res. 2015, 6, 749–752. [Google Scholar]
  142. Lawless, J.W.; Latham, M.C.; Stephenson, L.S.; Kinoti, S.N.; Pertet, A.M. Iron supplementation improves appetite and growth in anemic Kenyan primary school children. J. Nutr. 1994, 124, 645–654. [Google Scholar] [CrossRef]
  143. Lee, Y.M.; Lim, D.Y.; Choi, H.J.; Jung, J.I.; Chung, W.Y.; Park, J.H.Y. Induction of cell cycle arrest in prostate cancer cells by the dietary compound isoliquiritigenin. J. Med. Food 2009, 12, 8–14. [Google Scholar] [CrossRef]
  144. Kaiser, P.; Youssouf, M.S.; Tasduq, S.A.; Singh, S.; Sharma, S.C.; Singh, G.D.; Gupta, V.K.; Gupta, B.D.; Johri, R.K. Anti-allergic effects of herbal product from Allium cepa (bulb). J. Med. Food 2009, 12, 374–382. [Google Scholar] [CrossRef]
  145. Mohammadi-Motlagh, H.R.; Mostafaie, A.; Mansouri, K. Anticancer and anti-inflammatory activities of shallot (Allium ascalonicum) extract. Arch. Med. Sci. 2011, 7, 38–44. [Google Scholar] [CrossRef]
  146. Handbook of Chinese Herbs and Formulas; Yeung, H. (Ed.) Institute of Chinese Medicine: London, UK, 1985; Volume 1. [Google Scholar]
  147. Jannat, K.; Rahman, T.; Rahmatullah, M. Traditional uses, phytochemicals and pharmacological properties of Allium tuberosum Rottler ex spreng. J. Med. Plants Stud. 2019, 7, 214–220. [Google Scholar]
  148. Sabha, D.; Hiyasat, B.; Grtzinger, K.; Hennig, L.; Schlegel, F.; Mohr, F.W.; Rauwald, H.W.; Dhein, S. Allium ursinum L.: Bioassay-guided isolation and identification of a galactolipid and a phytosterol exerting antiaggregatory effects. Pharmacology 2012, 89, 260–269. [Google Scholar] [CrossRef]
  149. Carotenuto, A.; De Feo, V.; Fattorusso, E.; Lanzotti, V.; Magno, S.; Cicala, C. The flavonoids of Allium ursinum. Phytochemistry 1996, 41, 531–536. [Google Scholar] [CrossRef]
  150. Sobolewska, D.; Podolak, I.; Makowska-Wąs, J. Allium ursinum: Botanical, phytochemical and pharmacological overview. Phytochem. Rev. 2015, 14, 81–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Ivanova, A.; Mikhova, B.; Najdenski, H.; Tsvetkova, I.; Kostova, I. Chemical composition and antimicrobial activity of wild garlic Allium ursinum of Bulgarian origin. Nat. Prod. Commun. 2009, 4, 1059–1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Dong, Y.; Ruan, J.; Ding, Z.; Zhao, W.; Hao, M.; Zhang, Y.; Jiang, H.; Zhang, Y.; Wang, T. molecules Phytochemistry and Comprehensive Chemical Profiling Study of Flavonoids and Phenolic Acids in the Aerial Parts of Allium Mongolicum Regel and Their Intestinal Motility Evaluation. Molecules 2020, 25, 577. [Google Scholar] [CrossRef] [Green Version]
  153. Marefati, N.; Ghorani, V.; Shakeri, F.; Boskabady, M.; Kianian, F.; Rezaee, R.; Boskabady, M.H. A review of anti-inflammatory, antioxidant, and immunomodulatory effects of Allium cepa and its main constituents. Pharm. Biol. 2021, 59, 287–302. [Google Scholar] [CrossRef]
  154. Fossen, T.; Slimestad, R.; Andersen, Ø.M. Anthocyanins with 4′-glucosidation from red onion, Allium cepa. Phytochemistry 2003, 64, 1367–1374. [Google Scholar] [CrossRef]
  155. Xiao, H.; Parkin, K.L. Isolation and identification of potential cancer chemopreventive agents from methanolic extracts of green onion (Allium cepa). Phytochemistry 2007, 68, 1059–1067. [Google Scholar] [CrossRef]
  156. Fossen, T.; Andersen, Ø.M. Anthocyanins from red onion, Allium cepa, with novel aglycone. Phytochemistry 2003, 62, 1217–1220. [Google Scholar] [CrossRef]
  157. Lanzotti, V.; Romano, A.; Lanzuise, S.; Bonanomi, G.; Scala, F. Antifungal saponins from bulbs of white onion, Allium cepa L. Phytochemistry 2012, 74, 133–139. [Google Scholar] [CrossRef]
  158. Nile, A.; Nile, S.H.; Cespedes-Acuña, C.L.; Oh, J.W. Spiraeoside extracted from red onion skin ameliorates apoptosis and exerts potent antitumor, antioxidant and enzyme inhibitory effects. Food Chem. Toxicol. 2021, 154, 112327. [Google Scholar] [CrossRef]
  159. Nohara, T.; Fujiwara, Y.; El-Aasr, M.; Ikeda, T.; Ono, M.; Nakano, D.; Kinjo, J. Thiolane-type sulfides from garlic, onion, and Welsh onion. J. Nat. Med. 2021, 75, 741–751. [Google Scholar] [CrossRef] [PubMed]
  160. El-Aasr, M.; Fujiwara, Y.; Takeya, M.; Ikeda, T.; Tsukamoto, S.; Ono, M.; Nakano, D.; Okawa, M.; Kinjo, J.; Yoshimitsu, H.; et al. Onionin A from Allium cepa inhibits macrophage activation. J. Nat. Prod. 2010, 73, 1306–1308. [Google Scholar] [CrossRef] [PubMed]
  161. Terahara, N.; Yamaguchi, M.; Honda, T. Malonylated anthocyanins from bulbs of red onion, Allium cepa L. Biosci. Biotechnol. Biochem. 1994, 58, 1324–1325. [Google Scholar] [CrossRef]
  162. Pontin, M.; Bottini, R.; Burba, J.L.; Piccoli, P. Allium sativum produces terpenes with fungistatic properties in response to infection with Sclerotium cepivorum. Phytochemistry 2015, 115, 152–160. [Google Scholar] [CrossRef]
  163. Lanzotti, V.; Barile, E.; Antignani, V.; Bonanomi, G.; Scala, F. Antifungal saponins from bulbs of garlic, Allium sativum L. var. Voghiera. Phytochemistry 2012, 78, 126–134. [Google Scholar] [CrossRef] [PubMed]
  164. Okuyama, T.; Fujita, K.; Shibata, S.; Hoson, M.; Kawada, T.; Masaki, M.; Yamate, N. Effects of Chinese drugs “xiebai” and “dasuan” on human platelet aggregation (Allium bakeri, A. sativum). Planta Med. 1989, 55, 242–244. [Google Scholar] [CrossRef]
  165. Timité, G.; Mitaine-Offer, A.C.; Miyamoto, T.; Tanaka, C.; Mirjolet, J.F.; Duchamp, O.; Lacaille-Dubois, M.A. Structure and cytotoxicity of steroidal glycosides from Allium schoenoprasum. Phytochemistry 2013, 88, 61–66. [Google Scholar] [CrossRef]
  166. Fossen, T.; Slimestad, R.; Øvstedal, D.O.; Andersen, Ø.M. Covalent anthocyanin-flavonol complexes from flowers of chive, Allium schoenoprasum. Phytochemistry 2000, 54, 317–323. [Google Scholar] [CrossRef]
  167. Barile, E.; Bonanomi, G.; Antignani, V.; Zolfaghari, B.; Sajjadi, S.E.; Scala, F.; Lanzotti, V. Saponins from Allium minutiflorum with antifungal activity. Phytochemistry 2007, 68, 596–603. [Google Scholar] [CrossRef]
  168. Carotenuto, A.; Fattorusso, E.; Lanzotti, V.; Magno, S.; De Feo, V.; Cicala, C. The flavonoids of Allium neapolitanum. Phytochemistry 1997, 44, 949–957. [Google Scholar] [CrossRef]
  169. Chehri, Z.; Zolfaghari, B.; Sadeghi Dinani, M. Isolation of Cinnamic Acid Derivatives from the Bulbs of Allium tripedale. Adv. Biomed. Res. 2018, 7, 60. [Google Scholar] [CrossRef]
  170. Jan, K.; Michael, K. Cysteine sulfoxides and volatile sulfur compounds from Allium tripedale. J. Agric. Food Chem. 2010, 58, 1129–1137. [Google Scholar] [CrossRef]
  171. Fattorusso, E.; Lanzotti, V.; Taglialatela-Scafati, O.; Cicala, C. The flavonoids of leek, Allium porrum. Phytochemistry 2001, 57, 565–569. [Google Scholar] [CrossRef]
  172. Carotenuto, A.; Fattorusso, E.; Lanzotti, V.; Magno, S. Spirostanol saponins of Allium porrum L. Phytochemistry 1999, 51, 1077–1082. [Google Scholar] [CrossRef]
  173. Peng, J.P.; Yao, X.S.; Tezuka, Y.; Kikuchi, T. Furostanol glycosides from bulbs of Allium chinense. Phytochemistry 1996, 41, 283–285. [Google Scholar] [CrossRef]
  174. Kuroda, M.; Mimaki, Y.; Kameyama, A.; Sashida, Y.; Nikaido, T. Steroidal saponins from Allium chinense and their inhibitory activities on cyclic AMP phosphodiesterase and Na+/K+ ATPase. Phytochemistry 1995, 40, 1071–1076. [Google Scholar] [CrossRef]
  175. Baba, M.; Ohmura, M.; Kishi, N.; Okada, Y.; Shibata, S.; Peng, J.; Yao, S.S.; Nishino, H.; Okuyama, T. Saponins isolated from Allium chinense G. Don and antitumor-promoting activities of isoliquiritigenin and laxogenin from the same drug. Biol. Pharm. Bull. 2000, 23, 660–662. [Google Scholar] [CrossRef] [Green Version]
  176. Peng, J.; Yao, X.; Kobayashi, H.; Ma, C. Novel furostanol glycosides from Allium macrostemon. Planta Med. 1995, 61, 58–61. [Google Scholar] [CrossRef]
  177. Peng, J.P.; Wu, Y.; Yao, X.S.; Okuyama, T.; Narui, T. Two new steroidal saponins from Allium macrostemon. Yao Xue Xue Bao 1992, 27, 918–922. [Google Scholar] [PubMed]
  178. Peng, J.P.; Wang, X.; Yao, X.S. Studies on two new furostanol glycosides from Allium macrostemon Bunge. Yao Xue Xue Bao 1993, 28, 526–531. [Google Scholar]
  179. Usui, A.; Matsuo, Y.; Tanaka, T.; Ohshima, K.; Fukuda, S.; Mine, T.; Yakashiro, I.; Ishimaru, K. Ferulic acid esters of glucosylglucose from Allium macrostemon Bunge. J. Asian Nat. Prod. Res. 2017, 19, 215–221. [Google Scholar] [CrossRef]
  180. Kawashima, K.; Mimaki, Y.; Sashida, Y. Steroidal saponins from the bulbs of Allium schubertii. Phytochemistry 1993, 32, 1267–1272. [Google Scholar] [CrossRef]
  181. Zou, Z.M.; Yu, D.Q.; Cong, P.Z. A steroidal saponin from the seeds of Allium tuberosum. Phytochemistry 2001, 57, 1219–1222. [Google Scholar] [CrossRef]
  182. Sang, S.; Zou, M.; Xia, Z.; Lao, A.; Chen, Z.; Ho, C.T. New spirostanol saponins from Chinese chives (Allium tuberosum). J. Agric. Food Chem. 2001, 49, 4780–4783. [Google Scholar] [CrossRef] [PubMed]
  183. Sang, S.M.; Zou, M.L.; Zhang, X.W.; Lao, A.N.; Chen, Z.L. Tuberoside M, a new cytotoxic spirostanol saponin from the seeds of Allium tuberosum. J. Asian Nat. Prod. Res. 2002, 4, 67–70. [Google Scholar] [CrossRef] [PubMed]
  184. Mimaki, Y.; Kawashima, K.; Kanmoto, T.; Sashida, Y. Steroidal glycosides from Allium albopilosum and A. ostrowskianum. Phytochemistry 1993, 34, 799–805. [Google Scholar] [CrossRef]
  185. Zolfaghari, B.; Yazdiniapour, Z.; Sadeghi, M.; Akbari, M.; Troiano, R.; Lanzotti, V. Cinnamic acid derivatives from welsh onion (Allium fistulosum) and their antibacterial and cytotoxic activities. Phytochem. Anal. 2021, 32, 84–90. [Google Scholar] [CrossRef]
  186. Sang, S.; Lao, A.; Wang, Y.; Chin, C.K.; Rosen, R.T.; Ho, C.T. Antifungal constituents from the seeds of Allium fistulosum L. J. Agric. Food Chem. 2002, 50, 6318–6321. [Google Scholar] [CrossRef]
  187. Tsuruoka, T.; Ishikawa, K.; Hosoe, T.; Davaajab, D.; Duvjir, S.; Surenjav, U. A new cinnamoylphenethylamine derivative from a Mongolian Allium species, Allium carolinianum. J. Nat. Med. 2018, 72, 332–334. [Google Scholar] [CrossRef]
  188. Zamri, N.; Hamid, H.A. Comparative Study of Onion (Allium cepa) and Leek (Allium ampeloprasum): Identification of Organosulphur Compounds by UPLC-QTOF/MS and Anticancer Effect on MCF-7 Cells. Plant Foods Hum. Nutr. 2019, 74, 525–530. [Google Scholar] [CrossRef]
  189. Kang, L.-P.; Liu, Z.-J.; Zhang, L.; Tan, D.-W.; Zhao, Y.; Zhao, Y.; Chen, H.-B.; Ma, B.-P. New furostanol saponins from Allium ascalonicum L. Magn. Reson. Chem. Magn. Reson. Chem 2007, 45, 725–733. [Google Scholar] [CrossRef] [PubMed]
  190. Kubec, R.; Cody, R.B.; Dane, A.J.; Musah, R.A.; Schraml, J.; Vattekkatte, A.; Block, E. Applications of direct analysis in real time-mass spectrometry (DART-MS) in Allium chemistry. (Z)-butanethial S-oxide and 1-butenyl thiosulfinates and their S-(E)-1-butenylcysteine S-oxide precursor from Allium siculum. J. Agric. Food Chem. 2010, 58, 1121–1128. [Google Scholar] [CrossRef]
  191. Hu, X.-P.; Zhou, H.; Du, Y.-M.; Ou, S.-Y.; Yan, R.; Wang, Y. Two new flavonoids from the bark of Allium chrysanthum. J. Asian Nat. Prod. Res. 2017, 19, 229–234. [Google Scholar] [CrossRef] [PubMed]
  192. Kusterer, J.; Vogt, A.; Keusgen, M. Isolation and identification of a new cysteine sulfoxide and volatile sulfur compounds from Allium subgenus Melanocrommyum. J. Agric. Food Chem. 2010, 58, 520–526. [Google Scholar] [CrossRef] [PubMed]
  193. Morita, T.; Ushiroguchi, T.; Hayashi, N.; Matsuura, H.; Itakura, Y.; Fuwa, T. Steroidal saponins from elephant garlic, bulbs of Allium ampeloprasum L. Chem. Pharm. Bull. 1988, 36, 3480–3486. [Google Scholar] [CrossRef] [Green Version]
  194. Lee, K.T.; Choi, J.H.; Kim, D.H.; Son, K.H.; Kim, W.B.; Kwon, S.H.; Park, H.J. Constituents and the antitumor principle of Allium victorialis var. platyphyllum. Arch. Pharm. Res. 2001, 24, 44–50. [Google Scholar] [CrossRef]
  195. Akhov, L.S.; Musienko, M.M.; Piacente, S.; Pizza, C.; Oleszek, W. Structure of steroidal saponins from underground parts of Allium nutans L. J. Agric. Food Chem. 1999, 47, 3193–3196. [Google Scholar] [CrossRef]
  196. Mimaki, Y.; Matsumoto, K.; Sashida, Y.; Nikaido, T.; Ohmoto, T. New steroidal saponins from the bulbs of Allium giganteum exhibiting potent inhibition of cAMP phosphodiesterase activity. Chem. Pharm. Bull. 1994, 42, 710–714. [Google Scholar] [CrossRef] [Green Version]
  197. Ren, L.; Yaun-Fei, W.; Qian, S.; Hua-Bin, H. Chemical composition and antimicrobial activity of the essential oil from Allium hookeri consumed in Xishuangbanna, Southwest China. Nat. Prod. Commun. 2014, 9, 863–864. [Google Scholar]
  198. El-Saber Batiha, G.; Magdy Beshbishy, A.; Wasef, L.G.; Elewa, Y.H.A.; Al-Sagan, A.; Abd El-Hack, M.E.; Taha, A.E.; Abd-Elhakim, M.Y.; Prasad Devkota, H. Chemical Constituents and Pharmacological Activities of Garlic (Allium sativum L.): A Review. Nutrients 2020, 12, 872. [Google Scholar] [CrossRef] [Green Version]
  199. Kuda, T.; Iwai, A.; Yano, T. Effect of red pepper Capsicum annuum var. conoides and garlic Allium sativum on plasma lipid levels and cecal microflora in mice fed beef tallow. Food Chem. Toxicol. 2004, 42, 1695–1700. [Google Scholar] [CrossRef] [PubMed]
  200. Wallock-Richards, D.; Doherty, C.J.; Doherty, L.; Clarke, D.J.; Place, M.; Govan, J.R.W.; Campopiano, D.J. Garlic revisited: Antimicrobial activity of allicin-containing garlic extracts against Burkholderia cepacia complex. PLoS ONE 2014, 9, e112726. [Google Scholar] [CrossRef] [PubMed]
  201. Ross, Z.M.; O’Gara, E.A.; Hill, D.J.; Sleightholme, H.V.; Maslin, D.J. Antimicrobial properties of garlic oil against human enteric bacteria: Evaluation of methodologies and comparisons with garlic oil sulfides and garlic powder. Appl. Environ. Microbiol. 2001, 67, 475–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Pârvu, M.; Moţ, C.A.; Pârvu, A.E.; Mircea, C.; Stoeber, L.; Roşca-Casian, O.; Ţigu, A.B. Allium sativum Extract Chemical Composition, Antioxidant Activity and Antifungal Effect against Meyerozyma guilliermondii and Rhodotorula mucilaginosa Causing Onychomycosis. Molecules 2019, 24, 3958. [Google Scholar] [CrossRef] [Green Version]
  203. Fufa, B.K. Anti-bacterial and Anti-fungal Properties of Garlic Extract (Allium sativum): A Review. Microbiol. Res. J. Int. 2019, 28, 1–5. [Google Scholar] [CrossRef]
  204. Zhen, H.; Fang, F.; Ye, D.; Shu, S.; Zhou, Y.; Dong, Y.; Nie, X.; Li, G. Experimental study on the action of allitridin against human cytomegalovirus in vitro: Inhibitory effects on immediate-early genes. Antivir. Res. 2006, 72, 68–74. [Google Scholar] [CrossRef]
  205. Danquah, C.A.; Tetteh, M.; Amponsah, I.K.; Mensah, A.Y.; Buabeng, K.O.; Gibbons, S.; Bhakta, S. Investigating ghanaian Allium species for anti-infective and resistance-reversal natural product leads to mitigate multidrug-resistance in tuberculosis. Antibiotics 2021, 10, 902. [Google Scholar] [CrossRef]
  206. Satvati, S.A.R.; Shooriabi, M.; Amin, M.; Shiezadeh, F. Evaluation of the Antimicrobial Activity of Tribulus terrestris, Allium sativum, Salvia officinalis, and Allium hirtifolium Boiss Against Enterococcus faecalis. Int. J. Enteric. Pathog. 2017, 5, 63–67. [Google Scholar] [CrossRef] [Green Version]
  207. Abdel-Hafeez, E.H.; Ahmad, A.K.; Kamal, A.M.; Abdellatif, M.Z.M.; Abdelgelil, N.H. In vivo antiprotozoan effects of garlic (Allium sativum) and ginger (Zingiber officinale) extracts on experimentally infected mice with Blastocystis spp. Parasitol. Res. 2015, 114, 3439–3444. [Google Scholar] [CrossRef]
  208. Gruhlke, M.C.H.; Nicco, C.; Batteux, F.; Slusarenko, A.J. The Effects of Allicin—A Reactive Sulfur Species from Garlic, on a Selection of Mammalian Cell Lines. Antioxidants 2016, 6, 1. [Google Scholar] [CrossRef] [Green Version]
  209. Danquah, C.A.; Kakagianni, E.; Khondkar, P.; Maitra, A.; Rahman, M.; Evangelopoulos, D.; McHugh, T.D.; Stapleton, P.; Malkinson, J.; Bhakta, S.; et al. Analogues of Disulfides from Allium stipitatum Demonstrate Potent Anti-tubercular Activities through Drug Efflux Pump and Biofilm Inhibition. Sci. Rep. 2018, 8, 1150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Asdaq, S.M.B.; Inamdar, M.N. Pharmacodynamic and Pharmacokinetic Interactions of Propranolol with Garlic (Allium sativum) in Rats. Evid. Based. Complement. Alternat. Med. 2011, 2011, 824042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Jang, H.-J.; Lee, H.-J.; Yoon, D.-K.; Ji, D.-S.; Kim, J.-H.; Lee, C.-H. Antioxidant and antimicrobial activities of fresh garlic and aged garlic by-products extracted with different solvents. Food Sci. Biotechnol. 2018, 27, 219–225. [Google Scholar] [CrossRef] [PubMed]
  212. Abdel-Daim, M.M.; Shaheen, H.M.; Abushouk, A.I.; Toraih, E.A.; Fawzy, M.S.; Alansari, W.S.; Aleya, L.; Bungau, S. Thymoquinone and diallyl sulfide protect against fipronil-induced oxidative injury in rats. Environ. Sci. Pollut. Res. Int. 2018, 25, 23909–23916. [Google Scholar] [CrossRef]
  213. Putnoky, S.; Caunii, A.; Butnariu, M. Study on the stability and antioxidant effect of the Allium ursinumwatery extract. Chem. Cent. J. 2013, 7, 21. [Google Scholar] [CrossRef] [Green Version]
  214. Asgarpanah, J.; Ghanizadeh, B. Pharmacologic and medicinal properties of Allium hirtifolium Boiss. Afr. J. Pharm. Pharmacol. 2012, 6, 1809–1814. [Google Scholar] [CrossRef]
  215. Ahmad, T.A.; El-Sayed, B.A.; El-Sayed, L.H. Development of immunization trials against Eimeria spp. Trials Vaccinol. 2016, 5, 38–47. [Google Scholar] [CrossRef] [Green Version]
  216. Hobauer, R.; Frass, M.; Gmeiner, B.; Kaye, A.D.; Frost, E.A. Garlic extract (Allium sativum) reduces migration of neutrophils through endothelial cell monolayers. Middle East J. Anaesthesiol. 2000, 15, 649–658. [Google Scholar]
  217. Gu, X.; Wu, H.; Fu, P. Allicin attenuates inflammation and suppresses HLA-B27 protein expression in ankylosing spondylitis mice. Biomed. Res. Int. 2013, 2013, 171573. [Google Scholar] [CrossRef] [Green Version]
  218. Jeong, Y.Y.; Ryu, J.H.; Shin, J.-H.; Kang, M.J.; Kang, J.R.; Han, J.; Kang, D. Comparison of Anti-Oxidant and Anti-Inflammatory Effects between Fresh and Aged Black Garlic Extracts. Molecules 2016, 21, 430. [Google Scholar] [CrossRef] [Green Version]
  219. Jin, P.; Kim, J.-A.; Choi, D.-Y.; Lee, Y.-J.; Jung, H.S.; Hong, J.T. Anti-inflammatory and anti-amyloidogeniceffects of a small molecule, 2,4-bis(p-hydroxyphenyl)-2-butenal in Tg2576 Alzheimer’sdisease mice model. J. Neuroinflammation 2013, 10, 767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Krejčová, P.; Kučerová, P.; Stafford, G.I.; Jäger, A.K.; Kubec, R. Antiinflammatory and neurological activity of pyrithione and related sulfur-containing pyridine N-oxides from Persian shallot (Allium stipitatum). J. Ethnopharmacol. 2014, 154, 176–182. [Google Scholar] [CrossRef] [PubMed]
  221. Karunanidhi, A.; Ghaznavi-Rad, E.; Jeevajothi Nathan, J.; Abba, Y.; van Belkum, A.; Neela, V. Allium stipitatum Extract Exhibits In Vivo Antibacterial Activity against Methicillin-Resistant Staphylococcus aureus and Accelerates Burn Wound Healing in a Full-Thickness Murine Burn Model. Evid. Based Complement. Altern. Med. 2017, 2017, 1914732. [Google Scholar] [CrossRef] [Green Version]
  222. Kim, J.E.; Park, K.M.; Lee, S.Y.; Seo, J.H.; Yoon, I.S.; Bae, C.S.; Yoo, J.C.; Bang, M.A.; Cho, S.S.; Park, D.H. Anti-inflammatory effect of Allium hookeri on carrageenan-induced air pouch mouse model. PLoS ONE 2017, 12, e0190305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Li, Z.; Le, W.; Cui, Z. A novel therapeutic anticancer property of raw garlic extract via injection but not ingestion. Cell Death Discov. 2018, 4, 108. [Google Scholar] [CrossRef] [PubMed]
  224. Chhabria, S.V.; Akbarsha, M.A.; Li, A.P.; Kharkar, P.S.; Desai, K.B. In situ allicin generation using targeted alliinase delivery for inhibition of MIA PaCa-2 cells via epigenetic changes, oxidative stress and cyclin-dependent kinase inhibitor (CDKI) expression. Apoptosis 2015, 20, 1388–1409. [Google Scholar] [CrossRef]
  225. Fleischauer, A.T.; Arab, L. Garlic and cancer: A critical review of the epidemiologic literature. J. Nutr. 2001, 131, 1032S–1040S. [Google Scholar] [CrossRef] [Green Version]
  226. Piscitelli, S.C.; Burstein, A.H.; Welden, N.; Gallicano, K.D.; Falloon, J. The effect of garlic supplements on the pharmacokinetics of saquinavir. Clin. Infect. Dis. 2002, 34, 234–238. [Google Scholar] [CrossRef]
  227. Dall’Acqua, S.; Maggi, F.; Minesso, P.; Salvagno, M.; Papa, F.; Vittori, S.; Innocenti, G. Identification of non-alkaloid acetylcholinesterase inhibitors from Ferulago campestris (Besser) Grecescu (Apiaceae). Fitoterapia 2010, 81, 1208–1212. [Google Scholar] [CrossRef]
  228. Szychowski, K.A.; Rybczyńska-Tkaczyk, K.; Gaweł-Bęben, K.; Świeca, M.; Karaś, M.; Jakubczyk, A.; Matysiak, M.; Binduga, U.E.; Gmiński, J. Characterization of Active Compounds of Different Garlic (Allium sativum L.) Cultivars. Pol. J. Food Nutr. Sci. 2018, 68, 73–81. [Google Scholar] [CrossRef]
  229. Lu, S.-H.; Wu, J.W.; Liu, H.-L.; Zhao, J.-H.; Liu, K.-T.; Chuang, C.-K.; Lin, H.-Y.; Tsai, W.-B.; Ho, Y. The discovery of potential acetylcholinesterase inhibitors: A combination of pharmacophore modeling, virtual screening, and molecular docking studies. J. Biomed. Sci. 2011, 18, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Mathew, B.; Biju, R. Neuroprotective effects of garlic a review. Libyan J. Med. 2008, 3, 23–33. [Google Scholar] [CrossRef] [PubMed]
  231. Qidwai, W.; Ashfaq, T. Role of garlic usage in cardiovascular disease prevention: An evidence-based approach. Evid. Based. Complement. Alternat. Med. 2013, 2013, 125649. [Google Scholar] [CrossRef] [PubMed]
  232. Ashraf, R.; Aamir, K.; Shaikh, A.R.; Ahmed, T. Effects of garlic on dyslipidemia in patients with type 2 diabetes mellitus. J. Ayub Med. Coll. Abbottabad 2005, 17, 60–64. [Google Scholar]
  233. Zhai, B.; Zhang, C.; Sheng, Y.; Zhao, C.; He, X.; Xu, W.; Huang, K.; Luo, Y. Hypoglycemic and hypolipidemic effect of S-allyl-cysteine sulfoxide (alliin) in DIO mice. Sci. Rep. 2018, 8, 3527. [Google Scholar] [CrossRef] [Green Version]
  234. Lee, M.-S.; Kim, I.-H.; Kim, C.-T.; Kim, Y. Reduction of body weight by dietary garlic is associated with an increase in uncoupling protein mRNA expression and activation of AMP-activated protein kinase in diet-induced obese mice. J. Nutr. 2011, 141, 1947–1953. [Google Scholar] [CrossRef]
  235. Sobenin, I.; Andrianova, I.; Ionova, V.; Karagodin, V.; Orekhov, A. Anti-aggregatory and fibrinolytic effects of time-released garlic powder tablets. Med. Heal. Sci. J. 2012, 10, 47–51. [Google Scholar] [CrossRef]
  236. Embuscado, M.E. Bioactives from culinary spices and herbs: A review. J. Food Bioact. 2019, 6. [Google Scholar] [CrossRef]
  237. Mohammadi-rika, A.; Beigi-boroujeni, M.; Rajabzadeh, A.; Zarei, L. Effect of Extract of Allium stipitatum on Excisional Wound Healing in Rats. Iran. J. Vet. Surg. 2021, 16, 5–11. [Google Scholar]
  238. Velten, R.; Erdelen, C.; Gehling, M.; Ghrt, A.; Gondol, D.; Lenz, J.; Loekhoff, O.; Wachendorff, U.; Wendisch, D.; Cripowellin, A. Cripowellin A and B, Novel Type of Amaryllidaceae Alkaloid from Crinum powellii. Tetrahedron Lett. 1998, 39, 1737–1740. [Google Scholar] [CrossRef]
  239. Zvetkova, E.; Wirleitner, B.; Tram, N.T.; Schennach, H.; Fuchs, D. Aqueous extracts of Crinum latifolium and Camellia sinensis show immunomodulatory properties in human peripheral blood mononuclear cells. Sci. Pharm. 2001, 79, 2143–2150. [Google Scholar] [CrossRef]
  240. Ulrich, M.R.; Davies, F.T., Jr.; Koh, Y.C.; Duray, S.A.; Egilla, J.N. Micropropagation of Crinum `Ellen Bosanquet’ by tri-scales. Sci. Hortic. 1999, 82, 95–102. [Google Scholar] [CrossRef]
  241. Thi Ngoc Tram, N.; Titorenkova, T.V.; Bankova, V.S.; Handjieva, N.V.; Popov, S.S. Crinum L. (Amaryllidaceae). Fitoterapia 2002, 73, 183–208. [Google Scholar] [CrossRef]
  242. Singh, K.A.; Nayak, M.K.; Jagannadham, M.V.; Dash, D. Thrombolytic along with anti-platelet activity of crinumin, a protein constituent of Crinum asiaticum. Blood Cells. Mol. Dis. 2011, 47, 129–132. [Google Scholar] [CrossRef]
  243. Ghosal, S.; Rao, P.H.; Jaiswal, D.K.; Kumar, Y.; Frahm, A.W. Alkaloids of Crinum pratense. Phytochemistry 2007, 20, 2003–2007. [Google Scholar] [CrossRef]
  244. Yoshisuke, T.; Noriaki, K.; Vijaya, K. The Alkaloidal Constituents of Goda-Manel (Crinum zeylanicum L.), a Sri Lankan Folk Medicine. Chem. Pharm. Bull. 1984, 32, 3023–3027. [Google Scholar]
  245. Nordal, I.; Wahlstrom, R. Studies in the Crinum zeylanicum complex in East Africa. Nord. J. Bot. 1982, 2, 465–473. [Google Scholar] [CrossRef]
  246. Bastida, J.; Peeters, P.; Rubiralta, M.; Naturals, D.D.P.; De Farmficia, F.; Barcelona, U. De Alkaloids from crinum kirkii. Phytochemistry 1995, 40, 1291–1293. [Google Scholar] [CrossRef]
  247. Nair, J.J.; Machocho, A.K.; Campbell, W.E.; Brun, R.; Viladomat, F.; Codina, C.; Bastida, J. Alkaloids from Crinum macowanii. Phytochemistry 2000, 54, 5. [Google Scholar] [CrossRef]
  248. Elgorashi, E.E.; Drewes, S.E.; Staden, J. Van Alkaloids from Crinum bulbispermum. Phytochemistry 1999, 52, 533–536. [Google Scholar] [CrossRef]
  249. Tallini, L.R.; Carrasco, A.; Acosta León, K.; Vinueza, D.; Bastida, J.; Oleas, N.H. Alkaloid Profiling and Cholinesterase Inhibitory Potential of Crinum × amabile Donn. (Amaryllidaceae) Collected in Ecuador. Plants 2021, 10, 2686. [Google Scholar] [CrossRef] [PubMed]
  250. Tallini, L.R.; Torras-Claveria, L.; de Borges, W.S.; Kaiser, M.; Viladomat, F.; Zuanazzi, J.A.S.; Bastida, J. N-oxide alkaloids from Crinum amabile (Amaryllidaceae). Molecules 2018, 23, 1277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Panthong, K.; Ingkaninan, K. Amabiloid A from Crinum × amabile Donn ex Ker Gawl. Nat. Prod. Res. 2021, 35, 3220–3225. [Google Scholar] [CrossRef] [PubMed]
  252. Bordoloi, M.; Kotoky, R.; Mahanta, J.J.; Sarma, T.C.; Kanjilal, P.B. Anti-genotoxic hydrazide from Crinum defixum. Eur. J. Med. Chem. 2009, 44, 2754–2757. [Google Scholar] [CrossRef]
  253. Fennell, C.W.; Elgorashi, E.E.; Van Staden, J. Alkaloid production in Crinum moorei cultures. J. Nat. Prod. 2003, 66, 1524–1526. [Google Scholar] [CrossRef]
  254. Masi, M.; Koirala, M.; Delicato, A.; Di Lecce, R.; Merindol, N.; Ka, S.; Seck, M.; Tuzi, A.; Desgagne-Penix, I.; Calabrò, V.; et al. Isolation and Biological Characterization of Homoisoflavanoids and the Alkylamide N-p-Coumaroyltyramine from Crinum biflorum Rottb., an Amaryllidaceae Species Collected in Senegal. Biomolecules 2021, 11, 1298. [Google Scholar] [CrossRef]
  255. Elgorashi, E.; Drewes, S.E.; Van Staden, J. Alkaloids from Crinum moorei. Phytochemistry 2001, 56, 637–640. [Google Scholar] [CrossRef]
  256. Yu, M.; Wang, B.; Qi, Z.; Xin, G.; Li, W. Response surface method was used to optimize the ultrasonic assisted extraction of flavonoids from Crinum asiaticum. Saudi J. Biol. Sci. 2019, 26, 2079–2084. [Google Scholar] [CrossRef]
  257. Khumkhrong, P.; Piboonprai, K.; Chaichompoo, W.; Pimtong, W.; Khongkow, M.; Namdee, K.; Jantimaporn, A.; Japrung, D.; Asawapirom, U.; Suksamrarn, A.; et al. Crinamine Induces Apoptosis and Inhibits Proliferation, Migration, and Angiogenesis in Cervical Cancer SiHa Cells. Biomolecules 2019, 9, 494. [Google Scholar] [CrossRef] [Green Version]
  258. Sun, Q.; Shen, Y.H.; Tian, J.M.; Tang, J.; Su, J.; Liu, R.H.; Li, H.L.; Xu, X.K.; Zhang, W.D. Chemical constituents of Crinum asiaticum L. var. sinicum Baker and their cytotoxic activities. Chem. Biodivers. 2009, 6, 1751–1757. [Google Scholar] [CrossRef]
  259. Kim, S.C.; Kang, J.; Kim, M.K.; Hyun, J.H.; Boo, H.J.; Park, D.B.; Lee, Y.J.; Yoo, E.S.; Kim, Y.H.; Kim, Y.H.; et al. Promotion effect of norgalanthamine, a component of Crinum asiaticum, on hair growth. Eur. J. Dermatol. 2010, 20, 42–48. [Google Scholar] [CrossRef]
  260. Kogure, N.; Katsuta, N.; Kitajima, M.; Takayama, H. Two new alkaloids from Crinum asiaticum var. sinicum. Chem. Pharm. Bull. 2011, 59, 1545–1548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  261. Do, K.M.; Shin, M.K.; Kodama, T.; Win, N.N.; Prema, P.; Nguyen, H.M.; Hayakawa, Y.; Morita, H. Flavanols and Flavanes from Crinum asiaticum and their Effects on LPS Signaling Pathway through the Inhibition of NF-κB Activation. Planta Med. 2021. [Google Scholar] [CrossRef] [PubMed]
  262. Yu, M.; Chen, Y.; Liu, Y.; Xu, Y.; Wang, B. Efficient polysaccharides from Crinum asiaticum L.’s structural characterization and anti-tumor effect. Saudi J. Biol. Sci. 2019, 26, 2085–2090. [Google Scholar] [CrossRef] [PubMed]
  263. Endo, Y.; Sugiura, Y.; Funasaki, M.; Kagechika, H.; Ishibashi, M.; Ohsaki, A. Two new alkaloids from Crinum asiaticum var. japonicum. J. Nat. Med. 2019, 73, 648–652. [Google Scholar] [CrossRef]
  264. Min, B.S.; Gao, J.J.; Nakamura, N.; Kim, Y.H.; Hattori, M. Cytotoxic alkaloids and a flavan from the bulbs of Crinum asiaticum var. japonicum. Chem. Pharm. Bull. 2001, 49, 1217–1219. [Google Scholar] [CrossRef] [Green Version]
  265. Kim, Y.H.; Park, E.J.; Park, M.H.; Badarch, U.; Woldemichael, G.M.; Beutler, J.A. Crinamine from Crinum asiaticum var. japonicum inhibits hypoxia inducible factor-1 activity but not activity of hypoxia inducible factor-2. Biol. Pharm. Bull. 2006, 29, 2140–2142. [Google Scholar] [CrossRef] [Green Version]
  266. Machocho, A.K.; Bastida, J.; Codina, C.; Viladomat, F.; Brun, R.; Chhabra, S.C. Augustamine type alkaloids from Crinum kirkii. Phytochemistry 2004, 65, 3143–3149. [Google Scholar] [CrossRef]
  267. Presley, C.C.; Du, Y.; Dalal, S.; Merino, E.F.; Butler, J.H.; Rakotonandrasana, S.; Rasamison, V.E.; Cassera, M.B.; Kingston, D.G.I. Isolation, structure elucidation, and synthesis of antiplasmodial quinolones from Crinum firmifolium. Bioorganic Med. Chem. 2017, 25, 4203–4211. [Google Scholar] [CrossRef]
  268. Chen, M.X.; Huo, J.M.; Hu, J.; Xu, Z.P.; Zhang, X. Amaryllidaceae alkaloids from Crinum latifolium with cytotoxic, antimicrobial, antioxidant, and anti-inflammatory activities. Fitoterapia 2018, 130, 48–53. [Google Scholar] [CrossRef]
  269. Tian, H.; Liu, Q.J.; Wang, J.T.; Zhang, L. Antimicrobial crinane-type alkaloids from the bulbs of Crinum latifolium. J. Asian Nat. Prod. Res. 2021, 23, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  270. Nam, N.H.; Kim, Y.; You, Y.J.; Hong, D.H.; Kim, H.M.; Ahn, B.Z. New constituents from Crinum latifolium with inhibitory effects against tube-like formation of human umbilical venous endothelial cells. Nat. Prod. Res. 2004, 18, 485–491. [Google Scholar] [CrossRef]
  271. N’Tamon, A.D.; Okpekon, A.T.; Bony, N.F.; Bernadat, G.; Gallard, J.F.; Kouamé, T.; Séon-Méniel, B.; Leblanc, K.; Rharrabti, S.; Mouray, E.; et al. Streamlined targeting of Amaryllidaceae alkaloids from the bulbs of Crinum scillifolium using spectrometric and taxonomically-informed scoring metabolite annotations. Phytochemistry 2020, 179, 112485. [Google Scholar] [CrossRef] [PubMed]
  272. Berkov, S.; Romani, S.; Herrera, M.; Viladomat, F.; Codina, C.; Momekov, G.; Ionkova, I.; Bastida, J. Antiproliferative alkaloids from Crinum zeylanicum. Phytother. Res. 2011, 25, 1686–1692. [Google Scholar] [CrossRef] [PubMed]
  273. Ka, S.; Masi, M.; Merindol, N.; Di Lecce, R.; Plourde, M.B.; Seck, M.; Górecki, M.; Pescitelli, G.; Desgagne-Penix, I.; Evidente, A. Gigantelline, gigantellinine and gigancrinine, cherylline- and crinine-type alkaloids isolated from Crinum jagus with anti-acetylcholinesterase activity. Phytochemistry 2020, 175, 112390. [Google Scholar] [CrossRef]
  274. Cortes, N.; Sierra, K.; Alzate, F.; Osorio, E.H.; Osorio, E. Alkaloids of Amaryllidaceae as Inhibitors of Cholinesterases (AChEs and BChEs): An Integrated Bioguided Study. Phytochem. Anal. 2017, 29, 217–227. [Google Scholar] [CrossRef]
  275. Abebe, B.; Tadesse, S.; Hymete, A.; Bisrat, D. Antiproliferative Effects of Alkaloids from the Bulbs of Crinum abyscinicum Hochst. ExA. Rich. Evid. Based. Complement. Alternat. Med. 2020, 2020, 2529730. [Google Scholar] [CrossRef]
  276. Presley, C.C.; Krai, P.; Dalal, S.; Su, Q.; Cassera, M.; Goetz, M.; Kingston, D.G.I. New potently bioactive alkaloids from Crinum erubescens. Bioorganic Med. Chem. 2016, 24, 5418–5422. [Google Scholar] [CrossRef] [Green Version]
  277. Abdel-Halim, O.B.; Marzouk, A.M.; Mothana, R.; Awadh, N. A new tyrosinase inhibitor from Crinum yemense as potential treatment for hyperpigmentation. Pharmazie 2008, 63, 405–407. [Google Scholar] [PubMed]
  278. Abdel-Halim, O.B.; Morikawa, T.; Ando, S.; Matsuda, H.; Yoshikawa, M. New crinine-type alkaloids with inhibitory effect on induction of inducible nitric oxide synthase from Crinum yemense. J. Nat. Prod. 2004, 67, 1119–1124. [Google Scholar] [CrossRef]
  279. Aboul-Ela, M.A.; El-Lakany, A.M.; Hammoda, H.M. Alkaloids from the bulbs of Crinum bulbispermum. Pharmazie 2004, 59, 894–896. [Google Scholar] [CrossRef] [PubMed]
  280. Ramadan, M.A.; Kamel, M.S.; Ohtani, K.; Kasai, R.; Yamasaki, K. Minor phenolics from Crinum bulbispermum bulbs. Phytochemistry 2000, 54, 891–896. [Google Scholar] [CrossRef]
  281. Ali, A.; Ramadan, M.; Frahm, A. Alkaloidal Constituents of Crinum bulbispermum III: Bulbispermine—A New Alkaloid of Crinum bulbispermum. Planta Med. 1984, 50, 424–427. [Google Scholar] [CrossRef] [PubMed]
  282. Kissling, J.; Ioset, J.R.; Marston, A.; Hostettmann, K. Bio-guided isolation of cholinesterase inhibitors from the bulbs of Crinum x powellii. Phytother. Res. 2005, 19, 984–987. [Google Scholar] [CrossRef]
  283. Niño, J.; Hincapié, G.M.; Correa, Y.M.; Mosquera, O.M. Alkaloids of Crinum x powellii “Album” (Amaryllidaceae) and their topoisomerase inhibitory activity. Z. Naturforsch. C 2007, 62, 223–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Houghton, P.J.; Agbedahunsi, J.M.; Adegbulugbe, A. Choline esterase inhibitory properties of alkaloids from two Nigerian Crinum species. Phytochemistry 2004, 65, 2893–2896. [Google Scholar] [CrossRef]
  285. Nkanwen, E.R.S.; Gatsing, D.; Ngamga, D.; Fodouop, S.P.C.; Tane, P. Antibacterial agents from the leaves of Crinum purpurascens herb (Amaryllidaceae). Afr. Health Sci. 2009, 9, 264–269. [Google Scholar] [CrossRef] [PubMed]
  286. Refaat, J.; Kamel, M.S.; Ramadan, M.A.; Ali, A.A. Crinum; An endless source of bioactive principles: A Review. Part V. Biological Profile. Int. J. Pharm. Sci. Res. 2013, 4, 1239–1252. [Google Scholar]
  287. Mahomoodally, M.F.; Sadeer, N.B.; Suroowan, S.; Jugreet, S.; Lobine, D.; Rengasamy, K.R.R. Ethnomedicinal, phytochemistry, toxicity and pharmacological benefits of poison bulb—Crinum asiaticum L. S. Afr. J. Bot. 2021, 136, 16–29. [Google Scholar] [CrossRef]
  288. Kim, Y.H.; Kim, K.H.; Han, C.S.; Park, S.H.; Yang, H.C.; Lee, B.Y.; Eom, S.-Y.; Kim, Y.-S.; Kim, J.-H.; Lee, N.H. Anti-inflammatory activity of Crinum asiaticum Linne var. japonicum extract and its application as a cosmeceutical ingredient. J. Cosmet. Sci. 2008, 59, 419–430. [Google Scholar]
  289. Samud, A.M.; Asmawi, M.Z.; Sharma, J.N.; Yusof, A.P.M. Anti-inflammatory activity of Crinum asiaticum plant and its effect on bradykinin-induced contractions on isolated uterus. Immunopharmacology 1999, 43, 311–316. [Google Scholar] [CrossRef]
  290. Rahman, A.S.M.; Azad, H.; Nazim, U.A. Analgesic and anti-inflammatory effects of Crinum asiaticum leaf alcoholic extract in animal models. Afr. J. Biotechnol. 2013, 12, 212–218. [Google Scholar] [CrossRef] [Green Version]
  291. Minkah, P.A.B.; Danquah, C.A. Anti-infective, anti-inflammatory and antipyretic activities of the bulb extracts of Crinum jagus (J. Thomps.) Dandy (Amaryllidaceae). Sci. Afr. 2021, 12, e00723. [Google Scholar] [CrossRef]
  292. Ratnasooriya, W.D.; Deraniyagala, S.A.; Bathige, S.D.N.K.; Hettiarachchi, H.D.I. Leaf extract of Crinum bulbispermum has antinociceptive activity in rats. J. Ethnopharmacol. 2005, 97, 123–128. [Google Scholar] [CrossRef] [PubMed]
  293. Ahmad, B. Chemical composition and antifungal, phytotoxic, brine shrimp cytotoxicity, insecticidal and antibacterial activities of the essential oils of Acacia modesta. J. Med. Plants Res. 2012, 6, 4653–4659. [Google Scholar] [CrossRef]
  294. Alawode, T.T.; Lajide, L.; Owolabi, B.J.; Olaleye, M.T. Evaluation of Extracts of Leaves of Crinum jagus for Antimicrobial Properties. J. Appl. Sci. Environ. Manag. 2020, 24, 1197–1201. [Google Scholar] [CrossRef]
  295. Nguyen, H.M.; Nguyen, N.Y.T.; Chau, N.T.N.; Nguyen, A.B.T.; Tran, V.K.T.; Hoang, V.; Le, T.M.; Wang, H.C.; Yen, C.H. Bioassay-guided discovery of potential partial extracts with cytotoxic effects on liver cancer cells from vietnamese medicinal herbs. Processes 2021, 9, 1956. [Google Scholar] [CrossRef]
  296. Mannan, A.; Kawser, M.J.; Ahmed, A.M.A.; Islam, N.N.; Alam, S.M.M.; Emon, M.A.E.K.; Gupta, S. Das Assessment of antibacterial, thrombolytic and cytotoxic potential of cassia alata seed oil. J. Appl. Pharm. Sci. 2011, 1, 56–59. [Google Scholar]
  297. Yui, S.; Mikami, M.; Kitahara, M.; Yamazaki, M. The inhibitory effect of lycorine on tumor cell apoptosis induced by polymorphonuclear leukocyte-derived calprotectin. Immunopharmacology 1998, 40, 151–162. [Google Scholar] [CrossRef]
  298. Patel, D. Crinum asiaticum Linn: A Medicinal Herb as Well as Ornamental Plant in Central India. Int. J. Environ. Sci. Nat. Resour. 2017, 6, 1–7. [Google Scholar] [CrossRef]
  299. Hyun, J.H.; Kang, J.; Kim, S.C.; Kim, E.; Kang, J.H.; Kwon, J.M.; Park, D.B.; Lee, Y.J.; Yoo, E.S.; Kang, H.K. The effects of crinum asiaticum on the apoptosis induction and the reversal of multidrug resistance in hl-60/mx2. Toxicol. Res. 2008, 24, 29–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Yusoff, S.M. Anti-angiogenesis as a possible mechanism of action for anti-tumor (potential anti-cancer) activity of Crinum asiaticum leaf methanol extract. J. Angiother. 2017, 1, E012–E017. [Google Scholar] [CrossRef]
  301. Tan, W.-N.; Shahbudin, F.N.; Mohamed Kamal, N.N.S.N.; Tong, W.-Y.; Leong, C.-R.; Lim, J.-W. Volatile Constituents of the Leaf Essential Oil of Crinum asiaticum and their Antimicrobial and Cytotoxic Activities. J. Essent. Oil Bear. Plants 2019, 22, 947–954. [Google Scholar] [CrossRef]
  302. Lim, H.S.; Kim, Y.; Kim, Y.J.; Sohn, E.; Kim, J.H.; Jeong, S.J. The Effects of Crinum asiaticum var. japonicum Baker Seeds on Neuroprotection and Antineuroinflammation in Neuronal Cell Lines. Nat. Prod. Commun. 2020, 15, 10. [Google Scholar] [CrossRef]
  303. Seoposengwe, K.; van Tonder, J.J.; Steenkamp, V. In vitro neuroprotective potential of four medicinal plants against rotenone-induced toxicity in SH-SY5Y neuroblastoma cells. BMC Complement. Altern. Med. 2013, 13, 353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Ofori, M.; Danquah, C.A.; Ativui, S.; Doe, P.; Asamoah, W.A. In-Vitro Anti-Tuberculosis, Anti-Efflux Pumps and Anti-Biofilm Effects of Crinum Asiaticum Bulbs. Biomed. Pharmacol. J. 2021, 14, 1905–1915. [Google Scholar] [CrossRef]
  305. Goswami, S.; Das, R.; Ghosh, P.; Chakraborty, T.; Barman, A.; Ray, S. Comparative antioxidant and antimicrobial potentials of leaf successive extract fractions of poison bulb, Crinum asiaticum L. Ind. Crops Prod. 2020, 154, 112667. [Google Scholar] [CrossRef]
  306. Fu, L.; Zheng, Y.; Zhang, P.; Zhang, H.; Xu, Y.; Zhou, J.; Zhang, H.; Karimi-Maleh, H.; Lai, G.; Zhao, S.; et al. Development of an electrochemical biosensor for phylogenetic analysis of Amaryllidaceae based on the enhanced electrochemical fingerprint recorded from plant tissue. Biosens. Bioelectron. 2020, 159, 112212. [Google Scholar] [CrossRef]
  307. Min, B.S.; Kim, Y.H.; Tomiyama, M.; Nakamura, N.; Miyashiro, H.; Otake, T.; Hattori, M. Inhibitory effects of Korean plants on HIV-1 activities. Phytother. Res. 2001, 15, 481–486. [Google Scholar] [CrossRef]
  308. Naira, J.J.; Van Staden, J.; Bonnet, S.L.; Wilhelm, A. Antibacterial properties of the family amaryllidaceae: Evaluation of plant extracts in vitro. Nat. Prod. Commun. 2017, 12, 1145–1151. [Google Scholar] [CrossRef] [Green Version]
  309. Surain, P.; Aneja, K.R. Anticandidal potential of Crinum asiaticum leaves extract against selected oral and vaginal Candida pathogens. J. Innov. Biol. 2014, 6473, 27–30. [Google Scholar]
  310. Noubissi, P.A.; Fokam Tagne, M.A.; Fankem, G.O.; Ngakou Mukam, J.; Wambe, H.; Kamgang, R. Effects of Crinum jagus Water/Ethanol Extract on Shigella flexneri-Induced Diarrhea in Rats. Evid. -Based Complement. Altern. Med. 2019, 2019, 9537603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  311. Udegbunam, S.O.; Udegbunam, R.I.; Nnaji, T.O.; Anyanwu, M.U.; Kene, R.O.C.; Anika, S.M. Antimicrobial and antioxidant effect of methanolic Crinum jagus bulb extract in wound healing. J. Intercult. Ethnopharmacol. 2015, 4, 239–248. [Google Scholar] [CrossRef] [PubMed]
  312. Azikiwe, C.; Amazu, L. The potential organo-toxicity safety of Morpholine and Crinum jagus in rats. Discovery 2015, 10, 113–120. [Google Scholar]
  313. Akintola, A.O.; Kehinde, A.O.; Adebiyi, O.E.; Ademowo, O.G. Anti-tuberculosis activities of the crude methanolic extract and purified fractions of the bulb of Crinum jagus. Niger. J. Physiol. Sci. 2013, 28, 135–140. [Google Scholar]
  314. Ka, S.; Merindol, N.; Sow, A.A.; Singh, A.; Landelouci, K.; Plourde, M.B.; Pépin, G.; Masi, M.; Di Lecce, R.; Evidente, A.; et al. Amaryllidaceae Alkaloid Cherylline Inhibits the Replication of Dengue and Zika Viruses. Antimicrob. Agents Chemother. 2021, 65, e0039821. [Google Scholar] [CrossRef]
  315. Maroyi, A. A review of ethnoboatany, therapeutic value, phytochemistry and pharmacology of Crinum macowanii Baker: A highly traded bulbous plant in Southern Africa. J. Ethnopharmacol. 2016, 194, 595–608. [Google Scholar] [CrossRef]
  316. Ilavenil, S.; Kaleeswaran, B.; Sumitha, P.; Tamilvendan, D.; Ravikumar, S. Protection of human erythrocyte using Crinum asiaticum extract and lycorine from oxidative damage induced by 2-amidinopropane. Saudi J. Biol. Sci. 2011, 18, 181–187. [Google Scholar] [CrossRef] [Green Version]
  317. Uddin, Z.; Bin, T.; Kumar, A.; Jenny, A.; Dutta, M.; Morshed, M.; Kawsar, H. Anti-Inflammatory and Antioxidant Activity of Leaf extract of Crinum asiaticum. J. Pharm. Res. 2015, 5, 5553–5556. [Google Scholar]
  318. Indradevi, S.; Ilavenil, S.; Kaleeswaran, B.; Srigopalram, S.; Ravikumar, S. Ethanolic extract of Crinum asiaticum attenuates hyperglycemia-mediated oxidative stress and protects hepatocytes in alloxan induced experimental diabetic rats. J. King Saud. Univ. Sci. 2015, 24, 171–177. [Google Scholar] [CrossRef] [Green Version]
  319. Ghane, S.G.; Attar, U.A.; Yadav, P.B.; Lekhak, M.M. Antioxidant, anti-diabetic, acetylcholinesterase inhibitory potential and estimation of alkaloids (lycorine and galanthamine) from Crinum species: An important source of anticancer and anti-Alzheimer drug. Ind. Crops Prod. 2018, 125, 168–177. [Google Scholar] [CrossRef]
  320. Alawode, T.T.; Lajide, L.; Owolabi, B.J.; Olaleye, M.T. Studies on In vitro Antioxidant and Anti-Inflammatory Activities of Crinum jagus Leaves and Bulb Extracts. Int. J. Biochem. Res. Rev. 2019, 28, 1–9. [Google Scholar] [CrossRef]
  321. Adewusi, E.A.; Steenkamp, V. In vitro screening for acetylcholinesterase inhibition and antioxidant activity of medicinal plants from southern Africa. Asian Pac. J. Trop. Med. 2011, 4, 829–835. [Google Scholar] [CrossRef] [Green Version]
  322. Chahal, S.; Lekhak, M.M.; Kaur, H.; Shekhawat, M. Unraveling the medicinal potential and conservation of Indian Crinum Unraveling the medicinal potential and conservation of Indian Crinum (Amaryllidaceae) species. S. Afr. J. Bot. 2020, 136, 7–15. [Google Scholar] [CrossRef]
  323. Kang, J.; Choi, J.H.; Lee, J.G.; Yoo, E. The Mechanism of Crinum asiaticum var. japonicum on the Activation of Anagen. Korean J. Pharmacogn. 2017, 48, 148–154. [Google Scholar]
  324. Jeong, Y.J.; Sohn, E.H.; Jung, Y.H.; Yoon, W.J.; Cho, Y.M.; Kim, I.; Lee, S.R.; Kang, S.C. Anti-obesity effect of Crinum asiaticum var. japonicum Baker extract in high-fat diet-induced and monogenic obese mice. Biomed. Pharmacother. 2016, 82, 35–43. [Google Scholar] [CrossRef]
  325. Taiwe, G.S.; Tchoya, T.B.; Menanga, J.R.; Dabole, B.; De Waard, M. Anticonvulsant activity of an active fraction extracted from Crinum jagus L. (Amaryllidaceae), and its possible effects on fully kindled seizures, depression-like behaviour and oxidative stress in experimental rodent models. J. Ethnopharmacol. 2016, 194, 421–433. [Google Scholar] [CrossRef]
  326. Heinrich, M. Galanthamine from Galanthus and other Amaryllidaceae--chemistry and biology based on traditional use. Alkaloids. Chem. Biol. 2010, 68, 157–165. [Google Scholar] [CrossRef]
  327. Jilani, M.S.; Tagwireyi, D.; Gadaga, L.L.; Maponga, C.C.; Mutsimhu, C. Cognitive-Enhancing Effect of a Hydroethanolic Extract of Crinum macowanii against Memory Impairment Induced by Aluminum Chloride in BALB/c Mice. Behav. Neurol. 2018, 2018, 2057219. [Google Scholar] [CrossRef] [Green Version]
  328. Snijman, D.A.; Meerow, A.W. Floral and macroecological evolution within Cyrtanthus (Amaryllidaceae): Inferences from combined analyses of plastid ndhF and nrDNA ITS sequences. S. Afr. J. Bot. 2010, 76, 217–238. [Google Scholar] [CrossRef] [Green Version]
  329. Galley, C.; Bytebier, B.; Bellstedt, D.U.; Linder, H.P. The Cape element in the Afrotemperate flora: From Cape to Cairo? Proc. R. Soc. B Biol. Sci. 2007, 274, 535–543. [Google Scholar] [CrossRef] [Green Version]
  330. Mucina, L.; Rutherford, M.C. The Vegetation of South Africa, Lesotho and Swaziland; South African National Biodiversity Institute: Cape Town, South Africa, 2006. [Google Scholar]
  331. Born, J.; Linder, H.P.; Desmet, P. The Greater Cape Floristic Region. J. Biogeogr. 2007, 34, 147–162. [Google Scholar] [CrossRef]
  332. Cowling, R.M.; Procheş, Ş.; Vlok, J.H.J. On the origin of southern African subtropical thicket vegetation. S. Afr. J. Bot. 2005, 71, 1–23. [Google Scholar] [CrossRef] [Green Version]
  333. Bhat, R.B.; Jacobs, T.V. Traditional herbal medicine in Transkei. J. Ethnopharmacol. 1995, 48, 7–12. [Google Scholar] [CrossRef]
  334. Nwude, N.; Ebong, O.O. Some plants used in the treatment of leprosy in Africa. Lepr. Rev. 1980, 51, 11–18. [Google Scholar] [CrossRef]
  335. Rárová, L.; Ncube, B.; Van Staden, J.; Fürst, R.; Strnad, M.; Gruz, J. Identification of Narciclasine as an in Vitro Anti-Inflammatory Component of Cyrtanthus contractus by Correlation-Based Metabolomics. J. Nat. Prod. 2019, 82, 1372–1376. [Google Scholar] [CrossRef]
  336. Hutchings, A.; Scott, A.H.; University of Zululand; National Botanical Institute (South Africa). Zulu Medicinal Plants: An Inventory; University of Natal Press: Scottsville, South Africa, 1996; ISBN 0869808931. [Google Scholar]
  337. 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] [Green Version]
  338. Mahlangeni, N.T.; Moodley, R.; Jonnalagadda, S.B. Phytochemical analysis of Cyrtanthus obliquus bulbs from the informal street market of Kwazulu-Natal, South Africa. Afr. J. Tradit. Complement. Altern. Med. 2015, 12, 28–34. [Google Scholar] [CrossRef] [Green Version]
  339. Brine, N.D.; Campbell, W.E.; Bastida, J.; Herrera, M.R.; Viladomat, F.; Codina, C.; Smith, P.J. A dinitrogenous alkaloid from Cyrtanthus obliquus. Phytochemistry 2002, 61, 443–447. [Google Scholar] [CrossRef]
  340. Masi, M.; Mubaiwa, B.; Mabank, T.; Karakoyun; 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]
  341. Elgorashi, E.E.; Van Staden, J. Pharmacological screening of six Amaryllidaceae species. J. Ethnopharmacol. 2004, 90, 27–32. [Google Scholar] [CrossRef] [PubMed]
  342. Elgorashi, E.E.; Van Staden, J. Alkaloids from Cyrtanthus falcatus. S. Afr. J. Bot. 2003, 69, 593–594. [Google Scholar] [CrossRef] [Green Version]
  343. Weniger, B.; Italiano, L.; Beck, J.P.; Bastida, J.; Bergonon, S.; Codina, C.; Lobstein, A.; Anton, R. Cytotoxic activity of Amaryllidaceae alkaloids. Planta Med. 1995, 61, 77–79. [Google Scholar] [CrossRef] [PubMed]
  344. Elgorashi, E.E.; Stafford, G.I.; Mulholland, D.; Van Staden, J. Isolation of captan from Cyrtanthus suaveolens: The effect of pesticides on the quality and safety of traditional medicine. S. Afr. J. Bot. 2004, 70, 512–514. [Google Scholar] [CrossRef] [Green Version]
  345. Kim, H.W.; Wang, M.; Leber, C.A.; Nothias, L.F.; Reher, R.; Kang, K.B.; van der Hooft, J.J.J.; Dorrestein, P.C.; Gerwick, W.H.; Cottrell, G.W. NPClassifier: A Deep Neural Network-Based Structural Classification Tool for Natural Products. J. Nat. Prod. 2021, 84, 2795–2807. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical space of compounds identified from T. violacea. Blue circles are sulfur-containing compounds while red circles are compounds devoid of sulfur in their chemical structures. PCA analysis carried out using DataWarrior [54].
Figure 1. Chemical space of compounds identified from T. violacea. Blue circles are sulfur-containing compounds while red circles are compounds devoid of sulfur in their chemical structures. PCA analysis carried out using DataWarrior [54].
Molecules 27 04475 g001
Figure 2. Analysis of cLogP and molecular weight space occupied by compounds identified in T. violacea. Blue circles are sulfur-containing compounds while red circles are compounds devoid of sulfur in their chemical structures. Plot generated using DataWarrior [54].
Figure 2. Analysis of cLogP and molecular weight space occupied by compounds identified in T. violacea. Blue circles are sulfur-containing compounds while red circles are compounds devoid of sulfur in their chemical structures. Plot generated using DataWarrior [54].
Molecules 27 04475 g002
Figure 3. Chemical structures of compounds identified in T. violacea. (1) Marasmicin (1), (2) marasmin (2), allicin (3)—possesses antibacterial and antifungal activity, d-fructofuranosyl-β(2→6)-methyl-α-d-glucopyranoside (4), β-d-fructofuranosyl-(2→6)-α-d-glucopyranoside (5), methyl-α-d-glucopyranoside (6), bis(methylthiomethyl) disulfide (7)—found to constitute 48% of volatiles in aerial parts of T. violacea [55], methyl-2-thioethyl thiomethyl trisulfide (8)—found to constitute 16% of volatile compounds in aerial parts of T. violacea [55], methyl (methylthio)methyl disulfide (9)—found to constitute 10 % of volatile compounds in aerial parts of T. violacea [55], naphthalene (10)—interestingly observed to significantly increase in concentration in plants infected by the fungus Beauveria bassiana in comparison to untreated controls [59], nonanal (11)—also observed to significantly decrease in concentration in plants infected by the fungi Beauveria bassiana in comparison to untreated controls [59] and finally kaempferol (12)—which possesses multiple biological activities including antioxidant, anticancer and anti-inflammatory properties [60,61,62].
Figure 3. Chemical structures of compounds identified in T. violacea. (1) Marasmicin (1), (2) marasmin (2), allicin (3)—possesses antibacterial and antifungal activity, d-fructofuranosyl-β(2→6)-methyl-α-d-glucopyranoside (4), β-d-fructofuranosyl-(2→6)-α-d-glucopyranoside (5), methyl-α-d-glucopyranoside (6), bis(methylthiomethyl) disulfide (7)—found to constitute 48% of volatiles in aerial parts of T. violacea [55], methyl-2-thioethyl thiomethyl trisulfide (8)—found to constitute 16% of volatile compounds in aerial parts of T. violacea [55], methyl (methylthio)methyl disulfide (9)—found to constitute 10 % of volatile compounds in aerial parts of T. violacea [55], naphthalene (10)—interestingly observed to significantly increase in concentration in plants infected by the fungus Beauveria bassiana in comparison to untreated controls [59], nonanal (11)—also observed to significantly decrease in concentration in plants infected by the fungi Beauveria bassiana in comparison to untreated controls [59] and finally kaempferol (12)—which possesses multiple biological activities including antioxidant, anticancer and anti-inflammatory properties [60,61,62].
Molecules 27 04475 g003
Figure 4. Chemical structures of compounds isolated from the genus Allium. Quercetin (13), vanillic acid (14) and adenosine (15).
Figure 4. Chemical structures of compounds isolated from the genus Allium. Quercetin (13), vanillic acid (14) and adenosine (15).
Molecules 27 04475 g004
Figure 5. Chemical structures of compounds isolated from the genus Crinum. Lycorine (16), crinamine (17), galantamine (18) and crinine (19).
Figure 5. Chemical structures of compounds isolated from the genus Crinum. Lycorine (16), crinamine (17), galantamine (18) and crinine (19).
Molecules 27 04475 g005
Figure 6. Chemical structures of selected compounds from Crytanthus. Zephyranthine (20), 1,2-O-diacetylzephyranthine (21), haemanthamine (22), haemanthadine (23), galanthamine (18), 5,7-dihydroxy-6-methoxy-3-(4ꞌ-methoxybenzyl)chroman-4-one (24), 5,7-dihydroxy-6-methoxy-3-(4ꞌ-methoxybenzylidene)chroman-4-one (25), 5,7-dihydroxy-6-methoxy-3-(4ꞌhydroxybenzyl)chroman-4-one (26), 5,7-dihydroxy-3-(4ꞌhydroxybenzylidene)-chroman-4-one (27) and naciprimine (28).
Figure 6. Chemical structures of selected compounds from Crytanthus. Zephyranthine (20), 1,2-O-diacetylzephyranthine (21), haemanthamine (22), haemanthadine (23), galanthamine (18), 5,7-dihydroxy-6-methoxy-3-(4ꞌ-methoxybenzyl)chroman-4-one (24), 5,7-dihydroxy-6-methoxy-3-(4ꞌ-methoxybenzylidene)chroman-4-one (25), 5,7-dihydroxy-6-methoxy-3-(4ꞌhydroxybenzyl)chroman-4-one (26), 5,7-dihydroxy-3-(4ꞌhydroxybenzylidene)-chroman-4-one (27) and naciprimine (28).
Molecules 27 04475 g006
Table 1. Geographical distribution and traditional uses of Tulbaghia species.
Table 1. Geographical distribution and traditional uses of Tulbaghia species.
Plant SpeciesGeographical DistributionTraditional UsesReferences
T. violaceaIndigenous to the Eastern Cape, KwaZulu-Natal, Gauteng, Free State and Mpumalanga Provinces of South Africa.The leaves and bulbs are used in the management of fever and colds, tuberculosis, asthma, and stomach problems. The leaves are eaten as vegetables and for the management of oesophageal cancer. It is also used as a snake repellent. [8,50]
T. alliaceaNative to South Africa and grows mostly in the Eastern Cape and southern KwaZulu-Natal Provinces of South Africa.Its bruised rhizome is used locally in bathwater to relieve fever, rheumatism, and paralysis, and in small doses as a laxative. T. alliacea is used for the management of stomach problems, asthma, and pulmonary tuberculosis. Its rhizome infusion is administered as an enema.[8,51]
T. simmleriNative to the South African Drakensberg mountains growing as isolated plants on rocky ledges.Bulbs and leaves are used as a remedy for gastrointestinal ailments, enemas, high blood pressure, heart problems, chest complaints, high cholesterol, constipation, rheumatism, asthma, fever, pulmonary tuberculosis, earache, human immunodeficiency virus (HIV), paralysis, and cardiovascular diseases.[50,52]
T. acutilobaFound in the rainfall regions of southern Africa, occurring in the Eastern Cape, KwaZulu-Natal, Limpopo, Free State, Gauteng, North West, and Mpumalanga Provinces of South Africa, as well as in Lesotho, Swaziland and Botswana.T. acutiloba leaves are used as a culinary herb and snake repellent. It is used to treat barrenness, flu, bad breath, and as an aphrodisiac. It is also cultivated to keep snakes away from the homestead.[8]
T. natalensisAlthough native to South Africa, but is now grown worldwide.It is used as a culinary herb and snake repellent.[53]
T. cernuaCommonly found in the Eastern Cape, Free State, Gauteng, KwaZulu-Natal, Limpopo, Mpumalanga, North West and Western Cape Provinces of South Africa. It is used for ornamental purposes.[8]
T. leucanthaWidely distributed in southern Africa including Botswana, Lesotho, South Africa, Swaziland, Zambia, and Zimbabwe. Its rhizome is scraped clean and boiled in stews or roasted as a vegetable. Its leaves and stems are used as a culinary herb and protective charm.[53]
T. ludwigianaCommonly found in the Eastern Cape, KwaZulu-Natal, Northern Provinces of South Africa and in Swaziland. It is traditionally used as a love charm.[53]
Table 2. Published documents on the genus Tulbaghia per country.
Table 2. Published documents on the genus Tulbaghia per country.
CountryNo. of Documents *
South Africa99
United Kingdom15
United States12
Czech Republic8
Italy7
India6
Germany5
Australia3
China3
Belgium2
* Data retrieved following query of the Scopus database (https://www.scopus.com/, accessed on 22 February 2022) using the keyword “Tulbaghia”. The search was carried out on 22 February 2022.
Table 3. Number of published studies per specific disease or pharmacological area.
Table 3. Number of published studies per specific disease or pharmacological area.
DiseaseNo. of Published Studies #
Antimicrobial26
Cancer11
Antioxidant13
Diabetes2
Cardiovascular12
Antithrombogenic2
Miscellaneous17
# Studies considered are those published from 1997 to 2022. A number of these, published before 2013, have been succinctly discussed by Aremu and Van Staden [8].
Table 4. Antimicrobial activity of Tulbaghia species.
Table 4. Antimicrobial activity of Tulbaghia species.
Plant SpeciesExtraction SolventPlant Part UsedBiological ActivityReferences
T. violaceaDichloromethaneBulbsMIC ranging from 20 to 300 µg/mL against Bacillus subtilis, methicillin-resistant S. aureus, S. epidermidis, E. coli, K. pneumoniae, P. aeruginosa, C. albicans and C. parapsilosis.[67]
T. violaceaHexane and ethanolFlowers and callus culturesModerate to strong broad-antimicrobial (E. coli, P. aeruginosa, S. aureus, Aspergillus niger and C. albicans) activity observed by zone of inhibition in the agar well disc diffusion method.[68]
T. violaceaWaterBulbsSignificant reduction in A. flavus β-glucan and chitin synthesis corresponding to a dose-dependent inhibition of the enzymes β-glucan and chitin synthase, respectively.This results in inhibition of ergosterol production in the fungus.[70,71]
T. violaceaAcetoneBulbsVaried light intensities, pH and watering frequencies substantially impacted both growth and potency of plant extracts against the fungi F. oxysporum.[73,74]
T. violaceaWaterRoots, bulbs, leaves and flowersSignificantly compromised population densities of the nematode M. incognita race 2 on tomato roots and in the soil.[75]
T. violaceaDichloromethaneBulbsAntiparasitic activity against T. brucei (IC50 = 2.83 µg/mL) and L. tarentolae (IC50 = 6.29 µg/mL). [67]
Table 5. Anticancer activity of Tulbaghia.
Table 5. Anticancer activity of Tulbaghia.
Plant SpeciesExtraction SolventPlant Part UsedBiological ActivityReferences
T. violaceaMethanolLeaves and rootsMarked time- and dose-dependent cytotoxic effect on cancer cell lines. Induced apoptosis using p53-independent pathway.[78]
T. violaceaMethanol, butanol, and hexaneLeavesMethanol extract was prolific against multiple cell lines. Hela and ME-180 cell lines treated with methanol and hexane extracts showed an increase in caspase 3/7 activity. Both methanol and hexane extracts induced a 10-fold increase in expression of p53 gene in Hela cells.[79]
T. violaceaMethanol:water:formic acid (80:20:0.1, v/v/v)FlowersDemonstrated activity against ovarian tumor cells.[80]
T. violaceaWater and methanolLeavesWater-soluble extract emerged as the most cytotoxic (IC50 = 314 µg/mL), compared to the methanol extract (IC50 = 780 µg/mL), against the MDA-MB-231 triple-negative breast cancer cell line. Water-soluble extract prevented cell migration completely for 13 h at 300 µg/mL.[82]
T. violaceaHexane and ethanolFlowers and callus culturesExtracts showed marked cytotoxicity (60–74% growth inhibition at 250 µg/mL) against three different cell lines (Hep G2, PC-3 and MCF-7).[68]
T. violaceaAcetone and waterLeavesAnticancer activity against oral cancer with an IC50 (acetone extract) of 0.2 mg/mL; IC50 (water extract) of 1 mg/mL.[84]
T. violaceaMethanol:water (1:1)Whole plantsTwo pro-apoptotic glucopyranosides d-fructofuranosyl-β (2→6)-methyl-α-d-glucopyranoside and β-d-fructofuranosyl-(2→6)-α-d-glucopyranoside isolated and identified as active anticancer agents in the plant.[85]
T. violaceaWaterWhole plantsMoA of the three compounds, namely methyl-α-d-glucopyranoside, d-fructofuranosyl-β (2→6)-methyl-α-d-glucopyranoside and β-d-fructofuranosyl-(2→6)-α-d-glucopyranoside isolated from the water extract, deciphered to be through induction of apoptosis by targeting the mitochondrial (intrinsic) pathway [86]
Table 6. Antioxidant activity of Tulbaghia species.
Table 6. Antioxidant activity of Tulbaghia species.
Plant SpeciesExtraction SolventPlant Part UsedBiological ActivityReferences
T. violaceaWaterLeavesDose-dependent antioxidant activity measured using the DPPH (Log IC50 = 0.49 mg/mL) and ABTS (Log IC50 = 0.24 mg/mL) assays[92]
T. violaceaMethanol/water/formic acid (80:20:0.1, v/v/v)FlowersMarked antioxidant activity was observed using 3 different types of assays, namely DPPH, FRAP and TREC[80]
T. acutilobaHydro-methanolic extractsRoots, rhizomes, leaves and flowersDose-dependent antioxidant activity observed with the rhizome extract emerging as the most active plant part (IC50 DPPH = 0.202 mg/mL and peak scavenging activity of 95)[91]
T. violaceaHexane and ethanolFlowers and callus culturesDose-dependent antioxidant activity with IC50 ranging from 1.933 to 7.350 mg/mL in the DPPH assay[68]
T. violaceaAcetoneLeavesIC50 DPPH = 0.08 mg/mL; IC50 ABTS = 0.03 mg/mL [84]
T. acutilobaAcetoneLeavesIC50 DPPH = 0.16 mg/mL; IC50 ABTS = 0.07 mg/mL [84]
T. alliaceaAcetoneLeavesIC50 DPPH = 0.06 mg/mL; IC50 ABTS = 0.06 mg/mL [84]
T. cernuaAcetoneLeavesIC50 DPPH = 0.21 mg/mL; IC50 ABTS = 2.34 mg/mL [84]
T. leucanthaAcetoneLeavesIC50 DPPH = 0.39 mg/mL; IC50 ABTS = 0.03 mg/mL [84]
T. ludwigianaAcetoneLeavesIC50 DPPH = 0.26 mg/mL; IC50 ABTS = 0.09 mg/mL [84]
T. natalensisAcetoneLeavesIC50 DPPH = 2.70 mg/mL; IC50 ABTS = 0.04 mg/mL [84]
Table 7. Antidiabetic, anticardiovascular and antithrombogenic activity of Tulbaghia species.
Table 7. Antidiabetic, anticardiovascular and antithrombogenic activity of Tulbaghia species.
Plant SpeciesExtraction SolventPlant Part UsedBiological ActivityReferences
Diabetes
T. violaceaMethanolRhizomeAttenuated diabetes associated physiological complications in streptozotocin-induced diabetic rats.[96]
T. violaceaMethanolRhizomeNoticeably reduced blood glucose and serum lipid (TG, TC, and VLDL) levels while raising plasma insulin in a streptozotocin-induced diabetic rat model.[97]
Cardiovascular
T. violaceaMethanol LeavesMarkedly reduced systolic BP, diastolic BP, mean arterial pressure and the heart rate in both age-induced and spontaneous hypertensive rats.[98]
T. violaceaMethanolRhizome50 mg/kg significantly improved kidney function in vivo.[99]
T. acutilobaHydro-methanolic extractsRoots, rhizomes, leaves and flowersAll extracts inhibited the Angiotensin-1-Converting Enzyme in vitro (> 50 % inhibition at a concentration range of 125–1000 μg/mL). Extracts of leaves demonstrated activity comparable to that of the control drug ramipril.[91]
Antithrombogenic
T. violaceaWaterLeavesNoticeable inhibition of platelet adhesion by a novel scaffold consisting of polycaprolactone incorporated with 10 % (w/w) plant extracts.[90]
T. violaceaWaterLeavesMarked inhibition of platelet adhesion (70% inhibition at 0.1 mg/mL within 15 min post-exposure).[92]
Table 8. Miscellaneous biological properties of extracts of Tulbaghia species.
Table 8. Miscellaneous biological properties of extracts of Tulbaghia species.
Plant SpeciesExtraction SolventPlant Part UsedBiological ActivityReferences
T. violaceaMethanol/water/formic acid (80:20:0.1, v/v/v)FlowersReduced 1-42 β-amyloid peptide formation and arrested oxidative stress in vivo.[80]
T. violaceaMethanol LeavesDemonstrated in vivo anticonvulsant activity by attenuating tonic convulsions induced by either pentylenetetrazole, bicuculline, picrotoxin, strychnine or NMDLA.[102]
T. violaceaAcetoneMixture of leaves and bulbsMarked tick repellence properties of fungus-exposed plants at low treatment concentrations (5 % w/v and 10 % w/v).[59]
T. violaceaWaterLeaves, stems, and rootsInduced conspicuous genotoxicity effects at high test concentrations (250, 500 and 1000 µg/mL) in the A. cepa assay. [103]
T. violaceaWater and ethanolLeaves, stems, and rootsBroad murine macrophage antiproliferative and cytotoxicity activity influenced by both extract test concentrations, type of solvent and plant part used.[104]
Table 9. Traditional medicinal uses of Allium species.
Table 9. Traditional medicinal uses of Allium species.
Plant Species Mode of PreparationTraditional Medicinal UsesReference
A. cepaRaw, juice of bulb or rhizome, paste, decoctions, cataplasm, maceration, infusionAlopecia, antilithic (stone disease), anti-obesity, blood purifying, bronchitis, constipation, cardiovascular disease, cough, diabetes, eye diseases, erectile dysfunction, fever, hearing loss, headaches, hemorrhoids, epilepsy, oligomenorrhea, jaundice, lower gastrointestinal bleeding, prostate cancer, rheumatism, rubefacient, sinusitis, stomach pains, snake bites, skin diseases, teeth disorders, reduce flatulence, wound healing [133,134,135]
A. sativumExtracts of leaves or bulb Antiseptic, anthelmintic, antithrombotic, antilipidemic, aphrodisiac, anti-greying of hair, bronchitis, carminative, cough, colic, cancers (gastric, prostate, colorectal adenomatous polyps, squamous cell carcinoma), diabetes, diaphoretic, dysentery, eczema, facial paralysis, fever, flatulence, galactagogue, high blood pressure, intestinal worms, liver disorders, rheumatism, scabies, tetanus, stomach pains, tuberculosis[135,136,137,138,139]
A. umbilicatumRaw or cooked bulb, leaves, flowers Non-specific reduction in blood cholesterol levels, tonify digestive and circulatory systems [116]
A. victoralisFresh, pickled, boiled and salted flowers, leaves and rootsAppetizer, amenorrhea, pediatric otitis, bronchitis, diarrhea, dropsy, expectorant, hypofunction of stomach, inflammatory eye diseases, meteorism, gastroenteritis, heart diseases (atherosclerosis), rheumatism[140]
A. ascalonicum/A. cepa var aggregatumBulb and leavesAllergies, appetizer, cold, cancers, fever, obesity, rheumatoid arthritis, soothes nerves, diabetes, post-menopausal syndrome [141,142,143,144,145]
A. chinenseFlower, leaves, roots, seedpodsAngina pectoris, astringent, bronchitis, carminative, chest pains, diarrhea, expectorant, pleurisy, tenesmus in cases of dysentery, reducing cholesterol, tonic to the digestive and circulatory systems[146]
A. tuberosumRaw or cooked leaves, roots, oils from seed Asthma, abdominal pain, carminative, cuts and wounds, diabetes, diarrhea, kidney and bladder weakness, nocturnal emission, urinary incontinence, spermatorrhea, stomachic [147]
A. griffithianumLeaves and bulb Carminative, colic indigestion, dyspepsia, diabetes control
A. oreoprasumLeaves and bulb Cough and cold, diabetes control, diarrhea, dysentery, fever, gastritis, oedema, headache, jaundice, stomachache, rheumatism, numbness of limbs[124]
Table 10. Bioactive compounds isolated from Allium species.
Table 10. Bioactive compounds isolated from Allium species.
Plant SpeciesPlant PartCountryIsolated CompoundsBioactivityReferences
A. ursinum L.Leaves,
underground parts,
fresh flowers
Poland
Bulgaria
1,2-di-O-α-linolenoyl-3-O-β-d-galactopyranosyl-sn-glycerol; β-sitosterol3-O-β-d-glucopyranoside;
kaempferol 3-O-β-glucopyranoside and kaempferol 3-O-β-neohesperidoside.
(-S-)-spirost-5-en-3β-ol tetrasaccharide, (25R)-spirost-5,25(27)-dien-3 β-ol tetrasaccharide, 3-hydroxypregna-5,16-dien-20-one glycoside.
Thymidine, adenosine, astragalin (kaempferol-3-O-β-d-glucopyranoside, kaempferol-3-O-β-d-glucopyranosyl-7-O-β-d-glucopyranoside, kaempferol-3-O-β-d-neohesperoside, and kaempferol-3-O-β-d-neohesperoside-7-O-β-d glucopyranoside.
Anti-ADP-aggregation activity in human blood platelets.
Inhibition of human platelet aggregation.
Cytotoxic activity against murine melanoma B16 and sarcoma XC.
[148,149,150,151]
A. mongolicumAerial partsChinaMongoflavonoids A1, A2, A3, A4, B1, B2 and monogophenosides A1, A2, A3, B.Increase in the height of mouse small intestine.[152]
A. cepa.
A. cepa L.
Pigmented scales of red onion,
bulbs,
red onion skin waste
NaplesQuercetin.
3-O-(3″-O-β-glucopyranosyl-6″-O-malonyl-β-glucopyranoside)-4-O-β-glucopyranoside, cyanidin 3,4′-di-O-β-glucopyranoside, cyanidin-4′-O-β-glucoside, peonidin 3-O-(6 ″-O-malonyl-β-glucopyranoside).
5-hydroxy-3-methyl-4-propylsulfanyl-5H-furan-2-one, (hydroxymethyl) furfural, acetovanillone, methyl 4-hydroxyl cinnamate and ferulic acid methyl ester.
3-O-β-glucopyranoside and 3-O-(6″-O-malonyl-β-glucopyranoside) of 5-carboxypyranocyanidin.
Ceposide A, ceposide B and ceposide C.
Spiraeoside (4′-O-glucoside of quercetin).
Onionin A1, onionin A2, onionin A3, onionin B1 and B2.
Onionin A1 (3,4-dimethyl-5-(1E-propenyl)-tetrahydrothiophen-2-sulfoxide-S-oxide).
Cyanidin 3-glucoside (Cy 3-Glc), 3-malonylglucoside (Cy3-MaGlc), cyanidin 3-laminaribioside (Cy 3-Lam) and 3-malonyllaminaribioside (Cy 3-MaLam).
Anti-inflammatory and immunomodulatory effect.
Induction of quinone reductase.
Antifungal activity.
Radical scavenging, anti-inflammatory, inhibition of the expression of B-cell lymphoma 2.
Suppression of tumor progression in mouse ovarian cancer (Onionin A1).
Suppression of tumor-cell proliferation through the inhibition of polarization of M2 activated macrophages.
[153,154,155,156,157,158,159,160,161]
A. sativum.
A. sativum L. var. voghiera.
A. sativum L.
Root, protobulb, leaf sheath and blade,
bulbs,
tuber
Italy Nerolidol, α-pinene, terpinolene.
Voghieroside A1/A2, voghieroside B1/B2, voghieroside C1/C2, voghieroside D1/D2 and voghieroside E1/E2.
Adenosine and guanosine.
Antifungal activity against Sclerotium cepivorum.
Antimicrobial activity.
Strong inhibitory effect on human platelet aggregation generated by 2 μM ADP in both primary and secondary waves (adenosine).
[162,163,164]
A. schoenoprasumWhole plant,
pale-purple flowers
(20S, 25S)-spirost-5-en-3β, 12β,21-triol 3-O-α-L-rhamnopyranosyl-(1→2)-β-d-glucopyranoside, (20S, 25S)-spirost-5-en-3β, 11α,21-triol 3-O-α-L-rhamnopyranosyl-(1→2)-β-d-glucopyranoside, laxogenin 3-O-α-L-rhamnopyranosyl--(1→2)-[β-d-glucopyranosyl-(1→4)]-[β-d-glucopyranoside, (25R)-5α-spirostan-3β, 11α-diol 3-O-β-d-glucopyranosyl-(1→4)]-β-d-galactopyranoside.
(cyanidin 3-O-β-glucosideAII) (kaempferol 3-O-(2-O-β-glucosylFIII-β-glucosideFII)-7-O-β-glucosiduronic acid FIV) malonate AIII (AII-6→AIII-1, FIV-2→AIII-3), 1, (cyanidin 3-O-(3-O-acetyl-β-glucosideAII) (kaempferol 3-O-(2-O-β-glucosylFIII-β-glucosideFII)-7-O-β-glucosiduronic acid FIV) malonate AIII (AII-6→AIII-1, FIV-2→AIII-3), 2, and 7-O-(methyl-O-β-glucosiduronateFIV).
Cytotoxicity against HCT 116 and HT-29 human colon cancer lines.[165,166]
A. minutiflorum RegelBulbs Minutoside A, minutoside B, Minutoside C, alliogenin, neoagigeninAntifungal activity. [167]
A. neapolitanumExtracts 3-O-{[2-O-α-1-rhamnopyrnosyl-4-O-β-d-glucopyranosyl]-β-d-glucopyranoside}, isorhamnetin; 3-O-{[2-O-α-1-rhamnopyrnosyl-6-O-β-d-glucopyranosyl]-β-d-glucopyranoside}, isorhamnetin; 3-O-{[2-O-α-1-rhamnopyranosyl-4-O-β-d-glucopyranosyl]-β-d-glucopyranoside}-7-O-β-d-glucopyranoside and isorhamnetin; 3-O-{[2-O-α-1-rhamnopyranosyl-6-O-β-d-gentiobiosyl]-β-d-glucopyranoside}.Antiplatelet aggregation activity.[168]
A. tripedaleBulbs,
leaves
Iran6,7-dimethoxy-N-trans-caffeoyltyramine; N-trans-feruloyltyramine.
(+)-S-(1-butenyl)-L-cysteine sulfoxide (homoisoalliin), S-(1-butenyl)-L-cysteine (desoxyhomoisoalliin).
NR.[167,168]
A. porrum L.Bulbs Kaempferol 3-O-[2-O-(trans-3-methoxy-4-hydroxycinnamoyl)-β-d-galactopyranosyl]-(1→4)-O-β-d-glucopyranoside; Kaempferol 3-O-[2-O-(trans-3-methoxy-4-hydroxycinnamoyl)-β-d-glucopyranosyl]-(1→6)-O-β-d-glucopyranoside.
(25R)-5 α-spirostan-3 β, 6 β-diol 3-O-[O-β-d-glucopyranosyl-(1→2)-O-[β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside}; (25R)-5 α-spirostan-3 β, 6 β-diol 3-O-{O-β-d-glucopyranosyl-(1→3)-O-β-d-glucopyranosyl-(1→2)-O-[β-d-xylopyranosyl-(1→3)]-O-β-d-galactopyranosyl-(1→4)-β-d-galactopyranoside}
Antiplatelet aggregation activity.
Antifungal activity.
[169,170,171]
A. chinense.
A. chinense G. Don
Bulbs Chinenoside II and chinenoside III.
(25 R,S)-5 α-Spirostan-3β-ol tetrasaccharide, (25R)-3 β-hydroxy-5 α-spirostan-6-one di- and tri-saccharides.
Xiebai-saponin I (laxogenin 3-O-β-xylopyranosyl (1→4)-[α-arabinopyranosyl (1→6)-β-glucopyranoside), laxogenin 3-O-α-arabinopyranosyl (1→6)-β-glucopyranoside, laxogenin, isoliquiritigenin, isoliquiritigenin-4-O-glucoside, and β-sitosterol glucoside.
Inhibition of cAMP phosphodiesterase.
Antitumor-promoting activity (laxogenin).
[172,173,174,175]
A. macrostemon.
A. macrostemon Bunge
Bulbs,
leaves
JapanMacrostemonoside G (26-O-β-d-glucopyranosyl-22-hydroxy-5-β-furost-25(27)-ene-3 β,12 β,26 triol 3-O-β-d-glucopyranosyl(1-->2)-β-d-galactopyranoside) and I (26-O-β-d-glucopyranosyl-22-hydroxy-5 β-furost-25(27)-ene-12-one-3 β,26-diol 3-O-β-d-glucopyranosyl(1→2)-β-d-galactopyranoside).
tigogenin-3-O-β-d-glucopyranosyl(1-->2) [β-d-glucopyranosyl(1→3)1]-β-d-glucopyranosyl(1→4)-β-d-galactopyranoside (1) and tigogenin-3-O-β-d-glucopyranosyl(1→2)[β-d-glucopyranosyl (1→3)(6-O-acetyl-β-d-glucopyranosyl)] (1→4)-β-d-galactopyranoside (2).
Macrostemonoside E-(25R)-26-O-β-d-glucopyranosyl-5 α-furost-20(22)-ene-3 β,26-diol-3-O-β-d-glucopyranosyl (1→2) [β-d-glucopyranosyl (1→3)]-β-d-glucopyranosyl (1→4)-β-d-galactopyranoside; Macrostemonoside F(II)-(25R)-26-O-β-d-glucopyranosyl-5 β-furost-20(22)-ene-3 β,26-diol-3-O-β-d-glucopyranosyl (1→2)-β-d-galactoside.
Allimacronoid A (1-O-(E)-feruloyl-β-d-glucopyranosyl (1-2)-[β-d-glucopyranosyl (1-6)]-β-d-glucopyranose), Allimacronoid B (1-O-(E)-feruloyl-{β-d-glucopyranosyl (1-4)-[β-d-glucopyranosyl (1-2)]}-[β-d-glucopyranosyl (1-6)]-β-d-glucopyranose) and Allimacronoid Cn1-O-(E)-feruloyl-{β-d-glucopyranosyl (1-6)-[β-d-glucopyranosyl (1-2)]}-[β-d-glucopyranosyl (1-6)]-β-d-glucopyranose.
In vitro inhibition of ADP-induced human platelet aggregation (macrostemonoside G).
Inhibitory activity against rabbit platelet aggregation induced by ADP (1).
[176,177,178,179]
A. schubertiiBulbs (25R and S)-5 α-spirostan-2 α,3 β,6 β-triol 3-O-β-d-glucopyranosyl-(1→2)-O-[4-O-benzoyl-β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside, (25R and S)-5 α-spirostan-2α,3β,6 β-triol 3-O-β-d-glucopyranosyl-(1→2)-O-[3-O-benzoyl-β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside, (25R and S)-5 α-spirostan-2α,3β,6 β-triol 3-O-β-d-glucopyranosyl-(1→2)-O-[4-O-(3S)-3-hydroxy-3-methylglutaroyl-β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside and 26-O-β-d-glucopyranosyl-(25R and S)-5 α-furostan-2α,3β,6β,22 zeta,26-pentol 3-O-β-d-glucopyranosyl-(1→2)-O-[β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside.NR.[180]
A. tuberosumSeeds Shanghai (2α, 3β, 5α, 25S)-2,3,27-trihydroxyspirostane 3-O-α-L-rhamnopyranoyl-(1→2)-O-[α-L-rhamnopyranosyl-(1→4)]-β-d-glucopyranoside.
Tuberoside J-(25R)-5 α-spirostan-2α,3
β,27-triol 3-O-α-L-rhamnopyranosyl-(1-->2)-β-d-glucopyranoside; Tuberoside K-(25R)-5α-spirostan-2α,3β 27-triol 3-O-α-L-rhamnopyranosyl-(1→2)-[α-L-rhamnopyranosyl-(1→4)]-β-d-glucopyranoside; and Tuberoside L-27-O-β-d-glucopyranosyl-(25R)-5α-spirostan-2α,3
β,27-triol 3-O-α-d-rhamnopyranosyl-(1-->2)-[α-L-rhamnopyranosyl-(1→4)]-β-d-glucopyranoside.
Tuberoside M-(25S)-5β-spirostane-β,3 β-diol 3-O-α-L-rhamnopyranosyl-(1→4)-β-d-glucopyranoside.
Tuber-ceramide (N-(2′,3′-dihydroxy-tetracosenoyl)-2-amino-1,3,4-trihydroxy octadecane), and Cerebroside (N-(2′,3′-dihydrox-tetra-cosenoyl)-2-amino-1,3,4-trihydroxy octadecane).
Tuberoside M inhibits the proliferation of the human promyelocytic leukemia cell line (HL-60) [181,182,183]
A.albopilosum and A. ostrowskianumBulbs (25 R and S)-5 α-spirostane-2α, 3 β,6 β-triol 3-O-(O-β-d-glucopyranosyl-(1→2)-O-[3-O-acetyl-β-d-xylopyranosyl-(1→3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside), (25R)-2-O-[(S)-3-hydroxy-3-methylglutaroyl]-5 α-spirostane-2α, 3β, 6β-triol 3-O-(O-β-d-glucopyranosyl-(1→2)-O-[β-d-xylopyranosyl-(1—
>3)]-O-β-d-glucopyranosyl-(1→4)-β-d-galactopyranoside), (22S)-cholest-5-ene-1β, β,16 β,22-tetraol 1-O-α-L-rhamnopyranoside 16-O-(O-α-L-rhamnopyranosyl-(1→3)-β-d-glucopyranoside), 1β, 3β, 16β-trihydroxycholest-5-en-22-one 1-O-aα-L-rhamnopyranoside 16-O-(O-α-L-rhamnopyranosyl-(1→3)-β-d-glucopyranoside), 1β,3β,16 bβ-trihydroxy-5 α-cholestan-22-one 1-O-α-L-rhamnopyranoside 16-O-(O-α-L-rhamnopyranosyl-(1→3)-β-d-glucopyranoside) and (22S)-cholest-5-ene-1β,3β, 16β,22-tetraol 16-O-(O-β-d-glucopyranosyl-(1→3)-β-d-glucopyranoside).
NR.[184]
A.fistulosum.
A.fistulosum L.
Whole plant,
leaves,
seeds
Iran Fistulomidate A ((1Z,2E)-Methyl3-(3,4-dimethoxyphenyl)-N-(4-hydroxyphenethyl) acrilimidate) and Fistulomidate B ((1Z,2E)-Methyl3-(3,4-dihydroxyphenyl)-N-(4-hydroxyphenethyl)acrilimidat).
Onionin A1, onionin A2, and onionin A3.
Glycerol mono-(E)-8,11,12-trihydroxy-9-octadecenoate, tianshic acid, 4-(2-formyl-5-hydroxymethylpyrrol-1-yl) butyric acid, p-hydroxybenzoic acid, vanillic acid, and daucosterol.
Antibacterial and cytotoxic activity.
Suppression of tumor progression in mouse ovarian cancer (onionin A1).
Inhibition of the growth of Phytophtohora capsici on V8 media (glycerol mono-(E)-8,11,12-trihydroxy-9-octadecenoate and V).
[159,185,186]
A.carolinianum DCBulb MongoliaCinnamoylphenethylamine derivativeWeak cytotoxic activity[187]
A.ampeloprasum var. porrum (Leek)Plant parts A-β- d -glucopyranoside Anticancer activity against MCF-7 human breast cancer cell.[187]
A.ascalonicum L. China Ascalonicoside C-(25R)-26-O-β-d-glucopyranosyl-22-hydroxy-5α-furost-2-one-3β,5,6β, 26-tetraol-3-O-α-L-rhamnopyranosyl-(1→2)-β-d-glucopyranoside. Ascalonicoside d-(25R)-26-O-β-d-glucopyranosyl-22-methoxy-5α-furost-2-one-3β,5,6β, 26-tetraol-3-O-α-L-rhamnopyranosyl-(1→2)-β-d-glucopyranoside. (25R)-26-O-β-d-glucopyranosyl-22-hydroxy-5-ene-furostan-3β,26-diol-3-O-α-L-rhamnopyranosyl-(1→4)-α-L-rhamnopyranosyl-(1→4)-[ α-L-rhamnopyranosyl-(1→2)]-β-d-glucopyranoside. 25R)-26-O-β-d-glucopyranosyl-22-hydroxy-5-ene-furostan-3β, 26-diol-3-O-α-L-rhamnopyranosyl-(1→2)-[α-L-arabinofuranosyl-(1→4)]-β-d-glucopyranoside.NR. [188,189]
A.siculumBulbs Zwanenburg, The Netherlands(Z)-Butanethial S-oxide, (R(S),R(C),E)-S-(1-butenyl)cysteine S-oxide (homoisoalliin).NR. [190]
A.chrysanthumBarks Guangzhou, ChinaChrysanthumones A (6″,6″-dimethyl-4″,5″-dihydropyrano [2″,3″: 8,7]-6″′,6″′-dimethyl-prenyl-4″′,5″′-dihydropyrano [2″′,3″′:2′,3′]apigenin) and B
((E)-5,7-dihydroxy-2-(4-hydroxyphenyl)-8-(3-methylbut-1-enyl)-4H-chromen-4-one).
NR.[191]
A. L. melanocrommyum section Megaloprason.Bulbs Central AsiaL-(+)-S-(2-pyridyl)-cysteine sulfoxide. NR. [192]
A.ampeloprasum L.BulbsUnited States of America Ampeloside Bs1 (apigenin 3-O-β-glucopyranosyl (1 → 3)-β-glucopyranosyl (1 → 4)-β-galactopyranoside), ampelosides Bf1 ((25R)-26-O-β-glucopyranosyl-22-hydroxy-5α-furostane-2α,3β,6β,26-tetraol-3-O-β-glucopyranosyl(1 → 3)-β-glucopyranosyl-(1 → 4)-β-galactopyranoside) and Bf2 ((25R)-26-O-β-glucopyranosyl-22-hydroxy-5α-furostane-2α,3β,6β,26-tetraol-3-O-β-glucopyranosyl(1 → 4)-β-galactopyranoside). Weak antifungal activity by ampeloside Bs1.[193]
A.bakeri Reg.Tuber Adenosine, guanosine, and tryptophan, β-sitosterol β-d-glucoside. Strong inhibitory effect on human platelet aggregation generated by 2 μM ADP in both primary and secondary waves (adenosine).[162]
A.victorialis var. platyphyllumAerial parts, bulbsKorea Gitogenin 3-O-lycotetroside, astragalin and kaempferol 3, 4′-di-O-β-d-glucoside. Cytotoxic activity.[194]
A.nutans L.Underground plant parts Deltoside, nolinofuroside D, 25R Δ(5)-spirostan 3β-ol-3-O-α-L-rhamnopyranosyl(1-->2)-[β-d-glucopyranosyl(1→4)]-O-β-d-galactopyranoside and 25R Δ(5)-spirostan 1 β, 3β-diol 1-O-β-d-galactopyranoside.NR.[195]
A.giganteumBulbs Japan 3-O-acetyl-(24S,25S)-5α-spirostane-2α,3β,5α,6β,24-pentol 2-O-β-d-glucopyranoside. Inhibition of cAMP phosphodiesterase activity.[196]
A.hookeri ThwaitesRhizomes China Di-2-propenyl trisulfide, diallyl disulfide, and dipropyl trisulfide. Antimicrobial activity against Aspergillus fumigatus and C. albicans.[197]
NR: not reported.
Table 11. Bioactive compounds isolated from Crinum species.
Table 11. Bioactive compounds isolated from Crinum species.
Plant SpeciesPlant PartCountryIsolated CompoundsBioactivityReferences
C. x amabile
C. x amabile Donn ex Ker Gawl
Bulbs
Stems, roots
Ecuador
Brazil
Thailand
Haemanthamine/crimine-type
alkaloid.
Lycorine-type alkaloid
Galanthamine-type alkaloid.
Augustine N-oxide, buphanisine
N-oxide.
Amabiloid A.
Anticholinesterase
(anti-AChE) and antibutyrylcholinesterase (anti-BuChE) activity.
[249,250,251]
C. defixum Ker-GawlBulbsIndiaHydrazide derivative.
(E)-N-[(E)-2-butenoyl]-2-butenoylhydrazide.
Anti-genotoxic activity.[252]
C. mooreiBulblets Cherylline, crinamidine, crinine, epibuphanisine, lycorine, powelline, undulatine, 1-epideacetylbowdensine, 3-O-acetylhamayne.
3-[4′-(8′-aminoethyl) phenoxy] bulbispermine, mooreine.
NR.[253]
C. biflorumBulbs Senegal5,6,7-trimethoxy-3-(4 hydroxybenzyl) chroman-4-one, 3-hydroxy-5,6,7-trimethoxy-3-(4-hydroxybenzyl) chroman-4-one, 3-hydroxy-5,6,7-trimethoxy-3-(4-methoxybenzyl) chroman-4-one, 5,6,7-trimethoxy-3-(4-methoxybenzyl) chroman-4-one, (E)-N-(4-hydroxyphenethyl)-3-(4-hydroxyphenyl) acrylamide. Anticancer, anti-AChE, anti-glucosidase activity.[254,255]
C. asiaticum
C. asiaticum var. sinicum
C. asiaticum L.
C. asiaticum var. japonicum.
C. asiaticum L. var. sinicum.
Seeds,
rhizome, fruits
Bulbs, stems, leaves
Beijing, China,
Hainan Province,
Japan,
Island of Jeju in
Korea
Flavonoids
Isopowellaminone.
(2R,3S)-7-methoxyflavan-3-ol (1:), (2R,3S)-7-hydroxy-flavan-3-ol (2:), (2R,3S)-2 ′-hydroxy-7-methoxy-flavan-3-ol (3:).
Norgalanthamine.
Crinamine
CAL-n.
Crijaponine A, crijaponine B, ungeremine, lycorine, 2-O-acetyllycorine, 1,2-O-diacetyllycorine, (-)-crinine, 11-hydroxyvittatine, hamayne,(+)-epibuphanisine, crinamine, yemenine A, epinorgalanthamine.
Criasiaticidine A, pratorimine, Lycorine, 4′-hyd’oxy-7-methoxyflavan.
Crinamine, lycorine, norgalanthamine, epinorgalanthamine.
Asiaticumines A, asiaticumines B.
Inhibitory activity against LPS-induced nitric oxide production.
Anticancer activity (against cervical cancer SiHa cells).
Inhibition of platelet aggregation.
Promotion of hair growth through dermal papilla proliferation.
Inhibition of the growth of HepG2 tumor cells.
Anti-AChE activity, cytotoxic activity.
Cytotoxic against Meth-A (mouse sarcoma) and Lewis lung carcinoma (mouse lung carcinoma).
Inhibition of the activity of hypoxia inducible factor-1 (crinamine).
Cytotoxicity.
[256,257,258,259,260,261,262,263,264,265,266]
C. kirkii BakerBulbs Noraugustamine, 4aN-dedihydronoraugustamine, 3-O-acetylsanguinine, 1,2-diacetyllycorine.Antiparasitic activity against Trypanosoma brucei (T. brucei) rhodesiense, Trypanosoma cruzi (T. cruzi).[267,268]
C. macowaniiBulbs Macowine, lycorine, cherylline, crinine, krepowine, powelline, buphanidrine, crinamidine, undulatine, 1-epideacetylbowdensine, 4a-dehydroxycrinamabine.NR.[249]
C. firmifoliumLeaves Madagascar2-alkylquinolin-4(1H), 2-alkylquinolin-4(1H).Antiplasmodial activity.[269]
C. latifoliumBulbs
Leaves
China.
Hanoi, Vietnam
4,8-dimethoxy-cripowellin C. 4,8-dimethoxy-cripowellin D, 9-methoxy-cripowellin B, 4-methoxy-8-hydroxy-cripowellin B, cripowellin C.
C. latines A, C. latines B and C. latines C.
4-senecioyloxymethyl-3,4-dimethoxycoumarin, 5,6,3 ′-trihydroxy-7,8,4 ′-trimethoxyflavone.
4-methyloxysenecioyl-6,7-dimethoxycoumarin, 5,6,3′-trihydroxy-7,8,4′trimethoxyflavone.
Cytotoxic against tumor cell lines, antimicrobial activity, antioxidant activity.
Inhibitory activity against human umbilical venous endothelial cells.
[270,271,272]
C. scillifoliumBulbs Scillitazettine, scilli-N-desmethylpretazettine.Mild antiplasmodial activity[273]
C. zeylanicum (L)Bulbs, leaves, flowers, fruitsCuba
Sri Lanka
Crinine, Lycorine, 11-O-acetoxyambelline, ambelline, 6-hydroxybuphanidrine, 6-ethoxybuphanidrine, 3-acetylhamayne, 6-hydroxycrinamine, hamayne, 6-methoxycrinamine.Antiproliferative effect.[246,274]
C. jagus (J. Thomps) DandyBulbs, leavesSenegal
Ghana
Gigantelline, gigantellinine, gigancrinine, sanguinine, cherylline, lycorine, crinine, flexinine, hippadine.
Galanthamine, galanthamine N-oxide, powelline.
Anti-AChE activity,
inhibitors of TcAchE, hAChE and hBChE
[275,276]
C. abyscinicum Hochst. ExA. RichBulbsEthiopia6-hydroxycrinamine, lycorine.Antiproliferative activity against A2780 epithelial ovarian cancer and MV4-11 acute myeloid leukemia cell lines.[277]
C. erubescensAbove ground plant partsPuntarenas, Costa Rica
Cripowellin A, cripowellin B, cripowellin C, cripowellin D, hippadine.Antiplasmodial activity.[278]
C. yemenseBulbs Yemen6-hydroxy-2H-pyran-3-carbaldehyde.
Yemenines A, B and C, 1, (+)-bulbispermine, (+)-crinamine, (+)-6-hydroxycrinamine, (-)-lycorine.
Tyrosinase inhibitor.
Inhibit nitric oxide production, induce nitric oxide synthase.
[277,278,279,280]
C. bulbispermum
C. bulbispermum III
Bulbs Egypt8-hydroxylycorin-7-one, 2-deoxylycorine, vittatine, 11-hydroxyvittatine, hippamine.
4-hydroxy-2′,4′-dimethoxydihydrochalcone, 4,5-methylenedioxy-4′-hydroxy-2-aldehyde [1,1′-biphenyl], hippacine, 4′-hydroxy-7-methoxyflavan-3-ol, 2(S),3′,4′-dihydroxy-7-methoxy flavan, isolarrien, isoliquiritigenin, liquiritigenin.
Bulbispermine.
NR.[281,282,283]
C. powelliiBulbs Switzerland
Colombia
Linoleic acid ethyl ester, alkaloid hippadine, calleryanin, 4′-hydroxy-7-methoxyflavan.
Lycorine, 1-O-acetyllycorine, ismine.
AChE inhibitor (linoleic acid ethyl ester).
Inhibition of topoisomerase 1 activity.
[284,285]
C. glaucumBulbs NigeriaHamayne, lycorine, haemanthamane, crinamine.Choline esterase inhibitory activity.[286]
C. purpurascensLeavesCameroon4,5-ethano-9,10-methlenedioxy-7-phenanthridone, 4,5-ethano-9-hydroxy-10-methoxy-7-phenanthridone, α-d-glucopyranoside.Antibacterial activity[287]
NR: not reported.
Table 12. Geographical distribution of the Genus Cyrtanthus.
Table 12. Geographical distribution of the Genus Cyrtanthus.
LineageLocationSpeciesReferences
Clade ASouthern Africa Grassland
Southeastern African temperate grasslands
Grassland of the Highveld in the northern parts
Subtropical Indian Ocean Coastal Belt
East Africa and Angola
C. attenuatus, C. macowanii,
C. epiphyticus, C. mackenii subsp. cooperi, C. huttonii, C. macmasteri, C. suaveolens, C. stenanthus var. stenanthus,
C. flanaganii
C. tuckii var. transvaalensis
C. angustifolius, C. fergusoniae
C. aureolinus, C. mackenii subsp. Mackenii, C. brachyscyphus
C. breviflorus
[328]
Clade BBaviaanskloof Mountains and Eastern Cape (Fynbos and Albany Thicket Biomes)
Semi-arid Succulent Karoo
Greater Cape Region (“the Cape”)
Coastal and inland mountains of the southern Cape
Cape Peninsula into the Eastern Cape
C. labiatus, C. montanus
C. herrei
C. carneus, C. elatus, C. guthrieae, C. labiatus, C. leptosiphon, C. leucanthus,
C. montanus, C. odorus
C. collinus
C. ventricosus
[328]
Clade CAlbany Thicket Biome
Savanna Biome
Northwards from the Albany region through South Africa, Zimbabwe, Western Mozambique and East Africa into Sudan
Albany Thicket and Savanna Biomes
Extends beyond the Savanna Biome into the Sub-Escarpment
and Highveld grasslands
Fynbos Biome
Southern parts of the Nama Karoo
C. flammosus, C. spiralis
C. eucallus, C. galpinii
C. sanguineus
C. helictus
C. contractus
C. wellandii
C. smithiae
[328]
Table 13. A summary of the traditional uses, phytochemicals and pharmacological activities of Cyrtanthus species.
Table 13. A summary of the traditional uses, phytochemicals and pharmacological activities of Cyrtanthus species.
Plant SpeciesTraditional UsesCompoundsPharmacological ActivitiesReferences
C. obliquusChronic cough, headache and scrofula5,7-dihydroxy-6-methoxy-3-(4′-methoxybenzyl)chroman-4-one, 5,7-dihydroxy-6-methoxy-3-(4′-hydroxybenzyl)chroman-4-oneAntioxidant activity[338]
C. contractusMental illness, protective charm against evil spiritsNarciclasineNarciprimineAnti-inflammatory activity (via inhibition of E-selectin, blockade of the expression of endothelial adhesion molecule ICAM-1)Acetylcholinesterase inhibitor[337]
C. breviflorusEmesis, worm infestations, protective charm against evil spiritshaemanthamine, lycorine, crinamine hydrochloride and tazettine Antihelminthic[337,342]
C. elatusCough, headache, labor inductionHaemanthamine, zephyranthine, galanthamine and 1,2-O-diacetylzephyranthine Antiprotozoan activity, selective cytotoxic activity[43,44,337]
C. falcatusNot known to be used by the traditional South African peoplePapyramine, epipapyramine, maritidine, O-methylmaritidine and tazettineAntibacterial activity against B. subtilis S. aureus and E. coli, mutagenicity, cytotoxic activity [342,343,344]
C. suaveolensNo traditional use has been reportedCaptanMutagenicity, anti-inflammatory activity via inhibition of COX-2, fungicide[342,344]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Danquah, C.A.; Minkah, P.A.B.; Agana, T.A.; Moyo, P.; Ofori, M.; Doe, P.; Rali, S.; Osei Duah Junior, I.; Amankwah, K.B.; Somuah, S.O.; et al. The Phytochemistry and Pharmacology of Tulbaghia, Allium, Crinum and Cyrtanthus: ‘Talented’ Taxa from the Amaryllidaceae. Molecules 2022, 27, 4475. https://doi.org/10.3390/molecules27144475

AMA Style

Danquah CA, Minkah PAB, Agana TA, Moyo P, Ofori M, Doe P, Rali S, Osei Duah Junior I, Amankwah KB, Somuah SO, et al. The Phytochemistry and Pharmacology of Tulbaghia, Allium, Crinum and Cyrtanthus: ‘Talented’ Taxa from the Amaryllidaceae. Molecules. 2022; 27(14):4475. https://doi.org/10.3390/molecules27144475

Chicago/Turabian Style

Danquah, Cynthia Amaning, Prince Amankwah Baffour Minkah, Theresa A. Agana, Phanankosi Moyo, Michael Ofori, Peace Doe, Sibusiso Rali, Isaiah Osei Duah Junior, Kofi Bonsu Amankwah, Samuel Owusu Somuah, and et al. 2022. "The Phytochemistry and Pharmacology of Tulbaghia, Allium, Crinum and Cyrtanthus: ‘Talented’ Taxa from the Amaryllidaceae" Molecules 27, no. 14: 4475. https://doi.org/10.3390/molecules27144475

Article Metrics

Back to TopTop