Open Access Article
This Open Access Article is licensed under a
Creative Commons Attribution 3.0 Unported Licence

Naturally occurring prenylated flavonoids from African Erythrina plant species

Bienvenu Tsakem a, Fidele Ntie Kang bcd, Rémy Bertrand Teponno e and Xavier Siwe Noundou *a
aDepartment of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa. E-mail: xavier.siwenoundou@smu.ac.za
bCenter for Drug Discovery, Faculty of Science, University of Buea, P.O. Box 63, Buea, Cameroon
cDepartment of Chemistry Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
dInstitute of Pharmacy, Martin-Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle, Saale, Germany
eDepartment of Chemistry, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon

Received 16th May 2025 , Accepted 21st July 2025

First published on 18th August 2025


Abstract

Flavonoids refer to a large class of secondary metabolites with a unique skeleton comprising two aromatic rings linked together by a C3 unit. This class of compounds is largely distributed in plants and rarely occurs in fungi. Some of these compounds are directly bonded to prenyl units. Prenyl groups are C-5 carbon units derived from the mevalonate pathway, are reported to substantially improve the biological activities of diverse classes of secondary metabolites. Prenylated flavonoids display a broad spectrum of biological activities, including antibacterial, antioxidant, anticancer, antidiabetic and antiviral activities. Some flavonoids have already been formulated as either medicines or dietary supplements and are currently used in the management of certain medical conditions. The continuous search for bioactive molecules is a global concern; and encapsulating the contribution of each continent and/or country in terms of available resources should be a priority. This paper aims to methodically summarize the bioactive prenylated flavonoids characterized from plants of the genus Erythrina growing in Africa, as well as their distribution in the genus. Approximately 289 prenylated flavonoids have been isolated and characterized exclusively from plants belonging to the genus Erythrina growing in Africa, covering all the subclasses of flavonoids bearing prenyl group(s), namely, flavanones, flavones, chalcones, isoflavanones, isoflavones, isoflavans, isoflav-3-enes, pterocarpans and pterocarpenes isolated from 1981 to date. This review encompasses the data gathered from 202 peer-reviewed articles and covers the source, isolation, distribution of Erythrina plant species throughout the continent, structure elucidation of prenyl moieties, biological activities as well as the in silico tests where available towards some targets in drug discovery.


image file: d5ra03457d-p1.tif

Bienvenu Tsakem

Bienvenu Tsakem is a PhD student in the Department of Pharmaceutical Sciences at Sefako Makgatho Health Sciences University, South Africa. He obtained his undergraduate degree in Organic Chemistry from the University of Dschang, Cameroon, in 2016. In 2018, he completed a Master's degree in Natural Products Chemistry at the same university. He later joined the Humboldt Research Hub/CECANAPROF at the Higher Teacher Training College (HTTC), University of Yaoundé I, Cameroon, where he worked as a Junior Researcher from 2023 to 2024. His research focuses on the isolation and characterization of natural products from medicinal plants and fungi.

image file: d5ra03457d-p2.tif

Fidele Ntie Kang

Fidele Ntie-Kang is an Associate Professor of Pharmaceutical Chemistry at the Department of Chemistry, University of Buea in Cameroon, where he also heads the Centre for Drug Discovery. He completed a PhD in computer-aided drug design at the University of Douala in Cameroon, then moved to Germany, where he carried out postdoctoral research under Prof. Wolfgang Sippl, leading to a habilitation in pharmaceutical chemistry. He focuses on chemoinformatics for natural products research and is passionate about drug discovery from natural products identified from African medicinal plants. His major contribution is the development of the African Natural Products Database.

image file: d5ra03457d-p3.tif

Rémy Bertrand Teponno

Rémy Bertrand Teponno is Professor of Organic Chemistry at the Department of Chemistry, University of Dschang (Cameroon). He defended his PhD in 2009 and has been a TWAS-DFG fellow (2012), a Matsumae fellow (2014), a Humboldt fellow (2015, 2017, and 2018), and the winner of the TWAS-Atta-Ur-Rahaman Prize 2017. He has been visiting scientist at the University of Camerino (2011) and at the Helmholtz Centre for Infection Research, Braunschweig (2023 and 2024). He is currently the Master/PhD Coordinator at the Department of Chemistry, University of Dschang. His research interest focuses on bioactive secondary metabolites from medicinal plants and fungi.

image file: d5ra03457d-p4.tif

Xavier Siwe Noundou

Xavier Siwe Noundou is a Professor of Pharmaceutical Chemistry at the Department of Pharmaceutical Sciences, Sefako Makgatho Health Sciences University, South Africa. He holds a PhD in Chemistry, University of Johannesburg (2014). He was a CAPES Brazil visiting professor (2023), a GBMF Fellow in the USA (2017–2019), and an EU-FP7 Marie Curie Fellow in Hungary and Bulgaria (2015–2016). His research focuses on drug discovery from terrestrial and marine natural products, their structural derivatization for structure–activity relationship studies, and nano-formulations to improve drug delivery systems. His work bridges natural product chemistry, medicinal chemistry, and nanotechnology in pharmaceutical sciences.


1 Introduction

The search for new hits and leads from any source of secondary metabolites, including plants, fungi, and bacteria, has been a focus of interest for chemists and pharmacists over the last few decades.1–5 There is a continuous need to discover novel and more effective active principles, particularly during the emergence of new diseases and infections. This includes the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the drug resistant phenomena, as well as the existence of diseases without known or effective treatments, such as HIV-AIDS and many forms of cancer.

Natural products such as flavonoids are of paramount importance in drug discovery. Prenylated flavonoids are synthesized by plants as a response to microbial attacks.6 Interestingly, these natural compounds are reported to exhibit improved biological activities compared to the original skeleton without the prenyl groups.7 The increase in biological activities of prenylated flavonoids is generally ascribed to the increase of their lipophilicity due to the presence of prenyl moieties.7 The penetration of prenylated flavonoids into cells is facilitated due to the fact that the cell membrane of microbes is phospholipidic.

While several African species of the Erythrina genus have been extensively investigated, and have resulted in reports of a number of prenylated flavonoids,8,9 there is no available report documenting the significance of prenylated flavonoids in these African plant species from the Erythrina genus, or their distribution within the genus in Africa. There have been two review articles world-wide on non-alkaloidic compounds of the genus Erythrina, published in 2005 (ref. 10) and 2018.11 Further, there is one review article, published in 2021, summarizing all isolated flavonoids in general from any investigated Erythrina species,12 while a recent review in 2023 (ref. 13) reports on prenylated flavonoids from numerous plants, however, this only mentioned 10 species of Erythrina, while about forty species throughout the world have been already studied for their content in prenylated flavonoids.

Considering all the benefits of flavonoids to human health for cancer management, inflammation and immune system boosting, some of these molecules could be beneficial for further medical conditions such as COVID-19, AIDS, breast cancer, to mention a few. Unfortunately, these natural products are usually isolated in small amounts and sometimes biological assays are not conducted. Some flavonoids have just been submitted to in vitro assays, while others have been reported just from one source. Given that all these data are completely dispersed, this review is focussed only on prenylated flavonoids previously isolated from all Erythrina species distributed in Africa. The focus on African Erythrina plants has three objectives. Firstly, any information on the prenylated flavonoids in African Erythrina will be highlighted; secondly, as all information will be contained in a single document, it will facilitate the search for information regarding Erythrina genus in Africa, as shown in Fig. 1, and finally the review highlights the contribution of African plants as a source of potential lead compounds in drug discovery. It is worth mentioning that non-prenylated and prenylated flavonoids are currently used as medicines and dietary supplements.14–16


image file: d5ra03457d-f1.tif
Fig. 1 Distribution of studied Erythrina species in Africa.

The aim of this review is to methodically summarize the bioactive prenylated flavonoids isolated during the period 1981 to 2024 from plants of the genus Erythrina growing in Africa, by providing their source, isolation, distribution, structure elucidation and biological activities, as well as their drug target analysis.

2 Prenylated flavonoids

2.1 Definition

Flavonoids are a group of plant polyphenolic secondary metabolites featuring a common three-ring chemical structure C6–C3–C6 core (diphenylpropane derivatives). They consist of a skeleton containing two benzene rings connected by a C3 moiety. The first C6 ring is called the A ring, the second is called the B ring, and the C3 moiety can either be opened (forming an aliphatic chain) or closed (forming a six-membered ring attached to A ring) and is called the C ring.17 Flavonoids with a C-ring are also called benzopyran.18 Relative to the position of the linkage of the B ring to the benzopyran (chromano) moiety, flavonoids may be divided into thirteen sub-classes grouped as 2-phenylbenzopyrans (flavanones, flavones, chalcones, flavonols, flavononols, flavanes, flavens), isoflavonoids or 3-phenylbenzopyrans (isoflavanones, isoflavones, isoflavanes, isoflavens, pterocarpans, pterocarpens) and neoflavonoids or (4-phenylbenzopyrans) (Fig. 2).18
image file: d5ra03457d-f2.tif
Fig. 2 Common skeletons/subclasses of flavonoids.

2.2 Biosynthesis

Taking into consideration that the biosynthesis of flavonoids in general has been reported,19,20 we focus here on the prenylation, which, while it would seem to be a causal mechanism, there are a lot of specificities within flavonoid subclasses. Depending on the genes encoding for the biosynthesis of flavonoids in plants, some may carry a prenyl on the rings. Shi et al. in 2021 designated prenylated flavonoids as flavonoids that associate one or more prenyl groups linked either by C–C bond(s) or C–O bond(s) to their rings.7 The so-called prenyl groups derive from the dimethylallylpyrophosphate (DMAPP), resulting from the condensation of 3 units of acetyl coenzyme A.21

According to the report of Yazaki and coworkers, membrane-bound prenyltransferases which accept aromatic substrates are sub-divided into two main groups, namely, p-hydroxybenzoate (PHB) prenyltransferases and homogentisate (HG) prenyltransferases. Flavonoid prenyltransferases are derived from the HG prenyltransferase family.22 These enzymes are located in the plastid in plant cells23 and their catalytic action in the presence of a cofactor, Mg2+ is reported to be the best cofactor in these reactions.22

In an enzyme assay, dimethylallyl diphosphate (DMAPP) and the enzyme SfN8DT-1 identified in Sophora flavescens (naringenin 8-dimethylallyltransferase) were found to be specific to flavanone as a substrate.24 A paralogue flavonoid prenyltransferase SfFPT from S. flavescens (93% SfN8DT-1) has been shown to be non-specific only to flavanones, it also catalyzes the prenylation of flavones, flavanonols and even chalcones. It is worth mentioning that the amount of prenylated flavonoid is lower than that of flavanone as a substrate,6,25 and it only inserts a prenyl group at C-8, while when the hydroxyl at 7 is methylated or glycosylated, there is no prenylation.6 This might suggest the participation of this hydroxyl in the biosynthesis. The recombinant indole prenyltransferase 7-DMATS, identified from the fungus Aspergillus fumigatus, catalyzes C-6 prenylation in chalcones, isoflavonoids and flavanones. The enzyme AnaPT, equally identified in Aspergillus fumigatus has shown its ability to prenylate to the C-3′ position of chalcones.26–28

Pterocarpan subclass possesses a specific prenyltransferase which shares 50% of significant similarity with SfN8DTs. The gene has been identified previously in soybeans and was given the name GmG4DT,29–31 or more specifically GmPT20.32 It attaches DMAPP at C-8 of the native pterocarpan substrate, glycinol and the reaction is catalyzed by G4DT, while GmPT20 encodes for the prenylation on C-6 catalyzed by G2DT. Further cyclization reactions in these prenylated pterocarpan are catalyzed by glyceollin synthase (GS) or P450 cyclase.30 The enzyme SfG6DT inserts a prenyl group in genistein on carbon 6 and LaPT1 prenylate the B-rings in isoflavone such as genistein and 2′-hydroxygenistein. The chalcone-specific prenyltransferase SfiLDT has been shown to prenylate isoliquiritigenin.6 The prenyltransferase GuA6DT identified from Glycyrrhiza uralensis specifically introduces a prenyl group at position 6 of flavones; further studies by these authors showed no prenylated derivative in flavonol and flavone, with no hydroxyl groups at C-5 and C-7 or methoxyl group at C-7. Fig. 3 shows the biosynthetic routes of prenylations.


image file: d5ra03457d-f3.tif
Fig. 3 Biosynthetic routes of prenylation of flavonoids.

2.3 Importance of flavonoids in drug discovery

Several products containing natural molecules, usually referred to as dietary supplements, are progressively integrated in drug markets. Numerous medical conditions are of concern currently despite ongoing efforts in drug discovery and development. Synthetic molecules,33,34 natural products from plants and microoganisms1,5,35,36 have been at the centre of interest. Ebselen is an approved synthetic drug used as an antioxidant and anti-inflammatory and as a cytoprotective agent, however the same molecule has demonstrated toxicity towards normal cells.37 This is just one example amongst others, and this is the reason why some researchers in drug discovery focus on natural products. Numerous molecules from plants are currently used for their therapeutic properties worldwide, which include but are not limited to alkaloids, flavonoids, amino acid derivatives and terpenoids.38–41

Some reviews have already reported the implication of flavonoids in cancer evolution and their effects on the nervous system, as well as their antioxidant and anti-inflammatory effects.38,42–44 For example, three prenylated flavonoids i.e. glabridin, tephropurpurin, and 8-prenylnaringenin have been reported for their benefits in the management and prevention of cancers. Glabridin inhibits the activity of the enzyme CYP3A4 (Cytochrome P450 3A4), the largest class of CYP enzymes. This enzyme is expressed in the human liver and gastrointestinal tract and is involved in the metabolism of 50% of therapeutic agents, as well as in the activation of toxic and carcinogenic substances.42 This molecule is sold as a dietary supplement to lighten the skin, and is equally marketed as an anti-inflammatory, antibacterial and pro-apoptotic drug worldwide.45 Tephropurpurin induces the activity of NAD(P)H:quinone oxidoreductase, which results in the detoxification of carcinogens. The flavonoid 8-prenylnaringenin inhibits the enzyme CYP1A2 (Cytochrome P450 1A2) which mainly metabolizes important drugs such as phenacetin, theophylline, caffeine, imipramine and propranolol, and also converts some procarcinogens into carcinogens.42 Dietary supplements labelled as citrus bioflavonoids made of rutin and ascorbic acid, are consumed to boost the immune system; Nutrivein tart cherry® is marketed as a dietary supplement to relieve pains and for muscle recovery.45 Other dietary supplements include tart cherry®, Lipo-flavonoid plus®, Super flavonoids herbal supplement, Ester-C and flavonoids®, citrus bioflavonoids complex® and Bio-flavonoids®.45

Many other flavonoids such as quercetin, rutin, naringerin, equol and baicalein have been documented for their inhibition and induction effects on different enzymes contributing to cancer development. Lemos et al. reported in 2006 the vasorelaxant activity of floranol, a prenylated flavonoid from the roots of Dioclea grandiflora.46 Xanthohumol is a prenylated chalcone from Humulus lupulus L.; it is the active principle of the dietary supplement equally named Xanthohumol®. This molecule helps to fight oxidative stress and maintain cells in optimal health, promotes drowsiness and calms nervous tension and agitation, as well as supporting hormone balance.47

The flavonoids diosmin and hesperidin were isolated from Agathosma betulina, a South African plant and are also found in Citrus reticulata and Hyssopus officinalis.16 These flavonoids are used in the management of cholera, prostatitis, fever and many other conditions.48 These two molecules are active principles of the medicine sold under the name Diosmin Hesperidin® to treat pain and bleeding of haemorrhoids, chronic venous disease and chronic lymphedema. They are also sold as multivitamins and characterized by their high bioavailability.14–16 Some of the previous active ingredients from plants are summarized in Fig. 4.


image file: d5ra03457d-f4.tif
Fig. 4 Some flavonoids approved and marketed as drugs or dietary supplements.

The genus Erythrina has been investigated for prenylated flavonoid since 1981 and these investigations are still ongoing. Prenylated flavonoids represent one of the major classes of secondary metabolites emanated from those studies. As mentioned above, a number of prenylated flavonoids have been studied for their health benefits, and these studies resulted in the formulation of several drugs marketed as dietary supplements. Therefore, summarizing previous studies on the prenylated flavonoids content of Erythrina plants is of paramount importance. No report had focussed on this subclass of secondary metabolites before from the species Erythrina. The intention of this work is to attract the attention of pharmacists/pharmaceutics scientists on Erythrina prenylated flavonoids distributed in Africa.

3 Extraction and isolation

A total of twenty plant species of the Erythrina genus were collected and investigated within African countries (Table 1). It emerges from Table 1 that most chemical studies conducted on the Erythrina species have been in Cameroon, followed by Nigeria, Kenya and Botswana.
Table 1 African countries where studied Erythrina species were collected
Species investigated Country(ies) of origin and references
Erythrina caffra South Africa8
Botswana49
Egypt50
Erythrina mildbraedii Cameroon51–55
Nigeria56
Erythrina vogelii Cameroon57
Ivory coast58
Nigeria59
Erythrina sigmoidea Cameroon60–64
Nigeria65–67
Erythrina eriotricha Cameroon68
Erythrina indica Nigeria59,69
Erythrina lysistemon Cameroon70
Egypt71
Botswana72
Erythrina sacleuxii Kenya73–76
Erythrina addisoniae Cameroon77,78
Ghana79–81
Erythrina latissima Botswana82,83
South Africa84
Erythrina burana Ethiopia85
Erythrina abyssinica Botswana86
Kenya87–89
Uganda90–92
Erythrina livingstoniana Botswana93
Erythrina brucei Ethiopia94
Erythrina burttii Kenya95
Erythrina droogmansiana Cameroon96
Congo97
Erythrina senegalensis Cameroon98–101
Nigeria102–106
Mali107
Erythrina excels Cameroon9,108
Kenya109
Erythrina melanacantha Kenya110,111
Erythrina schliebenii Tanzania112


Maceration appears to have been largely used as an extraction method for the plants reported in this review. Several solvents such as MeOH, EtOH, EtOAc, Acetone, CHCl3, and CH2Cl2 were used. While generally a single solvent was used, some authors combined two solvents to perform their extraction. The commonly used combinations were CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1), and CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1), EtOH[thin space (1/6-em)]:[thin space (1/6-em)]H2O (2[thin space (1/6-em)]:[thin space (1/6-em)]1). With regards to the studied plant parts, mostly stem barks were investigated (Fig. 5).


image file: d5ra03457d-f5.tif
Fig. 5 Extracting solvents (A) and main studied plant parts (B) for the plants of Erythrina genus.

The description follows a specific trend, ranging from prenylated flavanones, flavones, chalcones, isoflavanones, isoflavones, isoflavans, isoflav-3-enes to pterocarpans and pterocarpenes. The structures are also organised in such a way that similar structures within the same subclass are gathered together independently of the species from which they were isolated.

3.1 Prenylated flavanones

Prenylated flavanones have been largely isolated from Erythrina plant species growing in Africa. They represent about 31% of compounds reported herein. The roots of E. abyssinica were extracted with MeOH, and chemical investigation of the extract led to the isolation of five flavanones, abyssinone I (1), abyssinone II (2), abyssinone III (3), abyssinone IV (4), and abyssinone V (5).113 Three flavanones, namely, sigmoidin A–C (6–8) were obtained from the chloroform extract of E. sigmoidea stem bark.60,64,114 Further investigation on the same plant materials led to the discovery of sigmoidin D (9).61 During continuous studies on the same plant, sigmoidin E (10) and sigmoidin F (11) were also isolated.62,115 Sigmoidin G (12), and eriotrinol (13) were reported from the EtOAc extract of E. sigmoidea stem bark.63 Unfortunately, no configuration was indicated for the stereocenter C-2, but according to Promsattha et al. (1989), the C-ring is behind the plane due to the high coupling constant value (J = 12.0 Hz).115 Furthermore, the hydroxyl groups on the heterocyclic ring possess either (R,S) or (S,R) configurations due to the value of the coupling constant reported by the authors. Another two prenylated flavanones, namely, sigmoidin L (14), and 3′-prenylnaringinin (15), were obtained from the EtOAc soluble fraction of E. sigmoidea stem bark.116 The configuration of the stereocenter was deduced according to the value of the coupling constant (J = 13.6 Hz) and the minimal interactions from the chair conformations. A year later, other sigmoidin derivatives, sigmoidins M (16) and N (17), were characterized from the MeOH extract of the stem bark of the same plant.66 From the stem bark MeOH extract of E. abyssinica, four prenylated flavanones were isolated, with their structures elucidated as abyssinone V 4′-methyl ether (18), abyssinoflavanones IV (19), V (20), and VI (21)87 (Fig. 6).
image file: d5ra03457d-f6.tif
Fig. 6 Prenylated flavanones from Erythrina plants.

Erythrisenegalone (22) was isolated from the chloroform extract of the stem bark of E. senegalensis105 while a prenylated flavanone named senegalensein (23) was isolated from the CHCl3 extract of the same plant.102 Similarly, 3′-prenylnaringenin (24) was isolated from the chloroform extract of the stem bark of E. eriotricha.117 Ichimaru and coworkers investigated the chemical constituents of the methanol crude extract of the stem bark of E. abyssinica and obtained three new prenylated flavanones, namely, abyssinin I (25), II (26), and III (27).89 The acetone extract of the stem bark of E. caffra Thunb was subjected to chemical studies and yielded burttinone (28).8 Two prenylated flavanones, i.e., erycaffra D (29), and erycaffra F (30), were isolated from the EtOAc extract of the stem bark of E. caffra.118 From the root bark using an EtOAc extract of E. mildbraedii, two flavanones were isolated, abyssinone IV-4′-O-methyl ether (31) and 7-hydroxy-4′-methoxy-3′-(3-hydroxy-3-methyl-trans-but-1-enyl)-5′-(3-methylbut-2-enyl)flavanone (32).51 Ali et al., in 2012 reported one previously undescribed prenylated flavanone, trivially named mildbone (33), from the methanol extract of E. mildbraedii roots.54 The flavanone licoflavanone-4-O-methyl ether (34) was obtained from the EtOAc crude extract of the root bark of E. mildbraedii.52 Structures 26–34 are reported in Fig. S1 (ESI data).

A biflavanone named bis-sigmodiol (35) and a flavanone isobavachin (36) were isolated and characterized from the MeOH extract of the stem bark of E. sigmoidea,119 in addition to 8-prenyl-7,3′,4′-trihydroxyflavanone (37), sigmone (38) and sigmotriol (39).67 Unfortunately, the structure of bis-sigmodiol (35) was not fully characterized,119 as the configuration of the stereocenter in the prenyl moieties was not determined. However, as the authors declared that the optical rotation was close to zero and used it to support the dimerization, it is possible that it might be the enantiomeric mixture of both monomers. These monomers have been isolated and characterized as brosimacutins A and B.120 Erylatissin C (40) was isolated82 from the stem wood of E. latissima, extracted with the mixture CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1).121 Wanjala and Majinda investigated the CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of E. latissima stem bark which led to the isolation and characterization of 4′,5,7-trihydroxy-3′-methoxy-5′-prenylflavanone (41) and 4′,5,7-trihydroxy-3′,5′-diprenylflavanone (42).83 Two flavanones, named abyssinoflavanone VII (43), and 5-deoxyabyssinin II (44), were obtained from the MeOH extract of the stem bark of E. abyssinica122 (Fig. 6).

From the methanol extract of the stem bark of E. abyssinica, twelve unreported flavanones were characterized, including 2(S)-5,5′,7-trihydroxy-2′-prenyl-(2′′,2′′-dimethylpyrano)-(5′′,6′′:3′,4′)flavanone (45), 2(S)-5,5′,7-trihydroxy-[2′′-(5′′-hydroxy)-methylpyrano]-(5′′,6′′:3′,4′)flavanone (46), 2(S)-5,7-dihydroxy-3′-methoxy-[2′′-(5′′-hydroxy)-methylpyrano]-(5′′,6′′:3′,4′)flavanone (47), 2(S)-5,7-dihydroxy-[(5′′,6′′:3′,4′)-(2′′,2′′-dimethylpyrano)-(5′′′,6′′′:5′,6′)]-(2′′′,2′′′-dimethylpyrano)flavanone (48), 2(S)-5,7-dihydroxy-5′-prenyl-[2′′,2′′-(3′′-hydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (49), 2(S)-5,7-dihydroxy-5′-methoxy-[2′′,2′′-(3′′-hydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (50), 2(S)-5,7-dihydroxy-[2′′,2′′-(3′′,4′′-dihydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (51), 2(S)-5,7-dihydroxy-5′-prenyl-[2′′,2′′-(3′′,4′′-dihydroxy)-dimethylpyrano)]-(5′′,6′′:3′,4′)flavanone (52), 2(S)-5,6′,7-trihydroxy-5′-prenyl-[2′′,2′′-(3′′,4′′-dihydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (53), 2(S)-5,5′,7-trihydroxy-[2′′,2′′-(4′′-chromanone)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (54), 2(S)-5′,7-dihydroxy-[2′′,2′′-(3′′-hydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (55) and 2(S)-5′,7-dihydroxy-[2′′,2′′-(3′′,4′′-dihydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (56)90 (Fig. 6).

The chemical study of the methanol extract of E. abyssinica stem bark led to the isolation and characterization of six unknown flavanones identified as (2S)-5,7-dihydroxy-3′-prenyl-2′′-(4′′-hydroxyisopropyl)dihydrofurano[1′′,3′′:4′,5']flavanone (57), (2S)-5,7-dihydroxy-3′-methoxy-2′′-(4′′-hydroxyisopropyl)dihydrofurano[1′′,3′′:4′,5']flavanone (58), (2S)-5,7,3′-trihydroxy-2′′-(4′′-hydroxyisopropyl)dihydrofurano[1′′,3′′:4′,5']flavanone (59), (2S)-5,7-dihydroxy-3′-prenyl-3′′-hydroxy-dihydrofurano[1′′,3′′:4′,5']flavanone (60), (2S)-5,7,3′-trihydroxy-2′′-(4′′-hydroxyisopropyl)-3′′-hydroxy-hydrofurano[1′′,3′′:4′,5']flavanone (61) and (2S)-5,7,3′-trihydroxy-2′-prenyl-2′′-(4′′-hydroxyisopropyl)-3′′-hydroxy-dihydrofurano[1′′,3′′:4′,5']flavanone (62).91 A total of nine prenylated flavonoids, namely, erylatissins D, E, G (63–65), dihydroabyssinin I (66), 3′4′-dihydro-3′-hydroxy-8′-methylether of sigmoidin C (67), 4′-O-methylsigmoidin B (68), abyssinoflavone IV (69) and V (70), were obtained from the crude MeOH extract of E. latissima stem bark.84 The chemical investigation of the CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of twigs and roots of E. abyssinica resulted in the isolation of one unknown flavanone, abyssinone VII (71).86 From the EtOAc crude extract of E. abyssinica root bark, several compounds, including 7-hydroxy-2-[4-methoxy-3-(3-methylbut-2-enyl)phenyl]chroman-4-one (72), erythribyssin G (73) and erythribyssin I (74)123 were reported. The EtOAc extract of the stem bark of E. livingstoniana was chemically investigated and led to the isolation of four prenylated flavanones, erylivingstone B (75), erylivingstone C (76), 5,7-dihydroxy-3′,4′-dimethoxy-5′-prenylflavanone (77) and 7,3′-dihydroxy-4′-methoxy-5′-prenylflavanone (78).93 Additionally, these authors investigated the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the twigs of the same plant and characterized two new flavanones, namely, 5,7,3′-trihydroxy-5′-(2,3-dihydroxy-3-methylbutyl)-4′-methoxy flavanone (79) and 5,7-dihydroxy-5′-(2,3-dihydroxy-3-methylbutyl)-3′,4′-dimethoxyflavanone (80).121 An investigation of the acetone extract of the stem bark of E. burttii led to the isolation of burttinonedehydrate (81).124 4′-Methoxylicoflavanone (82) was isolated from the EtOAc extract of roots bark of E. droogmansiana,125 while lupinifolin (83) was obtained from the combined hexane and CH2Cl2 extracts of the stem bark of E. excelsa.109 Three prenylated flavanones were isolated from the CH2Cl2 extract of stem bark from E. melanacantha ssp. melanacantha Taub. ex Harms, yielding glabranin (84), citflavanone (85) and exiguaflavanone (86).110 The chemical study of the CH2Cl2 extract of E. addisoniae stem bark led to the isolation of two flavanones, namely 2S-3′-(2-hydroxy-3-methylbut-3-enyl)licoflavone-4′-methyl ether (87) and 2S-3′-(2-hydroxy-3-methylbut-3-enyl)abyssinone II (88).80 Four flavanones were reported from the CH2Cl2 extract of the stem bark of E. addisoniae. These compounds included addisoniaflavanone I (89), II (90), and III (91) as well as 5,7-dihydroxy-5′-prenyl-[2′′,2′′-(3′′-hydroxy)-dimethylpyrano]-(5′′,6′′:3′,4′)flavanone (92)81 (Fig. 6). Structures 65–75 are found in Fig. S1 (ESI).

3.2 Prenylated flavones

From the crude MeOH extract of the stem bark of E. latissima 5′-prenylpratensein A (93) was characterized (Fig. 6).84 The chemical study of the ethanol leaf extract of the medicinal plant E. vogelii led to the isolation and characterization of one unreported flavone, vogeol (94), as well as known compounds carpachromene (95) and 5,7,4′-trihydroxy-6-prenylflavone (96).57 One new flavone, vogelin J (97), was characterized from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the stem bark of E. vogelii.126 The study of the MeOH extract of E. sigmoidea stem bark afforded atalantoflavone/limonianin (98) and neocyclomorusin (99)99 (Fig. 7).
image file: d5ra03457d-f7.tif
Fig. 7 Prenylated flavones from Erythrina plants.

3.3 Prenylated chalcones

The chemical investigation of the MeOH extract of root bark of E. abyssinica led to the isolation of abyssinone VI (100).113 From the EtOAc extract of the root bark of E. mildbraedii, one chalcone named abyssinone VI-4′-O-methyl ether (101) was obtained.51 In addition, mildbenone (102) was reported from the methanol extract of the roots of the same plant.54 Isobavachalcone (103) was isolated from the CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the stem wood of E. latissima.82 Licoagrochalcone A (104) is a prenylated chalcone obtained from the CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of twigs and roots of E. abyssinica.86 Compound 5-prenylbutein (105) was isolated from the EtOAc extract of the stem bark of the same plant.127 Further investigation on the MeOH extract of stem bark of the same plant also led to the discovery of four chalcones, i.e., abyssinone A–D (106–109)128 (Fig. 8).
image file: d5ra03457d-f8.tif
Fig. 8 Prenylated chalcones from Erythrina plants.

3.4 Prenylated isoflavanones

The isoflavanone eriotrichin B (110) was reported from the CH2Cl2 extract of the root bark of E. eriotricha. Its configuration at C-3 was not determined.129 The chemical investigation of the EtOAc extract of the stem bark of E. caffra led to the isolation of erycaffra A (111), erycaffra B (112) and lysisteisoflavanone (113) (configurations at C-3 were not determined).49,130 The chemical study of the CH2Cl2 extract of the roots of E. vogelii yielded two isoflavanones identified as vogelins A (114) and B (115).58 Another derivative, Vogelin D (116) was discovered from the CH2Cl2 crude extract of the root bark of the same plant.131 Another prenylated isoflavanone is 2,3-dihydroauriculatin (117) characterized from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the stem bark of E. vogelii.126 One isoflavanone, sigmoidin I (118) was reported from the MeOH crude extract of E. sigmoidea root bark,129 as well as its congener named sigmoidin J (119).132 (R)-saclenone (120), was obtained from the CH2Cl2 extract of the stem bark of E. sacleuxii.74 The investigation of the methanol extract of the stem bark of E. addisoniae led to the discovery of 5,2′,4′-trihydroxy-6-(γ,γ-dimethylallyl)-2′′′,2′′′-dimethyldihydropyrano[5′′′,6′′′]isoflavone/hydroxyosajin (121) and orientanol E (122).77 Unfortunately, the configuration of the stereogenic center of 122 was not indicated since its first isolation.133 This underscores the needs for continuous investigations on the Erythrina species to close some gaps encountered in the literature. The study of the chloroform extract of the leaves of E. lysistemon afforded 2′,5,7-trihydroxy-4-methoxy-5-prenylisoflavanone (123).72 The MeOH crude extract of the root bark of E. addisoniae afforded erythraddisons III and IV (124, 125).78 From the EtOAc crude extract of the root bark of E. abyssinica, erythribyssin E (126), erythribyssin J (127) and prostratol C (128), were isolated.123 The stem bark extract of E. brucei led to the characterization of bruceins A (129) and B (130), along with kenusanone F (131), 7-methylkenusanone F (132), and sophoraisoflavanone A (133).94 Two isoflavanones, erydroogmansin A (134) and erypoegin D (135), were isolated from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the root bark of E. droogmansiana.97 Sigmoidin H (136) was isolated from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the roots of E. senegalensis.108 The phytochemical investigation of the MeOH extract of the same plant led to the isolation of two new isoflavanones, erysenegalenseins B (137) and C (138) (Fig. 9). Unfortunately, the configurations of stereocenters of those compounds were not indicated.134 Structures 120–138 are incorporated in Fig. S1 (ESI).
image file: d5ra03457d-f9.tif
Fig. 9 Prenylated isoflavanones from Erythrina plants.

3.5 Prenylated isoflavones

This subclass represents 28% of prenylated flavonoids derived from Erythrina species distributed throughout Africa. The isoflavones 8-prenylluteone (139), 6,8-diprenylorobol (140), scandenone (141) and auriculasin (142) were isolated from the chloroform extract of the stem barks of E. eriotricha.135 Eriotriochin (143) was obtained from the chloroform extract of the stem bark of the same plant.136 Further investigations on the same species led to the isolation of 5,4′-dimethoxy-3′-prenylbiochanin A (144).137 The first glycosylated prenyl isoflavone auriculatin 4′-O-glucoside (145) and 8-prenylerythrinin C (146) were characterized from the chloroform extract of the stem bark of E. eriotricha.138 4′-O-Methylalpinumisoflavone (147) is one of the secondary metabolites reported from the EtOAc extract of E. sigmoidea stem bark.63 The acetone extract of the stem bark of E. caffra afforded two isoflavones, alpinumisoflavone (148) and 6,8-diprenylgenistein (149).8 Erysenegalensein D (150) and erysenegalensein E (151) are prenylated isoflavones obtained from the MeOH extract of the stem bark of E. senegalensis.139 The chemical study of the CH2Cl2 crude extract of the stem bark of the same plant led to the characterization of four prenylated isoflavones named auriculatin (152), erysenegalensein O (153), erysenegalensein N (154) and derrone (155)100,101 (Fig. 10).
image file: d5ra03457d-f10.tif
Fig. 10 Prenylated isoflavones from Erythrina plants.

Two epoxy isoflavones reported as erysenegalensein F (156) and erysenegalensein G (157) were isolated from the MeOH extract of E. senegalensis stem bark.140 Erysenegalensein L (158) and erysenegalensein M (159) were obtained from the MeOH extract of seeds of the same plant.139 Erycaffra C (160), isoerysenegalensein E (161), isosenegalensein (162), erythrinin C (163) and erysubin B (164) were obtained from the stem bark EtOAc extract of E. caffra.49,87 Another study was later conducted by the same authors on the EtOAc extract of stem bark of E. caffra to yield laburnetin (165).130 Tchokouaha and coworkers reported several prenylated isoflavones from the dichloromethane extract of the stem bark of E. mildbraedii. These compounds comprise two previously unreported derivatives named erymildbraedins A (166) and B (167) (Fig. S1, ESI) as well as 5,4′-dihydroxy-2′-methoxy-8-(3,3-dimethylallyl)-2′′,2′′dimethylpyrano[5,6[thin space (1/6-em)]:[thin space (1/6-em)]6,7]isoflvone (168) and eryvarin B (169).53 Three isoflavones 2′,7-dihydroxy-4-methoxy-5′-prenylisoflavone (170), erythrinin B (171), and parvisoflavone B (172), were reported from the EtOAc crude extract of E. mildbraedii root bark.52 The chemical study of the ethanol extract of the leaves of E. vogelii led to the isolation of vogeliiol (173), euchrenone b10 (174) and 5,4′-dihydroxy-8-(3′′-methylbut-2′′-enyl)-2′′′-(4′′′-hydroxy-4′′′-methylethyl)-furano-[4′′′,5′′′:6,7] isoflavone (175)57 (Fig. S1, ESI).

From the roots of E. vogelii, two isoflavones were isolated with their structures being elucidated as vogelin C (176) and isowighteone (177).58 Three unreported isoflavones, vogelins E (178), F (179) and G (180), in addition to five reported isoflavones, namely isolupalbigenin (181), ficuisoflavone (182), ulexone A (183), isochandalon (184) and isoderrone (185) were obtained from the CH2Cl2 crude extract of E. vogelii root bark.131 Two new isoflavones named vogelins H (186) and I (187) were characterized from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of stem bark of the same plant.126 Corylin (188), and neobavaisoflavone (189) were isolated from the MeOH crude extract of E. sigmoidea root bark.129 One isoflavone, lupiwighteone (190), was isolated from the MeOH extract of the stem bark of the above mentioned plant.119 Two isoflavones, fleminphilippinin B (191) and 8-prenyldaidzein (192), were isolated from the EtOAc extract of E. sigmoidea stem bark.68 Two isoflavones, indicanine C (193) and 5,4′-di-O-methylalpinumisoflavone (194) were reported from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH extract of the roots bark of E. indica.59 The novel isoflavones indicanines D (195) and E (196) along with wighteone (197) were isolated from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the stem bark of E. indica. Compound 195 featured an alkane chain containing 26 carbon atoms attached to the hydroxyl at position 4′.69 Four isoflavones erysacleuxins A (198) and B (199), 5′-prenylpratensein (200), and 3′-prenylbiochanin A (201) were reported from both the chloroform and EtOAc extracts of E. sacleuxii stem bark.73,76 The unreported isoflavonoid 5-deoxy-3′-prenylbiochanin A (202) along with the known erysubin F (203) were isolated and characterized from the acetone extract of the root bark of E. sacleuxii.75 One isoflavone, 2,3-dehydrokievitone (204), was obtained from the CH2Cl2 extract of the stem bark of the same plant74 (Fig. 10).

The chemical study of the methanol stem bark extract of E. addisoniae led to the isolation of warangalone 4′-methyl ether (205).77 The EtOAc extract of the twigs of E. lysistemon yielded 4′,7-dihydroxy-2′′,2′′-dimethylpyrano [5′′,6'':5,6]-isoflavone (206) and 4′,5,7-trihydroxy-6-(2′′-hydroxy-3′′-prenyl)isoflavone (207).72 The MeOH crude extract of the root bark of E. addisoniae led to the isolation of erythraddisons I (208) and II (209).78 Erylatissin F (210) and glycyrrhizoflavone (211) were isolated from the crude MeOH extract of E. latissima stem bark.84 The chemical investigation of the CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the twigs and roots of E. abyssinica resulted in the identification of semilicoisoflavone B (212).86 However, 212 might also result from the direct Claysen cyclization of the prenyl group at C-3′ and the hydroxyl at C-4′ in 211. The investigation of the acetone extract of the stem bark of E. burttii yielded 7-O-methylluteone (213).124 The isoflavone erydroogmansin B (214) was isolated from the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the root bark of E. droogmansiana.97 Excelsanone (215) was obtained from the EtOH[thin space (1/6-em)]:[thin space (1/6-em)]H2O (8[thin space (1/6-em)]:[thin space (1/6-em)]2) extract of the stem bark of E. excelsa.9

The chemical study of the MeOH extract of the stem bark of E. schliebenii led to the isolation and characterization of schliebenones A (216) and C (217), 5,7-dihydroxy-4′-methoxy-3′-(2,3-dihydroxy-3-methylbutyl)isoflavone (218) and 5′-methoxy-3′-prenylbiochanin A or piscerythrinetin (219), while schliebenone B (220) is a secondary metabolite of the MeOH extract of the root bark of this plant.112 Osajin (221) was isolated from the EtOH[thin space (1/6-em)]:[thin space (1/6-em)]H2O (3[thin space (1/6-em)]:[thin space (1/6-em)]2) extract of the stem bark of E. senegalensis106 (Fig. 10). Structures 205–216 are shown in Fig. S1 (ESI).

3.6 Prenylated isoflavanes and isoflav-3-enes

El-Masry et al. (2010) reported the isolation and characterization of two prenylated isoflavanes, 3S (+) 2′-O-methylphaseollidinisoflavan (222) and 3R (−) erythbidin A (223) from the ethanol extract of the stem bark of E. caffra.50 The isoflavane (3R)-2,7-dihydroxy-3-prenyl-2,2-dimethylpyrano[5,6:4,5]isoflavan (224) was isolated from the EtOAc crude extract of E. mildbraedii root bark.52 Studies of the CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) extract of the root bark of E. livingstoniana led to the characterization of 7-hydroxy-2′-methoxy-[6′′,6′′-dimethylpyrano (2′′,3′′:4′,5′)]isoflavan (225), 7,2′-dihydroxy-[6′′,6′′-dimethyl pyrano (2′′,3′′:4′,5′)]isoflavan (226), 3R 2′-methoxyphaseollinisoflavan (227), and 7,4′-dihydroxy-2′-methoxy-3'-(3-methylbut-2-enyl) isoflavan (228).141 The chemical study of the CH2Cl2 root bark extract of E. lysistemon led to the isolation of two isoflavans, namely, eryzerin C (229), and eryvarin C (230).142 The acetone extract of the root bark of E. burttii Ball.f. were investigated and resulted in three isoflav-3-enes simply named burttinol A–C (231–233)143 (Fig. 11).
image file: d5ra03457d-f11.tif
Fig. 11 Prenylated isoflavanes and isoflav-3-enes from Erythrina plants.

3.7 Prenylated pterocarpans and pterocarpenes

As this represents the third largest group of prenylated flavonoids, this section presents the different prenylated pterocarpans. The roots of E. abyssinica were extracted with methanol, and the study of its chemical contents revealed the isolation of erythrabyssin I, also named cristacarpin (234), erythrabyssin II (235), phaseollin (236) and phaseollidin (237).113 Three new pterocarpans, erybraedins A–C (238–240) and the known isoneorautenol (241) were isolated from the ethanol extract of E. mildbraedii roots.56 Erybraedins D (242) and E (243) were reported from the MeOH extract of the stem bark of E. eriotricha.144 The continuous search of bioactive flavonoids led to the isolation of neorautenol (244).132 El-Masry and coworkers reported one pterocarpan, sandwicensin (3-hydroxy-10-dimethylallyl-9-methoxypterocarpan) (245) from the ethanol extract of E. caffra.50 Compound 1-methoxyphaseollidin (246) was isolated from the CH2Cl2 root extract of E. vogelii.58 A pterocarpan orientanol A (247) was discovered in the MeOH extract of the stem bark of E. sigmoidea.119 Two pterocarpans, gangetinin (248) and calopocarpin (249) were isolated from the EtOAc extract of E. sigmoidea stem bark68 (Fig. 12).
image file: d5ra03457d-f12.tif
Fig. 12 Prenylated pterocarpans from Erythrina plants.

From the methanol extract of the stem bark of E. lysistemon, three novel pterocarpans erylysin A–C (250–252) were reported, conjointly with known orientanol C (253), erysubin D (254), eryvarin D (255) and erystagallin C (256).70 Shinpterocarpin (257) was isolated from the CH2Cl2 extract of the stem bark of E. sacleuxii.74

Sophorapterocarpan A (258), and 6α-hydroxyphaseollidin (259) were isolated and characterized from the MeOH extract of E. sigmoidea stem barks.99 Nguyen and coworkers studied the chemical composition of the MeOH extract of E. abyssinica. They identified two new prenylated pterocarpans, erythribyssin A (260) and C (261), along with five known derivatives identified as eryvarin K (262), 3,9-dihydroxy-4-prenyl-[6aR:11 aR]pterocarpan (263), folitenol (264), erysubin E (265) and erystagallin A (266).145 The stem and root bark of E. brucei were extracted with CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1) and further chemical investigations resulted in the isolation of eryvarin J (267), 2-prenyl-6α-hydroxyphaseollidin (268), and erycristagallin (269).94 The acetone extract of the stem bark of E. burttii led to the isolation of 3-O-methylcalopocarpin (270).124 Erybraedin F (271) was isolated from the CH2Cl2 extract of the stem bark of E. senegalensis.107 The chemical structures 250–271 are reported in Fig. S1 (ESI).

Compound 2′,3′-epoxyhomoedudiol (3-hydroxy-8-(3,3-dimethyl-oxiranylmethyl)pterocarpane (272) was isolated from the CH2Cl2 extract of the stem bark of E. melanacantha.111 The chemical study of the CH2Cl2 root bark extract of E. schliebenii led to the isolation and characterization of 3-hydroxy-10-(2,3-dihydroxy-3-methylbutyl)-9-methoxypterocarpan (273) and orientanol B (274).112 Erythribyssin O (275), erythribyssin L (276), erythribyssin D (277) and erythribyssin M (278) were isolated from the EtOAc extract of the stem bark of E. abyssinica.146 The chemical study of the CH2Cl2 root bark extract of E. lysistemon led to the isolation of nine unreported pterocarpans, namely, (6aR,11aR)-3,9-dihydroxy-4-(γ,γ-dimethylallyl)-10-(2′′-hydroxy-3′′-methylbut-3-enyl) pterocarpan (279), (6aR,11aR)-3,9-dihydroxy-10-(γ,γ-dimethylallyl)-4-(2′′-hydroxy-3′′-methylbut-3-enyl)pterocarpan (280), (6aR,11aR)-2′,2′-dimethylpyrano[6′,5′:3,4]-2′′,2′′ dimethylpyrano[6′′,5′′:9,10]pterocarpan (281), (6aR,11aR)-3,9-dihydroxy-10-(γ,γ-dimethylallyl)-2′-hydroxyisopropyl dihydrofurano[5′,6′:3,4]pterocarpan (282), (6aR,11aR)-3-methoxy-9-hydroxy-4,l0-di(γ,γ-dimethylallyl)-pterocarpan (283), (6aR,11aR)-3-Hydroxy-9-methoxy-4,10-di(γ,γ-dimethylallyl)-pterocarpan (284), (6aR,11aR)-9-hydroxy-10-(γ,γ-dimethylallyl)-2′,2′-dimethylpyrano[6′,5′:3,4]pterocarpan (285), (6aR,11aR)-3,9-dihydroxy-4-(γ,γ-dimethylallyl)-2′′-hydroxyisopropyl dihydrofurano[5′′,6′′:9,10]pterocarpan (286) and (6aR,11aR)3-hydroxy-4(γ,γ-dimethylallyl)-2′,2′-(3′′-hydroxy)-dimethylpyrano[6′′,5′′:9,10]pterocarpan (287), as well as the reported 8-methoxyneorautenol (288).142 The pterocarpen sigmoidin K (289) was obtained from the MeOH crude extract of E. sigmoidea root bark132 (Fig. 12).

In these studies of prenylation of flavones from African Erythrina, flavanones tend to orient on the B ring cycle, in comparison to isoflavonoids where the prenylation was oriented on both the A and B rings. This preference in flavone and flavanone might be related to high enzyme AnaPT content in the corresponding plants.26

4 Distribution of prenylated flavonoids in African Erythrina

Considerable chemical studies have been conducted on the plant species of Erythrina genus (Fabaceae) across the world. Data from previous studies lend to the theory that plants of this genus produce mainly flavonoids10–12 and alkaloids.147,148 Prenylated flavonoids are relatively dominant in the genus and this section discusses the occurrence of different subclasses of prenylated flavonoids already reported. Some structural differences have often been noted in compounds isolated from the same genus according to geographical location.149 In ess ence, there is no difference between the skeletons of prenylated flavonoids occurring in African Erythrina species and others worldwide. Focusing the review on African Erythrina is only for the purpose of collecting information regarding studies already published. Several structures reported in this review have been equally reported in species growing in Japan, Brazil and India.150–154 This section therefore collates the information to support the occurrence of prenylated flavonoids within the genus Erythrina. This genus belongs to the Fabaceae family. While the comparison of prenylated flavonoids occurring within the Erythrina genus across the world was not the objective of the present review, during the investigation we did notice the occurrence of isoflavonoids and pterocarpans in the species growing in Asia.150–152,155–157 There is no review differentiating these compounds from species of other continents, and further reviews might focus on this. A review by Wei et al. focussed on all prenylated flavonoids in plants kingdom world-wide, but this review only mentioned ten species of Erythrina with prenylated flavonoids.13 Another review targeting therapeutic prenylated flavonoids from the Fabaceae family did not provide enough information on the Erythrina genus, particularly the species from Africa.158 In the present review, the phytochemistry of twenty species of Erythrina occurring only in Africa is discussed. We limited our investigation only to those species of the genus Erythrina in Africa.

Prenylated flavanones represent the higher percentage (31%) of prenylated flavonoids in African Erythrina, followed by isoflavones (28%) and pterocarpans (19%) (Fig. 13). Within these three subclasses, certain specific compounds have been isolated several times within the genus.


image file: d5ra03457d-f13.tif
Fig. 13 Subclasses of prenylated flavonoids from African Erythrina.

From this summary of prenylated flavonoids isolated and characterized from the plants of the genus Erythrina it appears that abyssinone V (5), sigmoidin A (6), abyssinone V-4′-O-methyl ether (18), warangalone (141), 6,8-diprenylgenistein (149) and phaseollidin (237) can be considered to be their chemical markers. Concerning the subclass of prenylated flavanones, abyssinone V (5) and abyssinone V-4′-O-methyl ether (18) specifically have been reported from more than ten chemical studies within the genus. According to these reported data, twenty-five isoflavones have been isolated and characterized from more than one Erythrina species. Warangalone/scandenone (141), alpinumisoflavone (148), 6,8-diprenylgenistein (149), erysenegalensein E (151), auriculatin (152), neobavaisoflavone (189) and 5′-prenylpratensein (200) are reported here as the chemical markers of the prenylated isoflavones in the genus Erythrina growing in Africa. Specifically, warangalone/scandenone (141), and 6,8-diprenylgenistein (149) were characterized from ten phytochemical investigations in the genus. These isoflavones are largely distributed in the stem barks of various Erythrina species. As noted from these isolations, pterocarpans represent 19% of compounds isolated from the genus Erythrina, with 20 compounds identified in more than one Erythrina species. It emerges from this study that erythrabyssin II (235), phaseollin (236), phaseollidin (237), erybraedin A (238), isoneorautenol (241), cristacarpin (234) and calopocarpin (249) are the chemical markers of Erythrina genus in Africa for the prenylated pterocarpans, with phaseollidin (237) reported in eleven chemical studies. Regarding the location in erythrina plant parts, in general root bark extracts yielded pterocarpans (Table 2). Pterocarpans were mainly isolated from E. abyssinica, E. senegalensis, E. burttii and E. eriotricha, however, these species are distributed across Cameroon, Nigeria, Mali, Kenya and Botswana, and unfortunately this not indicate any specificity regarding their occurrence. The same analysis was deduced from flavanones and isoflavones.

Table 2 Distribution of prenylated flavonoids in Erythrina genus
Isolated compounds Species (part studied) Extract and references
Abyssinone II (2) E. latissima (stem wood) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)82,141
E. abyssinica (root bark) EtOAc123
Abyssinone III (3) E. abyssinica (root bark) EtOAc123
Abyssinone IV (4) E. sigmoidea (stem bark) CHCl3 (ref. 115)
E. mildbraedii (root bark) EtOAc51
E. sigmoidea (stem bark) MeOH99
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. addisoniae (stem bark) CH2Cl2 (ref. 80)
Abyssinone V (5) E. sigmoidea (stem bark) CHCl3 (ref. 62)
E. abyssinica (root bark) EtOAc123
E. eriotricha (stem bark) CHCl3 (ref. 117)
E. abyssinica (stem bark) MeOH89
E. mildbraedii (root bark) EtOAc51
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. melanacantha (stem bark) CH2Cl2 (ref. 110)
E. addisoniae (stem bark) CH2Cl2 (ref. 80)
E. burttii (stem bark) CHCl3 (ref. 159)
Sigmoidin A (6) E. abyssinica (stem bark) MeOH89
E. latissima (stem bark) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)83
E. sigmoidea (stem bark) EtOAc68
E. abyssinica (stem bark) MeOH122
E. latissima (stem bark) MeOH84
Sigmoidin B (7) E. sigmoidea (stem bark) EtOAc33
E. abyssinica (stem bark) MeOH58
E. latissima (stem bark) MeOH84
E. latissima (stem bark) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)83
E. abyssinica (stem bark) MeOH122
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
Sigmoidin C (8) E. eriotricha (stem bark) CHCl3 (ref. 138)
E. abyssinica (stem bark) MeOH89
E. latissima (stem bark) MeOH84
E. abyssinica (stem bark) MeOH122
E. latissima (stem bark) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)83
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
Sigmoidin D (9) E. abyssinica (stem bark) MeOH87
E. latissima (stem bark) MeOH84
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
Sigmoidin E (10) E. mildbraedii (root bark) EtOAc51
E. sacleuxii (stem bark) EtOAc and CHCl3 (ref. 73 and 76)
E. abyssinica (stem bark) EtOAc127
Sigmoidin F (11) E. abyssinica (stem bark) MeOH89
E. abyssinica (stem bark) MeOH122
E. latissima (stem bark) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)83
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. latissima (stem bark) MeOH84
Sigmoidin G (12) E. sigmoidea (stem bark) EtOAc68
Sigmoidin L (14) E. sigmoidea (stem bark) EtOAc68
Abyssinone V 4′-O-methyl ether (18) E. caffra (stem bark) Acetone8
EtOAc118
E. addisoniae (stem bark) CH2Cl2 (ref. 80)
E. schliebenii (stem bark) MeOH112
E. mildbraedii (root bark) EtOAc51
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
E. sacleuxii (stem bark) EtOAc and CHCl3 (ref. 73 and 76)
E. burttii (stem bark) CHCl3 (ref. 143 and 160)
E. burttii (stem bark) Acetone124
E. droogmansiana (root bark) EtOAc96
E. melanacantha (stem bark) CH2Cl2 (ref. 110)
Senegalensein (23) E. senegalensis (stem bark) CHCl3 (ref. 103)
E. caffra (stem bark) EtOAc130
E. indica (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)69
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
E. addisoniae (stem bark) MeOH77
Abyssinin III (27) E. abyssinica (stem bark) MeOH91
E. latissima (stem bark) MeOH84
Burttinone (28) E. caffra (stem bark) EtOAc130
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
E. sacleuxii (stem bark) EtOAc and CHCl3 (ref. 73 and 76)
E. burttii (stem bark) CHCl3 (ref. 143 and 160)
E. burttii (stem bark) Acetone124
E. schliebenii (stem bark) MeOH112
Erycaffra D (29) E. caffra (stem bark) EtOAc130
Erycaffra F (30) E. caffra (stem bark) EtOAc130
Erylatissin C (40) E. abyssinica (stem bark) MeOH91
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
Abyssinoflavanone VII (43) E. addisoniae (stem bark) CH2Cl2 (ref. 80)
5-Deoxyabyssinin II (44) E. abyssinica (root bark) EtOAc92
E. abyssinica (stem bark) EtOAc127
Sigmoidin B-4′-methyl ether (68) E. livingstoniana (stem bark) EtOAc93
E. burttii (stem bark) CHCl3 (ref. 143 and 160)
E. melanacantha (stem bark) CH2Cl2 (ref. 110)
E. burttii (stem bark) CHCl3 (ref. 159)
Carpachromene (95) E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
E. senegalensis (stem bark) CH2Cl2 (ref. 107)
Limonianin (98) E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
Abyssinone VI (100) E. abyssinica (stem bark) MeOH122
Isobavachalcone (103) E. burttii (stem bark) CHCl3 (ref. 143 and 160)
Licoagrochalcone A (104) E. abyssinica (stem bark) EtOAc127
E. abyssinica (stem bark) MeOH122
Eriotrichin B/bidwillon A (110) E. eriotricha (stem bark) MeOH129
E. burttii (stem bark) CHCl3 (ref. 160)
E. sigmoidea (stem bark) MeOH99
E. lysistemon (root bark) CH2Cl2 (ref. 142)
E. burttii (stem bark) CHCl3 (ref. 159)
2,3-Dihydroauriculatin (117) E. addisoniae (stem bark) MeOH77
Sigmoidin I (118) E. sigmoidea (stem bark) MeOH99
Lysisteisoflavanone (113) E. lysistemon (stem bark) CH2Cl2 (ref. 71)
Orientanol E (122) E. lysistemon (root bark) CH2Cl2 (ref. 142)
Sophoraisoflavanone A (133) E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
8-Prenylluteone (139) E. senegalensis (stem bark) MeOH104
E. senegalensis (stem bark) CH2Cl2 (ref. 100 and 101)
E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
Warangalone/scandenone (141) E. senegalensis (stem bark) MeOH140
E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
E. caffra (stem bark) EtOAc130
E. mildbraedii (root bark) EtOAc55
E. mildbraedii (stem bark) CH2Cl2 (ref. 53)
E. vogelii (leaves) EtOH57
E. sigmoidea (stem bark) MeOH65
E. sigmoidea (stem bark) EtOAc68
E. addisoniae (stem bark) MeOH77
E. senegalensis (stem bark) CHCl3 (ref. 105)
Alpinumisoflavone (148) E. senegalensis (stem bark) CH2Cl2 (ref. 100 and 101)
E. caffra (stem bark) EtOAc130
E. indica (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)69
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
E. lysistemon (twigs) EtOAc72
6,8-Diprenylgenistein (149) E. senegalensis (stem bark) MeOH104
E. senegalensis (stem bark) CH2Cl2 (ref. 100 and 101)
E. mildbraedii (root bark) EtOAc51
E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
E. sigmoidea (stem bark) MeOH65
E. sigmoidea (stem bark) EtOAc68
E. lysistemon (twigs) EtOAc72
E. senegalensis (stem bark) CHCl3 (ref. 98)
E. excels (stem bark) EtOH[thin space (1/6-em)]:[thin space (1/6-em)]H2O (8[thin space (1/6-em)]:[thin space (1/6-em)]2)9
E. senegalensis (stem bark) EtOH[thin space (1/6-em)]:[thin space (1/6-em)]H2O (3[thin space (1/6-em)]:[thin space (1/6-em)]2)106
Erysenegalensein E (151) E. caffra (stem bark) EtOAc130
E. indica (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)69
E. indica (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)69
E. lysistemon (twigs) EtOAc72
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
Auriculatin (152) E. senegalensis (stem bark) MeOH140
E. caffra (stem bark) EtOAc49
E. vogelii (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)126
E. senegalensis (stem bark) CHCl3 (ref. 98)
Derrone (155) E. caffra (stem bark) EtOAc130
E. lysistemon (twigs) EtOAc72
Erysenegalensein M (159) E. mildbraedii (stem bark) CH2Cl2 (ref. 53)
Erycaffra C (160) E. caffra (stem bark) EtOAc130
Isoerysenegalensein E (161) E. caffra (stem bark) EtOAc130
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
E. lysistemon (twigs) EtOAc72
Isosenegalensein (162) E. caffra (stem bark) EtOAc130
E. lysistemon (stem bark) CH2Cl2 (ref. 71)
Erythrinin C (163) E. caffra (stem bark) EtOAc130
Erysubin B (164) E. caffra (stem bark) EtOAc130
Parvisoflavone B (172) E. schliebenii (root bark) MeOH112
Euchrenone b10 (174) E. addisoniae (root bark) MeOH78
Vogelin C (176) E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
Isolupalbigenin (181) E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
Corylin (190) E. sacleuxii (root bark) Acetone75
Neobavaisoflavone (189) E. sigmoidea (stem bark) MeOH99
E. latissimi (stem wood) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)82
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. senegalensis (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)108
Dimethylalpinumisoflavone (194) E. indica (stem bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)69
Wighteone (197) E. lysistemon (stem bark) CH2Cl2 (ref. 71)
Erysubin F (203) E. addisoniae (root bark) MeOH78
E. brucei (root bark) CH2Cl2–MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
5′-Prenylpratensein (200) E. latissimi (stem bark) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)83
E. abyssinica (twigs and roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. burttii (stem bark) Acetone124
E. schliebenii (root bark) CH2Cl2 (ref. 112)
3′-Prenylbiochanin A (201) E. schliebenii (stem bark) MeOH112
Erythrabyssin I/cristacarpin (234) E. droogmansiana (root bark) EtOAc125
E. lysistemon (stem bark) MeOH70
E. lysistemon (leaves) CHCl3 (ref. 72)
E. latissimi (stem wood) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)82
E. brucei (root bark) CH2Cl2–MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
E. abyssinica (stem bark) EtOAc146
E. burana (stem bark) CHCl3 (ref. 85)
Erythrabyssin II (235) E. mildbraedii (root bark) EtOH56
E. sigmoidea (root bark) MeOH132
E. abyssinica (twigs, roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. abyssinica (stem bark) EtOAc91
E. brucei (root bark) CH2Cl2–MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
E. burttii (root bark) Acetone143
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Phaseollin (236) E. lysistemon (stem bark) MeOH70
E. burttii (root bark) Acetone143
E. senegalensis (stem bark) CH2Cl2 (ref. 107)
E. melanacantha (stem bark) CH2Cl2 (ref. 111)
E. abyssinica (stem bark) EtOAc146
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Phaseollidin (237) E. sigmoidea (root bark) MeOH129
E. burana (bark) CHCl3 (ref. 85)
E. lysistemon (leaves) CHCl3 (ref. 72)
E. latissimi (stem wood) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)82
E. abyssinica (twigs, roots) CHCl3[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)86
E. abyssinica (stem bark) EtOAc145
E. burttii (root bark) Acetone143
E. droogmansiana (root bark) EtOAc125
E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
E. melanacantha (stem bark) CH2Cl2 (ref. 111)
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Erybraedin A (238) E. eriotricha (stem bark) MeOH129
E. eriotricha (root bark) CH2Cl2 (ref. 129)
E. lysistemon (stem bark) MeOH70
E. burttii (root bark) Acetone143
E. senegalensis (stem bark) CH2Cl2 (ref. 107)
E. melanacantha (stem bark) CH2Cl2 (ref. 111)
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Erybraedin B (239) E. abyssinica (stem bark) EtOAc145
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Erybraedin C (240) E. eriotricha (stem bark) MeOH129
E. eriotricha (root bark) CH2Cl2 (ref. 144)
E. abyssinica (stem bark) EtOAc145
E. senegalensis (stem bark) CH2Cl2 (ref. 107)
Isoneorautenol (241) E. eriotricha (stem bark) MeOH129
E. eriotricha (root bark) CH2Cl2 (ref. 144)
E. lysistemon (stem bark) MeOH70
E. livingstoniana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)141
E. excelsa (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)108
E. melanacantha (stem bark) CH2Cl2 (ref. 111)
E. abyssinica (stem bark) EtOAc146
Erybraedin D (242) E. eriotricha (root bark) CH2Cl2 (ref. 144)
E. abyssinica (stem bark) EtOAc145
E. senegalensis (stem bark) CH2Cl2 (ref. 107)
Erybraedin E (243) E. eriotricha (root bark) CH2Cl2 (ref. 144)
Neorautenol (244) E. abyssinica (stem bark) EtOAc91
E. burttii (stem bark) CHCl3 (ref. 160)
E. schliebenii (root bark) CH2Cl2 (ref. 112)
E. abyssinica (stem bark) EtOAc146
E. burttii (stem bark) CHCl3 (ref. 159)
Sandwicensin (245) E. brucei (root bark) CH2Cl2–MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
Calopocarpin (249) E. lysistemon (stem bark) MeOH70
E. livingstoniana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)141
E. brucei (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
E. burttii (stem bark) CHCl3 (ref. 160)
E. burttii (root bark) Acetone143
E. abyssinica (stem bark) EtOAc146
E. burttii (stem bark) CHCl3 (ref. 159)
Erysubin D (254) E. abyssinica (stem bark) EtOAc146
Eryvarin D (255) E. abyssinica (stem bark) EtOAc146
E. lysistemon (root bark) CH2Cl2 (ref. 142)
E. abyssinica (root bark) Acetone127
Shinpterocarpin (257) E. senegalensis (stem bark) CH2Cl2 (ref. 107)
E. sacleuxii (root bark) Acetone75
E. abyssinica (root bark) Acetone161
Sophorapterocarpan A (258) E. abyssinica (stem bark) EtOAc145
E. abyssinica (stem bark) EtOAc146
E. melanacantha (stem bark) CH2Cl2 (ref. 111)
Eryvarin K (262) E. senegalensis (stem bark) CH2Cl2 (ref. 107)
E. lysistemon (root bark) CH2Cl2 (ref. 142)
Erysubin E (265) E. brucei (root bark) CH2Cl2–MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)94
E. abyssinica (stem bark) EtOAc146
Erystagallin A (266) E. droogmansiana (root bark) CH2Cl2[thin space (1/6-em)]:[thin space (1/6-em)]MeOH (1[thin space (1/6-em)]:[thin space (1/6-em)]1)97
E. abyssinica (stem bark) EtOAc146
Erycristagallin (269) E. mildbraedii (root bark) EtOAc55
E. abyssinica (root bark) Acetone127
E. abyssinica (root bark) Acetone161
E. burttii (stem bark) CHCl3 (ref. 159)


5 Characterization of prenyl moieties

Several spectroscopy methods are commonly used in the characterization of flavonoids, such as Circular Dichroism, Infrared and NMR spectroscopy.162 In general, NMR spectroscopy is helpful in the identification of prenyl moieties; this is fundamentally because the characteristics of prenyl groups in UV and IR spectroscopy interfere with those of the flavonoids. The first noticeable characteristics of prenyl groups in 1H NMR spectra are the chemical shifts of protons of the hydroxyl groups; these signals resonate between 5 and 8 ppm. In 2-hydroxy-3-methylbut-3-enyl the configuration of the double bond is usually trans, as evidenced by coupling constants J = 16 Hz.93 In chromans, furanose or any open prenyl groups containing asymmetric carbon, the wide ranges in chemical shifts could be due to the absolute configurations, but unfortunately these configurations were not all determined. Meanwhile the configuration of C-3′′ in sigmoidin D (9) was determined by using the Horeaus' method with (+) 2-phenylbutanoic anhydride, and was attributed to “S”.61 In addition, the positions of these prenyl moieties relative to aromatic rings can equally affect the chemical shift values. Tables 3 and 4 provide the chemical shift ranges characterizing some commonly occurring prenyl groups.
Table 3 Chemical shift ranges characteristic of open prenyl groups
Prenyl groups Position, δC and δH in ppm References
1 2 3 4 5
image file: d5ra03457d-u1.tif 21–22 121–122 132–136 25–26 17–18 57, 119, 67, 82, 77 and 51
3.15–3.45 5.00–5.3   1.50–1.80 1.50–1.80
image file: d5ra03457d-u2.tif 28–34 74–78 145–150 109–112 17–19 69, 80, 100, 112 and 163
2.80–3.30 4.00–4.50   4.70–5.00 1.75–1.85
image file: d5ra03457d-u3.tif 120–125 135–142 70–83 25–31 25–31 51, 141 and 151
6.80–6.95 6.30–6.60   1.30–1.50 1.30–1.50
image file: d5ra03457d-u4.tif 122–124 133–135 142–44 118–119 18–19 112 and 151
6.70–6.90 6.40–7.00   5.00–5.2 1.8–2.00
image file: d5ra03457d-u5.tif 32 129.5 140 72.4 18.5 131
3.4 5.4   3.95 1.80
image file: d5ra03457d-u6.tif 31 154 141 198 10.5 53
3.6 6.5 9.3   1.8
image file: d5ra03457d-u7.tif 39 148 113.3 27.1 27.1 143
  6.2 5.33 1.4 1.4


Table 4 Chemical shift ranges characteristic of heterocyclic prenyl groups
Prenyl groups Position, δC and δH in ppm References
2 3 4 5 6
image file: d5ra03457d-u8.tif 76–80 127–132 115–123 27–29 27–29 52, 67, 82, 90, 97 and 98
5.40–5.60 6.20–6.65 1.40–1.50 1.40–1.50
image file: d5ra03457d-u9.tif 79–81 76–77 68–70 25–28 19–20 66, 90 and 128
3.50–3.60 4.50–4.60 1.20–1.25 1.40–1.55
image file: d5ra03457d-u10.tif 76–80 69–70 26–33 25–27 20–21 87, 90, 92, 128, 131 and 146
3.65–3.85 2.40–2.80 1.30–1.40 1.10–1.27
image file: d5ra03457d-u11.tif 81.1 49.3 192.2 25.6 25.6 90
2.80 1.48 1.48
image file: d5ra03457d-u12.tif 75–76 32–34 23–24 26–27 26–28 54 and 87
1,80–1.90 2.75–2.85 1.30–140 1.30–140
image file: d5ra03457d-u13.tif 90–92 26–32 71–73 25–27 24–26 57, 91, 126, 138 and 142
4.60–3.75 3.00–3.25 1.25–1.30 1.20–1.27
image file: d5ra03457d-u14.tif 97–98 73–75 71–72 25–27 25–27 91
3.30–3.50 5.10–5.40 1.65–1.72 1.65–1.72


6 Biological activities

Flavonoids represent a major class of natural products from plants, and given that many plant-derived flavonoids are commonly used as food, they are generally considered to be non-toxic to human organisms.164 Therefore, targeting their importance as bioactive ingredients should be beneficial to humans, both as drugs and for food supplements. Recent reports have stated that the substitution of the flavonoid ring system by a prenyl side chain confers a strong affinity to biological membranes to the molecule due to the increase of their lipophilicity.165

6.1 Antimicrobial activity

Some reported data have suggested that flavonoids mainly interact with the cell membranes of Gram-positive bacteria. These suggestions were supported by the study of Juang et al.; the lipophilicity might be a key factor which increases the activity against Gram positive bacteria.6 However, it is reported that some modifications such as carbonylation, hydroxylation and methoxylation and/or cyclisation of the prenyl and/or geranyl side chains negatively impact the activity.166

Abyssinone V 4′-O-methyl ether (18), 6,8-diprenylgenistein (149), alpinumisoflavone (148) and burttinone (28) were assayed for their effect on bacteria growth using the microdilution method. Results showed that 18 and 148 had significative antibacterial activity on E. coli ATCC 11775 and K. pneumonia, with the same MIC value of 3.9 μg mL−1 (9.24 and 11.6 μM, respectively) compared to the positive control neomycin (MIC = 1.6 μg mL−1, 2.6 μM). Alpinumisoflavone (148) equally showed the same activity against S. aureus (neomycin MIC = 0.8 μg mL−1, 1.3 μM) whereas 6,8-diprenylgenistein (149) exhibited an activity of 7.8 μg mL−1 (19.2 μM) towards the growth of S. aureus ATCC 12600, E. coli, K. pneumonia (ATCC 13883) and 15.6 against B. subtilis ATCC 6051. The positive control was in the range 0.78 to 1.6 μg mL−1. B. subtilis was mainly affected by compound 149, only with a MIC of 7.8 μg mL−1 (19.2 μM) relative to the MIC value of 1.3 μM exhibited by neomycin. Given that compound 28 was the less active (31 < MIC < 125 μg mL−1), the hydroxylation of the prenyl group at 3′ could have been more negatively affected than 148 which has no hydroxyl on its prenyl groups.8 Sandwicensin (245) and 3R (−) erythbidin A (223) when evaluated for their effect on S. aureus ATCC6538P both showed a MIC value of 62.5 μg mL−1 (183.8 μM) (positive control not indicated).50 Abyssinone VII (71), abyssinone IV (4), abyssinone V (5), phaseollidin (237), erythrabyssin II (236), sigmoidin B (7), sigmoidin C (8), sigmoidin D (9), sigmoidin F (11), 5′-prenylpratensein (200), sandwicensin (245), neobavaisoflavone (189), semilicoisoflavone B (212) and licoagrochalcone A (104) were tested against some microbes using the bioautographic agar overlay method, and all these compounds strongly inhibited the growth of B. subtilis and S. aureus with MIC values in the range of 0.5 to 4 μg mL−1 relative to chloramphenicol (MIC = 0.001 μg mL−1). Only sigmoidin B (7) and Abyssinone VII (71) strongly affected the growth of E. coli, with a MIC value of 5.0 μg mL−1 (14.0 and 14.7 μM) (MIC = 0.05 μg mL−1 or 0.05 μM for chloramphenicol). Sigmoidin C (8), abyssinone VII (71), erythrabyssin II (235), phaseollidin (237), sandwicensin (245), neobavaisoflavone (189), 5′-prenylpratensein (200) and semilicoisoflavone B (212) exhibited strong activities against Saccharomyces cerevisiae with MIC values of 0.5, 5.0, 2.0, 0.5, 1.0, 0.5, 0.5, and 3.0 μg mL−1, respectively (MIC = 0.005 μg mL−1 for miconazole).86 It has been also reported that sigmoidin L (14) inhibited the growth of S. aureus and P. vulgaris with respective MICs of 4.0 and 7.0 μg mL−1 (11.3 and 17.8 μM), however, no positive control was reported in this study.116 Additionally, its congener sigmoidin M (16) exhibited significant antibacterial activity against S. aureus with a MIC of 4.0 μg mL−1 or 10.3 μM (no positive control).66 Erybraedins A–C (238–240), erythrabyssin II (235) and isoneorautenol (241) were assayed for their effect on the growth of certain bacteria, and erythrabyssin II (235) potentially inhibited the growth of S aureus ATCC 13709 and Mycobacterium smegmatis ATCC 607, with MIC values of 3.12 and 0.78 μg mL−1 (8.8 and 2.2 μM), respectively, in comparison to tests against streptomycin with MICs of 5.0 and 1.25 μg mL−1 (8.6 and 2.1 μM), respectively. Other compounds showed MICs in the range 6.25 and 25 μg mL−1 against these two bacteria and none of these compounds affected the growth of, E. coli ATCC 9637, Salmonella gallinarum ATCC 9184, Klebsiella pneumoniae ATCC 10031, Candida albicans ATCC 10231 or Pseudomonas aeruginosa ATCC 27853.56 Erybraedin A (238), erythrabyssin II (235), erystagallin A (266) and erycristagallin (269) exhibited strong antibacterial activities against several species and strains of Streptococcus and Staphylococcus as well as several strains of MRSA (Multi Resistant Staphylococcus aureus), with MICs ranging from 0.78 to 6.25 μg mL−1, relative to vancomycin and oxacillin (0.09 < MIC < 256 μg mL−1). These are promising results with regard to the fight against antibacterial resistance, and these compounds represent some potential antibiotics isolated from medicinal plants.167 Brucein B (130) displayed moderate antibacterial activities, with MIC of 62 μg mL−1 or 167.6 μM against Bacillus cereus (ATCC 33019) compared to chloramphenicol (MIC = 15 μg mL−1, 46.4 μM).94 The five compounds isolated from E. livingstoniana were evaluated for their antibacterial activity. Sigmoidin B-4′-methyl ether (68) displayed a good activity with MIC of 5.0 μg mL−1 against E. coli (DSM 1116), B. subtilis (DSM 1088), and E. coli (DSM 682). 7,3 ′-dihydroxy-4′-methoxy-5′-prenylflavanone (78) showed a MIC value of 2.0 μg mL−1 or 5.4 μM against B. subtilis (DSM 1088) in comparison to Streptomycin and gentamycin with MIC = 5.0 μg mL−1 or 8.6 μM and 1.0 μg mL−1 or 2.1 μM and a MIC of 5 μg mL−1 or 13.5 μM against the two strains of E. coli (streptomycin and gentamycin MIC = 1.0 μg mL−1 or 1.7 and 2.1 μM).93 Erythrabyssins I (234) and II (235), phaseollin (236), phaseollidin (237), abyssinone I (1), abyssinone II (2), abyssinone III (3), abyssinone IV (4), abyssinone V (5) and abyssinone VI (100) were assessed for their antimicrobial activities. Erythrabyssin I (234) and phaseollin (236) showed MICs of 12.5 and 6.25 μg mL−1, respectively, against S. aureus and B. subtilis. Erythrabyssin II (235) exhibited strong antibacterial activity against S. aureus, B. subtilis and Micrococcus lysodeikticus with a MIC value of 3.13 μg mL−1, whereas abyssinone V (5) showed a MIC of 12.5 μg mL−1 against Micrococcus lysodeikticus (positive control was not reported). Regarding antifungal activity, erythrabyssin I (234) showed a MIC of 6.25 μg mL−1 or 18.5 μM against Sclerotinia libertiana and phaseollin (236) displayed an activity of 12.5 μg mL−1 or 31.9 μM against Sclerotinia libertiana, Mucor mucedo and Rhizopus chinensis. In addition, abyssinones I (1) and II (2) exhibited a MIC of 12.5 μg mL−1 (32.1 and 38.6 μM) against Sclerotinia libertiana.113 Sigmoidin I (118), corylin (188), neobavaisoflavone (189) and phaseollidin (237) were assessed for their capacity to affect the growth of Candida albicans, Cryptococcus neoformans, Aspergillus fumigatus and S. aureus. Only neobavaisoflavone (189) showed a MIC of 50 μg mL−1 or 156.3 μM against C. neoformans and A. fumigatus,129 and neobavaisoflavone (189) displayed a MIC of 3.2 μg mL−1 or 10.0 μM against S. aureus.132 Sigmoidin A (6) inhibited the growth of S. aureus, M. luteus (WS) and M. luteus (IPC) with MICs of 12.5, 25 and 50 μg mL−1 (29.5, 59.0 and 117.9 μM); with inhibition diameter varying from 10 to 13 mm; sigmoidin B (7) only affected the growth of S. aureus with the same MICs.114 Eriotrichin B (110), erybraedins A (238) and C (240) exhibited good antibacterial activities against S. aureus with MICs of 8.3, 13.6, and 12.8 μg mL−1 (20.3, 42.0 and 32.8 μM), respectively, relative to a MIC = 6 μg mL−1 or 18 μM for penicillin.144 Abyssinone IV-4′-methylether (31) exhibited an activity of 25 μg mL−1 or 63.8 μM against S. aureus, P. stuartii ATCC 29916 and E. aerogenes ATCC1 3048 relative to 8.0, 128 and 32 μg mL−1, respectively, for ciproflaxacin.125 Neobavaisoflavone (189) exhibited good to moderate antibacterial activity against E coli ATCC8739 (MIC = 8.0 μg mL−1 or 25.0 μM), E. coli AG100 ΔacrAB mutant AG100, with an over-expressing acrF gene (MIC = 32.0 μg mL−1 or 100 μM), Enterobacter cloacae Clinical MDR isolates (MIC = 8.0 μg mL−1 or 25.0 μM), Klebsiella pneumonia Clinical MDR isolate (MIC = 8 μg mL−1 or 25.0 μM), Providencia stuartii Clinical MDR isolate (MIC = 8 μg mL−1 or 25.0 μM) and Pseudemonas aeruginosa (MIC = 8.0 μg mL−1 or 25.0 μM).168 4′,5,7-Trihydroxy-6-(2′′-hydroxy-3′′-prenyl)isoflavone (207) exhibited a good antimicrobial activity against E. coli, S. aureus and Candida mycoderma with respective MICs of 10.0, 5.0, and 10.0 μg mL−1 (29.8, 14.9 and 29.8 μM). 6,8-diprenylgenistein (149) inhibited the growth of S. aureus and C. mycoderma at respective MICs of 1.0 and 5.0 μg mL−1 (2.4 and 12.3 μM), erysenegalensein E (151) and isoerysenegalensein E (161) inhibited the growth of E. coli, B. subtilis and C. mycoderma at MIC values of 10.0, 5.0, and 10.0 μg mL−1, respectively. Sandwicensin (245) showed a MIC of 10.0 μg mL−1 or 31.0 μM against E. coli comparatively to chloramphenicol and miconazole had respective MICs of 0.01 and 1.0 μg mL−1 against these microorganisms.72 Erysubin E (265) showed an IC50 of 1.30 μM against C. perfringens, cristacarpin (234), and erystagallin A (266) exhibited a good antibacterial activity with IC50 values of 2.28 and 2.04 μM against C. perfringens. In addition, eryvarin D (255) and erythribyssin O (275) exhibited antibacterial activity with IC50s of 2.09 and 1.32 μM, as well as 3.30 and 0.35 μM, respectively, against C. perfringens and V. cholera comparative to quercetin (IC50 = 25.34 μM).146 The poor activity of phaseollin (236) could result from the cyclisation of the prenyl group relative to 255 and 275.

6.2 Antioxidant activity

Mildbone (33) and mildbenone (102) demonstrated strong scavenging activity with respective IC50 values of 20.2 and 28.5 μM compared to the positive control butylated hydroxyanisole (IC50 = 44.2 μM).54 Sigmoidin A (6) and B (7) showed DPPH scavenging activity at a dose of 100 μM. This activity was found to be better than that of the similar flavonoid (quercetin 3-O-β-D-glucopyranoside) with the same hydroxyl pattern, illustrating that the prenyl group might contribute to their scavenging effect.64 In addition, these two compounds were assessed for their effect in reducing superoxide anion radicals produced by rat alveolar macrophages in vivo, and they inhibited the superoxide anion at 90% and 65% respectively for sigmoidins A (6) and B (7) at a concentration of 100 μM each. The high effect of sigmoidin A (6) might be associated with the contribution of its second prenyl group at 6'.169 The isolated compounds from E. brucei, namely, bruceins A (129) and B (130), kenusanone F (131), sophoraisoflavanone A (133), cristacarpin (234), eryvarin J (267) and erycristagallin (269) were equally evaluated for their scavenging capacity, and they showed activities with IC50 values of 6.3, 13.3, 22.2, 21.2, 6.4, 1.4, and 1.1 μM relative to trolox (IC50 = 0.62 μM).94 Similarly, erylivingstone B (75), erylivingstone C (76), 5,7-dihydroxy-3′,4′-dimethoxy-5′-(3-methylbut-2-enyl) flavanone (77), sigmoidin B-4′-methyl ether (68) and 7,3′-dihydroxy-4′-methoxy-5′-(3-methylbut-2-enyl) flavanone (78) displayed respective IC50 values of 11.9, 11.4, 21.1, 12.9 and 10.7 μM (Trolox IC50 = 4.4 μM).93 The lower activity of 68 is likely related to its poor number of free hydroxyl groups.

6.3 Cytotoxic activity

The cytotoxic activity of a component is declared significant when the IC50 value is less than 10 μM or between 4–5 μg mL−1.170 Several reports claim that prenylated flavonoids have better anticancer effects than corresponding flavonoids and glycosides. It can be suggested that the hydrophilicity of these compounds could play an important role in the invasion of cancer cell lines.171 Several mechanisms of action are associated with the cell invasion, such as the initiation of apoptosis, the induction of autophagy, the inhibition of tumour angiogenesis and inhibition of cellular migration.172

Lysisteisoflavanone (113), erycaffra C (160), alpinumisoflavone (148), derrone (155), warangalone (141), isoerysenegalensein E (161), erysenegalensein E (151), laburnetin (165), senegalensein (23), isosenegalensein (162) and burttinone (28) exhibited a cytotoxic effect on human cervix carcinoma KB-3-1 cells, with respective IC50 values of 183, 104, 71.5, 230, 73.4, 99, 58.4, 250, 37.8, 53.8 and 58.8 μM, however, unfortunately no positive control was used during their assay.130 Erycaffra B (112) was reported to affect KB cells with an ED50 value of 12.3 μM.63 Lipoxygenase are expressed in tumour cells, epithelial and immune cells and play an important function in inflammation, skin disorders and tumorigenesis.173 Mildbone (33) and mildbenone (102) showed a moderate inhibitory effect on the enzyme with IC50 values of 41.8 and 59.7 μM relative to 22.6 μM of baicalein.54 Erymildbraedins A (166) and B (167), scandenone (141), erysenegalensein M (159), 5,4′-dihydroxy-2′-methoxy-8-(3,3-dimethylallyl)-2′′,2′′dimethylpyrano[5,6:6,7]isoflvone (168) and eryvarin B (173) were evaluated for their effect on the growth of MCF-7 breast cancer cells, LNCaP prostate cancer cells and Ishikawa endometrial cancer cells using MTT and/or SRB. Scandenone (141), 5,4′-dihydroxy-2′-methoxy-8-(3,3-dimethylallyl)-2′′,2′′dimethylpyrano[5,6:6,7]isoflvone (168) and eryvarin B (169) strongly inhibited the growth of MCF-7 breast cancer cells with EC50 values of 7.0, 6.8, and 7.1 μM, respectively. In addition, they inhibited the growth of Ishikawa endometrial cancer cells with EC50 values of 7.4, 7.4, and 7.7 μM, respectively. Compound 168 and 169 strongly affected the growth of LNCaP prostate cancer cells with EC50 values of 4.1 and 4.6 μM, respectively, while the activity of 141 was moderate (EC50 = 6.9 μM). In all cases, the EC50 values of the reference Faslodex® were between 7 and 30 μM.53 It was evident that the prenyl group at position 8 of these structures played a crucial role in their cytotoxic effect against these cells compared to other congeners where this prenyl was either oxygenated or the double bond was not at the same position. Phaseollidin (237) and cristacarpin (234) were assessed for their cytotoxicity towards several cancer cell lines and compound 237 exhibited an activity of 12.3 μM (no positive control).85 Erythribyssin A (260), erybraedin B (239), folitenol (264), erybreadin D (242), and erybreadin C (240) all showed cytotoxic effects on certain cancer cell lines (with the exception of erybreadin D (242) which had no effect on MCF7 and MDA-MB-231 or folitenol (264)); these compounds exhibited good to moderate cytotoxicity against MCF7 and MDA-MB-231 (human breast carcinoma cells), and the multidrug-resistant cell lines MCF7/TAMR and MCF7/ADR with IC50 values ranging from 5.6 to 28.0 μM, comparative to the positive control (Tamoxifen) which showed IC50 values in the range 10.9–12.4 μM.145 As an ongoing part of the same investigation, the authors evaluated the activity of erythraddisons I and II (208, 209), euchrenone b10 (174) and erysubin F (204) and erythraddisons III and IV (124, 125) and results showed that erythraddison II (209), erythraddisons III and IV (124, 125) and echrenone b10 (174) exhibited good cytotoxicity against MCF7 and MDA-MB-231 human breast carcinoma cells and the Adriamycin resistant cell line MCF7/ADR, with the IC50 values ranging from 4.32–11.41 μM (Tamoxifen, IC50 = 11.44, 11.13, and 12.41 μM, respectively).78 Neorautenol (244), phaseollin (236), calopocarpin (249), isoneorautenol (241), orientanol C (253) and cristacarpin (235) were studied for their effects on H4IIE rat hepatoma cells and phaseollin and neorautenol showed prominent toxicity on H4IIE cells, inducing apoptotic cell death at a dose of 2 μM.79 Sigmoidin A (6) was assessed for its cytotoxic effect on B16 melanoma and RAW 264.7 cell lines. It was found that this compound exhibited a dose-dependent cytotoxicity towards the two cells, with the higher activity observed at a concentration of 100 μM, which reduced the cell concentration to zero, compared to its congener eriodictyol with no prenyl fragments which inhibited less than 40% cytotoxicity on both cells. It appears from these results that the prenyl groups increase the cytotoxicity effect of flavonoids.174 Prenylated flavonoids isolated by Zarev et al. (2017) were evaluated for their antigenotoxic activities against aflatoxin B1 induced genotoxicity, and in the Vitotox assay, sigmoidin A (6), and B (7) showed good antigenotoxic activity with MIC values of 53.9 and 52.5 μM compared to curcumin (IC50 = 50 μM), however 4′-O-methylsigmoidin B (68), and abyssinins I (25), II (26), III (27) showed moderate activities with respective IC50 values of 68.1, 59.2, 68.1, and 61.4 μM.84 Sigmoidin I (118), sophorapterocarpan A (258), and 6α-hydroxyphaseollidin (259) induced apoptosis in Leukemia cells (CCRF-CEM) with IC50 values of 4.24, 3.73, and 3.36 μM, respectively in comparison to doxorubicin (IC50 = 0.20 μM). In addition, 6α-hydroxyphaseollidin (259) revealed good activity towards MDA-MB-231- pcDNA (breast cancer cells), HCT116 (p53+/+) (colon cancer cells), U87MG (glioblastoma cells) and HepG2 (Hepatocarinoma cells) with respective IC50 values of 5.70, 5.68, 4.71, and 6.44 relative to doxorubicin (IC50 = 1.1, 1.41, 1.06, 3.83 μM).99 Neobavaisoflavone (189), sigmoidin H (136), and isoneorautenol (241) were tested for their ability to affect the growth of certain cancer cell lines and isoneorautenol (241) exhibited a prominent cytotoxicity towards MDA-MB-231-BCRP (cDNA for the breast cancer resistance protein, BCRP) and knockout clones HCT116 (p53−/−) (colon cancer cells), with respective IC50 values of 2.67 and 9.89 μM compared to the positive control doxorubicin (IC50 = 7.83 and 4.06 μM).108 Alpinumisoflavone (148) and abyssinone V-4′-methyl-ether (18) showed a good binding affinity to ERα with an IC50 value of 4.5 μM, as well as to ERβ with IC50 = 15 μM for both compounds.175 Burttinone (28) exhibited a good cytotoxicity towards the colon cancer cell line HCC-2998 with an IC50 of 20 μM, however, no positive control was reported for the assay.71 Indicanines D (195), wighteone (197), alpinumisoflavone (148), erysenegalensein E (151) and 8-prenylerythrinin C (146) were assessed for their effects on human KB cells. They showed respective ED50 values of 12.5, 0.78, 4.13, 6.25, and 13.0 μg mL−1 (no positive control was reported).69 Excelsanone (215) and 6,8-diprenylgenistein (149) inhibited the DU145 prostate carcinoma cells at doses of 1, 10 and 20 μg mL−1, but only excelsanone (215) showed similar activity against PC3 prostate carcinoma cells.9 Addisoniaflavanones I (89) and II (90) reduced the viability of H4IIE hepatoma cells with respective EC50 values of 5.25 and 8.5 μM.81 The flavanones abyssinone V (5), 4′-methylabyssinone V (18), abyssinone IV (4), and abyssinoflavanone VII (43) showed good cytotoxicity with respective IC50 values of 15.0, 5.0, 15.0, and 3.5 μM. These values were good in comparison to those of their respective flavanone skeleton without prenyl groups (IC50 > 100 μM), illustrating the enhancement of the activity by the prenyl groups.80 Abyssinones A, C and D (106, 108 and 109) exhibited a cytotoxic activity against human colorectal cancer cell line Caco2, with IC50 values of 13.3, 15.1, and 11.1 μM, respectively. Abyssinone B (107) poorly affected these cells (IC50 > 30 μM).128 Erybraedin A (238), erythrabyssin II (235), phaseollin (236), eryzerin C (229), eriotrichin B (110), (6aR,11aR) 3-hydroxy-4(γ,γ-dimethylallyl)-2′,2′-(3′′-hydroxy)-dimethylpyrano[6′′,5′′:9,10]pterocarpan (287) and eryvarin D (255) showed cytotoxicity effects on human retinal endothelial cells (HRECs), with respective IC50 values of 4.21, 2.57, 3.65, 4.65, 5.85, 4.67 and 5.91 μM, compared to the positive control SH-11037 with IC50 = 0.018 μM.142

According to studies on molecular docking, the prenylflavonoids induced apoptosis by increasing the p53 protein. They are also believed to decrease the anti-apoptotic protein Bcl-2 and activate the caspase family in A549 cells. The prenyl groups attached to flavonoids interact with leucine, alanine, valine and lysine, which might be associated with the aforementioned apoptosis.176 Other studies have supported the up-regulation of the tumour necrosis factor-related apoptosis-inducing ligand and a down-regulation of the death receptor 5, thus contributing to the production of apoptotic amplificators.177

6.4 Anti-inflammatory activity

Prenylated flavonoids have been shown to have higher bioavailability than non-prenylated ones. The high pain-relieving effects of these flavonoids have therefore been associated with their bioavailability. Specific studies have been conducted on 8-prenylnaringenin and 8-prenylquercetin and these compounds were found to be accumulated in the liver, muscles and kidney.178,179 Other studies showed that icariin had a long term suppressive effect on paclitaxel-induced neuroinflammation and mechanical allodynia.180

Sigmodins A (6) and B (7) were tested against TPA-induced oedema and were all effective at a dose of 0.25 mg per ear by decreasing oedema by 89 and 83%, respectively, relative to the positive control indomethacin, which had a percentage of 83% at 0.5 mg per ear.64 The effect of erycristagallin (269) on ear inflammation induced by multiple topical applications of TPA was assessed and it inhibited swelling at 34% and the production of neutrophil infiltration at 59%, at a dose of 0.1 mg per ear.64 Abyssinone V-4′-methyl ether (18) was evaluated for its effects towards xylene induced-ear edema in mice and cotton pellet-induced granuloma model in rats; the best activity was obtained with a dose of 10 mg kg−1 of abyssinone V-4′-methyl ether (18), which inhibited the oedema at 71.43% compared to 2.5 mg kg−1 of dexamethasone (61.9% of inhibition) in xylene induced-ear edema in mice. This compound equally inhibited cotton pellet-induced granuloma model in rats at a dose of 10 mg kg−1 (61.32%) compared to dexamethasone at a dose of 2.5 mg kg−1 (68.72%).181 These pain-relieving activities are likely related to a high accumulation of the tested prenylated flavonoids in the muscles of the mice. The mechanism of action of these compounds might be related to the suppression of certain pro-inflammatory markers, including tumour necrosis factor α (TNF-α), interleukin 1β (IL-1β) and interleukin 6 (IL-6), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) phosphorylation (p65) in the spinal cord of mice.180 Other findings support that this mechanism of action of prenylated flavonoids might be via the inhibition of cyclooxygenase-2 (COX-2), as demonstrated by the prenylated flavonoid cudaflavone B.7

6.5 Antiplasmodial activity

Abyssinone V (5), 5′-prenylpratensein A (93) and shinterocarpin (263) exhibited antiplasmodial activity with respective IC50 values of 4.9, 6.3, and 6.6 μM against the chloroquine-sensitive strain of Plasmodium falciparum (chloroquine IC50 = 0.008 μM). They equally displayed IC50 values of 6.1, 8.7, and 8.3 μM against a chloroquine-resistant strain (chloroquine IC50 = 0.075 μM).75 In addition, abyssinone IV (4) and erythrabyssin II (235) showed good antiplasmodial activities, with IC50 values of 7.7 and 6.5 μM against a chloroquine resistant strain (chloroquine IC50 = 0.093 μM) as well as 9.0 and 8.1 μM against a chloroquine sensitive strain (chloroquine IC50 = 0.008 μM).88 Abyssinin III (27), abyssinones IV (4) and V (5), sigmoidins A (6) and B (7) exhibited good antiplasmodial activity with IC50 values of 5.8, 5.4, 4.9, 5.8 and 8.1 μM against a chloroquine sensitive strain (chloroquine IC50 = 0.009 μM), as well as 5.2, 5.9, 6.1, 5.9 and 9.3 μM towards a chloroquine-resistant strain (chloroquine IC50 = 0.08 μM).127 Burttinol A (231) and C (233) and abyssinone V (5) displayed good activity with IC50 values 7.6, 9.3, and 5.7 μM against a chloroquine-sensitive strain (chloroquine IC50 = 0.009 μM) and 8.5, 9.1, and 6.6 μM against a chloroquine resistant strain (chloroquine IC50 = 0.08 μM), and Abyssinone V 4′-methyl ether (18) showed moderate activity (IC50 = 10.7 and 11.9 μM) against these respective strains, and in addition, the methyl at 4′ decreased its activity relative to abyssinone V (5).182

6.6 Antidiabetic activity

Overcoming insulin resistance is considered to be one of the major challenges for conventional anti-diabetic medication. The Protein Tyrosine Phosphatase 1B (PTP-1B) is recognized as a key element in the regulation of insulin signal transduction pathways, and is unfortunately considered to be a negative regulator of the insulin receptor pathway along with the leptin receptor pathway.183 Therefore, inhibiting this enzyme will contribute to overcoming insulin resistance.

The chemical constituents of E. addisoniae 5,2′,4′-trihydroxy-6-(γ,γ-dimethylallyl)-2′′′,2′′′-dimethyldihydropyrano[5′′′,6′′′] isoflavone (121), orientanol E (122), senegalensein (23), warangalone (141), warangalone 4′-methyl ether (205) and 2,3-dihydroauriculatin (117) were assayed for their inhibitory effect on the protein PTP1B, and orientanol E (122), 2,3-dihydroauriculatin (117) and 5,2′,4′-trihydroxy-6-(γ,γ-dimethylallyl)-2′′′,2′′′-dimethyldihydropyrano[5′′′,6′′′]isoflavone (121) exhibited the activities with IC50 values of 10.1, 2.6, and 4.1 μM, relative to the reference ursolic acid (IC50 = 2.5 μM). In contrast, the weak activity of other compounds might be related to the double 2,3.77 Compounds 45, 47, 49, 50, 53 and 54 exhibited good inhibitory effects on PTP1B with IC50 values of 13.9, 14.9, 18.2, 19.0, and 18.2 μM, respectively, compared to ursolic acid (3.6 μM).90 Additionally, compounds 51–2 and 40 were also assayed for their impact on the protein PTP1B. With the exception of 53 and 55 these metabolites showed dose-dependent activities, with IC50 values ranging from 15.2 to 19.6 μM compared to RK-682 (IC50 = 4.7 μM).91 Erylysin B (251), eryvarin D (255) and erybraedin A (238) also exhibited activity, with IC50 values of 6.0, 4.1, and 1.01 μM, respectively (ursolic acid IC50 = 2.5 μM).70 Neorautenol (244), erybreadin B (239), folitenol (264), erybreadin D (242), erysubin E (265) and erybreadin C (240) exhibited good activities against PTP1B protein with IC50 values of 7.6, 4.2, 7.8, 6.4, 8.8, and 7.3 μM, respectively (ursolic acid IC50 = 3.6 μM). Further investigation by Nguyen et al. in 2011 aimed to evaluate the activities of erythribyssin E (126), 5-deoxyabyssinin II (44), abyssinone III (3) 7-hydroxy-2-[4-methoxy-3-(3-methylbut-2-enyl)phenyl]chroman-4-one (72), abyssinone V (5), abyssinone II (2), prostratol C (128), erythribyssin G (73), erythribyssin I (74) and erythribyssin J (127) against PTP1B. Apart from erythribyssin I (74), the compounds exhibited moderate dose-dependent activities, with the IC50 range of 14.9–98.1 μM (IC50 = 3.6 μM for ursolic acid).123 Additionally, erythraddison II (211), euchrenone b10 (174) and erysubin F (203), as well as erythraddison III and IV (124, 125) showed good inhibitory effect on PTP3B, with the IC50 values ranging from 4.6–17.4 μM (ursolic acid: IC50 = 3.6 μM).78 Compound (3R)-2,7-dihydroxy-3-(3-methylbut-2-enyl)-2,2-dimethylpyrano[5,6:4,5]isoflavan (224) exhibited an IC50 value of 5.5 μM (ursolic acid: IC50 = 3.6 μM).52 Some compounds isolated from E. mildbraedii were evaluated for their inhibitory activity on the protein tyrosine phosphatase-1B (PTP1B) and abyssinone IV (4), and abyssinone VI-4′-O-methyl ether (101) potentially inhibited the activity of this protein with the respective IC50 values of 16 and 14.8 μM. Other compounds, abyssinone V-4′-O-methyl ether (18), abyssinone IV-4′-O-methyl ether (31), abyssinone V (5), sigmoidin E (10) and alpinumisoflavone (148) showed IC50 values of 26.3, 21.2, 39.7, 39.2, and 41.5 μM, respectively, with ursolic acid used as a positive control (IC50 = 3.6 μM). Regarding the activities of 4 and 101 compared to 18, 5, 10, and 148, the carbonyl in 4 and 101 could have improved the activity by chelating the hydrogen atoms in the protein.51 It is worthy to note that all the chemical structures of the compounds which exhibited antidiabetic activity beared the unmodified prenyl moieties.

6.7 Antiviral activity

8-Prenylluteone (139), auriculatin (152), erysenegalensein O (153), erysenegalensein D (150), erysenegalensein N (154), derrone (155), alpinumisoflavone (148) and 6,8-diprenylgenistein (149) were evaluated for their ability to inhibit the protease responsible for maturation of HIV-1 copies and they exhibited good to moderate activities with respective IC50 values of 4.0, 3.5, 5.0, 2.5, 4.5, 18.2, 30.1, and 0.5 μM compared to acetylpepstatin (IC50 = 0.09 μM). It was evidenced that the 6,8-diprenyl groups might improve the potency of HIV-1 PR inhibition in 4′-hydroxy isoflavonoids.101

6.8 Other activities

Erycaffrain A (150) showed moderate estrogenic activity with a MAC value of 1.25 μg mL−1 following the β-glucuronidase plant assay system.163 Clonic seizures were induced in mice by the ip injection of pentylenetetrazol (70 mg kg−1), picrotoxine (7.5 mg kg−1) and pilocarpine (375 mg kg−1); these animals then received injections of abyssinone V-4′-methyl ether (18). The best activities with this compound were recorded at 100 mg kg−1 in PTZ-induced (clonazepam, 0.1 mg Kg−1) and 25 mg kg−1 in PIC-induced (0.4 mg kg−1 clonazepam) and PILO-induced (Diazepam, 0.3 mg kg−1), thus illustrating the anticonvulsant effect of abyssinone V-4′methyl ether (18) in vivo.96 Erybraedin D (242) was evaluated for its ability to inhibit the enzyme 15-lipoxygenase, and displayed activity with an IC50 of less than 32 μM, while the positive control quercetin had an IC50 of 30 μM.107 2,3-Dihydro-2′-hydroxyosajin (121), osajin (221) and 6,8-diprenylgenistein (143) showed hepatoprotective activities in CCl4 induced hepatotoxicity at respective percentages of 71.8, 67.54 and 69.41%, compared to 63.8% of silymarin.106

From studies on the biological activities of these prenylated flavonoids, it appears that a large number have exhibited cytotoxic effects towards cancer cell lines. The second most prominent activity was their antidiabetic and antimicrobial potential. However, few of these compounds were evaluated for their antiviral and anti-inflammatory activities. In addition, they were largely investigated for their antioxidant effect but had poor scavenging effects (Fig. 14). According to some specific subclasses, mainly isoflavones, pterocarpanes and flavanones exhibited cytotoxic activity. More than 20 flavanones derivatives contributed to antidiabetic effects of prenylated flavonoids occurring in the genus Erythrina. About 8 pterocarpans also showed antidiabetic potential. Overall, the prenylated flavonoids exhibited promising activities on certain bacteria, mainly S. aureus and B. subtilis (two Gram-positive bacteria), and 6,8-diprenylgenistein (149) exhibited a lower lethal dose (1 mg mL−1 or 2.46 mM) while showing good activity against B. subtilis, E. coli (Gram-negative bacteria) and Candida mycoderma.72 Unfortunately, not all these reported studies evaluated the toxicities of these antimicrobial compounds against normal cells. This is one of the challenges in terms of the search for lead or hit molecules, as many prenylated flavonoids were active. However, it is imperative that these studies be revised using state-of-the-art methodologies and analyses to re-evaluate these activities and carry out the cytotoxicity assays. Another challenge worth mentioning is the non-use of positive controls in certain assays, which renders the results less accurate. Certain methods used to assess biological activities in previous studies are obsolete and no longer in use. Hence, the results reported might not reflect the up-to-date challenges in drug discovery research.184 For example, the agar dilution method assay was not automated until recently. The well diffusion assay is well limited, and the 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radical assay, ferric reducing antioxidant power (FRAP) assay and other methods of assessment of biological activity are no longer considered to be efficient.184 Various assays were only based on one method, whereas it is advised consider more than one in vitro assay to characterise the biological activity of a molecule184


image file: d5ra03457d-f14.tif
Fig. 14 Summary of biological activities of prenylated flavonoids.

This review therefore encourages more biological assays on prenylated flavonoids from Erythrina plants and other plant species. We also recommend multiple in vivo studies on the anticancer, antimicrobial, and antidiabetic activities of any prenylated flavonoids.

7 Some targeted proteins in drug analysis

7.1 Human epidermal growth factor receptor 2: 1N8Z

The HER2 is a protein belonging to the tyrosine kinase family that comprises more than one thousand amino acids. Its ligand-binding site contains 632 amino acids185 and does not bind any ligand as the other variants do (HER1, HER3, and HER4).186 This protein accounts for about 20% of breast cancers and 10 to 30% of gastric/gastroesophageal cancers.187,188 Jeong et al., studying the breast cancer cell line SKBR3 reported that HER2 is co-localized with actin (phalloidin) in punctate regions of the plasma membrane that protrude from the apical aspect of these cells.189 The HER2 causes uncontrolled growth and division in breast cells, leading to breast cancer.187,190 Breast cancer can possess up to 25 to 50 copies of HER2 and up to 40 to 100-fold increase in HER2 protein, resulting in two million receptors expressed at the tumour cell surface.191 The activities of HER2 can be inhibited using tyrosine kinase inhibitors or monoclonal antibodies, thus suppressing the tumour cell growth.192 We should mention here that the variant HER1, generally referred to as EGFR1 or simply EGFR, is reported to be implicated in both lung and breast cancers,193–195 specifically, EGFR undergoes certain mutations to generate the subvariant EGFR TKI resistant;194 this protein has more affinity with ATP and its mechanism is thought to resist gefitinib or erlotinib196 and is encoded in PDB as 1M17.197

6,8-Diprenylgenistein (149) and phaseollin (236) were docked in silico method for their ability to bind to the active site human oestrogen receptor- (hER-), B-cell lymphoma 2 (Bcl-2), cyclin-dependent kinase (CDK-2), ikappaB kinase (IkB) and growth factor receptor epidermal layer (EGFR); 6,8-diprenylgenistein (149) and phaseollin (236) showed binding energies with respective ΔG values of −10.66, and −9.22 kcal mol−1 (Fig. 15) with ERα receptor, compared to the positive controls 17β-estradiol (−10.40 kcal mol−1) and tamoxifen (−11.35 kcal mol−1); they equally showed respective binding affinities with CDK-2 receptor of −10.14, and −8.03 kcal mol−1 relative to roniciclib and 106 (oxindole) (ΔG = −7.86 and −9.24 kcal mol−1). These compounds also showed binding affinities with respective ΔG values of −9.51 and −9.06 kcal mol−1 with EGFR receptor in comparison to −11.22 kcal mol−1 for the positive control (canertinib).198


image file: d5ra03457d-f15.tif
Fig. 15 Binding energies of compounds 149, 236, 242 and 248.

7.2 Angiotensin-converting enzyme 2: ACE2

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the pandemic COVID-19. The invasion of the host cells by the virus is facilitated by the Angiotensin-converting enzyme 2 (ACE2) receptor located at the surface of host cells.199 These receptors are expressed in various human organs.200,201 A recent study conducted by Herlina and coworkers revealed that erybraedin D (242) and gangetinin (248), out of 18 flavonoids from the genus Erythrina, had a better affinity with the ACE2 receptor.202 These two compounds were docked against the ACE2 receptor and had respective binding affinities of −7.028 and −6.379 kcal mol−1202 (Fig. 15). Therefore, these two flavonoids could play an important function in the pathogenicity of SARS-CoV-2.

8 Conclusion

In conclusion, twenty species of Erythrina were collected and investigated throughout Africa, and these studies resulted in the isolation and characterization of 289 prenylated flavonoids. A high percentage of these compounds proved to be prenylated flavanones. This is not surprising given that other subclasses of flavonoids derive from naringenin (flavanone subclass). According to the distribution, abyssinone V (5) and abyssinone V-4′-O-methyl ether (18), 6,8-diprenylgenistein (149) and phaseollidin (237) can be considered to be the chemical markers of the genus Erythrina as far as prenylated flavonoids are concerned. Pterocarpans were mainly characterized from E. abyssinica, E. eriotricha, E. burtii and E. senegalensis. No pterocarpan was isolated from E. caffra or E. sigmoidea. The flavanone subclass was largely characterized from E. caffra, E. sigmoidea, E. latissimi, E. addisoniae and E. burtii. Finally, the subclass isoflavone was mostly isolated from E. caffra, E. abyssinica, E. burtii and E. senegalensis. Most of these compounds have been evaluated for their biological effects, and were found to exhibit good antibacterial, anticancer and antidiabetic activities. About 68 prenylated flavonoids summarized in this work exhibited good cytotoxic effects against numerous cancer cell lines. 46 and 44 flavonoids exhibited promising antimicrobial and antidiabetic activities. Of these compounds, 20 prenylated flavanones displayed anti-diabetic properties. As some flavonoids are already marketed as dietary supplements for bacterial infections, antioxidant effect, immune system booster, prostatitis, and many more, the prenylated flavonoids herein reported have exhibited promising biological effects and therefore could be further investigated for their pharmacological activities.

The chemical shift ranges of different prenyl groups were highlighted to assist future structure elucidation or rapid identification of prenyl moieties. However, many prenyl groups possessing stereogenic centres were not fully characterized and further research will be needed to complete their structure elucidation.

Data availability

The data supporting this article have been included in the manuscript and as part of the ESI.

Author contributions

Conceptualization: B. Tsakem, X. Siwe-Noundou. Original draft preparation: B. Tsakem. Writing, review and editing: B. Tsakem, F. Ntie-Kang, R. B. Teponno, X. Siwe-Noundou. Supervision: F. Ntie-Kang, R. B. Teponno, X. Siwe-Noundou. Funding acquisition: F. Ntie-Kang, X. Siwe-Noundou. All authors have read and agreed to the published version of the manuscript.

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgements

The work reported herein was made possible through funding from the South African Medical Research Council through its Division of Research Capacity Development under the Research Capacity Development Initiative (RCDI) Programme to XSN. The content and findings reported/illustrated are the sole deduction, view and responsibility of the researchers and do not reflect the official position and sentiments of the SAMRC. XSN thanks the South Africa National Research Foundation/Competitive Support for Unrated Researchers (NRF/CSUR) Grant Number: SRUG2203291031. We acknowledge financial support from the Bill & Melinda Gates Foundation through the Calestous Juma Science Leadership Fellowship awarded to Fidele Ntie-Kang (grant award number: INV-036848 to the University of Buea). FNK also acknowledges joint funding from the Bill & Melinda Gates Foundation and LifeArc (award number: INV-055897 and Grant ID: 10646) under the African Drug Discovery Accelerator program. FNK acknowledges further funding from the Alexander von Humboldt Foundation for a Research Group Linkage project.

References

  1. R. B. Teponno, S. Kusari and M. Spiteller, Nat. Prod. Rep., 2016, 33, 1044–1092 Search PubMed .
  2. D. J. Newman and G. M. Cragg, J. Nat. Prod., 2020, 83, 770–803 Search PubMed .
  3. B. Y. Kianfé, J. Tchamgoue, A. Narmani, R. B. Teponno, A.-L. Njouonkou, M. Stadler and S. Fogue Kouam, Molecules, 2023, 28, 3120 Search PubMed .
  4. T. R. Fouedjou, B. Tsakem, X. Siwe-Noundou, H. P. Dongmo Fogang, A. Tiombou Donkia, B. Kemvoufo Ponou, M. Poka, P. H. Demana, R. B. Teponno and L. Azefack Tapondjou, Biomolecules, 2023, 13, 1783 Search PubMed .
  5. B. Tsakem, G. Li and R. B. Teponno, Bioorg. Chem., 2024, 150, 107572 Search PubMed .
  6. X. Chen, E. Mukwaya, M.-S. Wong and Y. Zhang, Pharm. Biol., 2014, 52, 655–660 Search PubMed .
  7. S. Shi, J. Li, X. Zhao, Q. Liu and S.-J. Song, Phytochemistry, 2021, 191, 112895 Search PubMed .
  8. J. C. Chukwujekwu, F. R. Van Heerden and J. Van Staden, Phytother. Res., 2011, 25, 46–48 Search PubMed .
  9. A. J. Y. Gbaweng, H. Daïrou, S. Zingué, T. Emmanuel, A. T. Tchinda, M. Frédérich and J. T. Mbafor, Nat. Prod. Res., 2020, 34, 659–667 Search PubMed .
  10. R. R. T. Majinda, C. C. W. Wanjala and B. F. Juma, in Studies in Natural Products Chemistry, Elsevier, 2005, vol. 32, pp. 821–853 Search PubMed .
  11. N. M. Fahmy, E. Al-Sayed, M. El-Shazly and A. N. Singab, Ind. Crops Prod., 2018, 123, 500–538 Search PubMed .
  12. N. T. Son and A. I. Elshamy, Comb. Chem. High Throughput Screening, 2021, 24, 20–58 Search PubMed .
  13. H.-W. Lv, Q.-L. Wang, M. Luo, M.-D. Zhu, H.-M. Liang, W.-J. Li, H. Cai, Z.-B. Zhou, H. Wang, S.-Q. Tong and X.-N. Li, Arch. Pharmacal Res., 2023, 46, 207–272 Search PubMed .
  14. G. Diana, M. Catanzaro, A. Ferrara and P. Ferrari, Clin. Ter., 2000, 151, 341–344 Search PubMed .
  15. M. Gohel and A. Davies, Curr. Vasc. Pharmacol., 2009, 7, 303–308 Search PubMed .
  16. S. Mustafa, M. Akbar, M. A. Khan, K. Sunita, S. Parveen, J. S. Pawar, S. Massey, N. R. Agarwal and S. A. Husain, Curr. Res. Pharmacol. Drug Discov., 2022, 3, 100122 Search PubMed .
  17. B. Yang, H. Liu, J. Yang, V. K. Gupta and Y. Jiang, Trends Food Sci. Technol., 2018, 79, 116–124 Search PubMed .
  18. J. P. J. Marais, B. Deavours, R. A. Dixon and D. Ferreira, in The Science of Flavonoids, ed. E. Grotewold, Springer New York, New York, NY, 2006, pp. 1–46 Search PubMed .
  19. T. Tohge, L. P. De Souza and A. R. Fernie, J. Exp. Bot., 2017, 68, 4013–4028 Search PubMed .
  20. S. Liga, C. Paul and F. Péter, Plants, 2023, 12, 2732 Search PubMed .
  21. P. M. Dewick, Medicinal Natural Products: A Biosynthetic Approach, Wiley, 1st edn, 2009 Search PubMed .
  22. K. Yazaki, M. Kunihisa, T. Fujisaki and F. Sato, J. Biol. Chem., 2002, 277, 6240–6246 Search PubMed .
  23. Y. Asada, W. Li and T. Yoshikawa, Phytochemistry, 2000, 55, 323–326 Search PubMed .
  24. K. Sasaki, K. Mito, K. Ohara, H. Yamamoto and K. Yazaki, Plant Physiol., 2008, 146, 1075–1084 Search PubMed .
  25. J. Li, R. Chen, R. Wang, X. Liu, D. Xie, J. Zou and J. Dai, ChemBioChem, 2014, 15, 1673–1681 Search PubMed .
  26. H. Zhou, Y. Yuan, Y. Liu, W. Deng, J. Zong, Z.-Y. Bian, J. Dai and Q.-Z. Tang, Exp. Ther. Med., 2014, 7, 1116–1122 Search PubMed .
  27. S.-M. Li, Phytochemistry, 2009, 70, 1746–1757 Search PubMed .
  28. Y. Yang, L. Tao, Y. Li, Y. Wu, Q. Ran, D. Li, S.-M. Li, X. Yu, C.-M. Yuan and K. Zhou, J. Agric. Food Chem., 2024, 72, 8018–8026 Search PubMed .
  29. T. Akashi, K. Sasaki, T. Aoki, S. Ayabe and K. Yazaki, Plant Physiol., 2009, 149, 683–693 Search PubMed .
  30. K. Yoneyama, T. Akashi and T. Aoki, Plant Cell Physiol., 2016, 57, 2497–2509 Search PubMed .
  31. K. Yazaki, K. Sasaki and Y. Tsurumaru, Phytochemistry, 2009, 70, 1739–1745 Search PubMed .
  32. A. Sukumaran, T. McDowell, L. Chen, J. Renaud and S. Dhaubhadel, Plant J., 2018, 96, 966–981 Search PubMed .
  33. S. Santoro, J. B. Azeredo, V. Nascimento, L. Sancineto, A. L. Braga and C. Santi, RSC Adv., 2014, 4, 31521–31535 Search PubMed .
  34. M. A. Moreira Costa, N. F. De Sousa, C. L. Mansur Pontes, M. T. Scotti, F. F. De Assis, A. L. Braga and L. P. Sandjo, Fitoterapia, 2024, 173, 105784 Search PubMed .
  35. B. Tsakem, J. Tchamgoue, R. T. Kinge, G. L. M. Tiani, R. B. Teponno and S. F. Kouam, Fitoterapia, 2024, 106154 Search PubMed .
  36. J. R. N. Kuete, O. G. Fadeyi, B. A. Olou, F. W. T. Kenmoe, B. Tsakem, R. B. Teponno, L. A. Tapondjou, S. F. Kouam and N. S. Yorou, Phytochem. Lett., 2024, 65, 19–23 Search PubMed .
  37. G. K. Azad and R. S. Tomar, Mol. Biol. Rep., 2014, 41, 4865–4879 Search PubMed .
  38. S. Hou, J. Evidence-Based Complementary Altern. Med., 2022, 2022, 1–10 Search PubMed .
  39. N. Nasim, I. S. Sandeep and S. Mohanty, Nucleus, 2022, 65, 399–411 Search PubMed .
  40. J. J. Guzmán-Rodríguez, A. Ochoa-Zarzosa, R. López-Gómez and J. E. López-Meza, BioMed Res. Int., 2015, 2015, 1–11 Search PubMed .
  41. B. Tsakem, S. Mokhele, L. Mosima, M. Poka, P. H. Demana, R. B. Teponno and X. Siwe Noundou, Med. Chem. Res., 2025, 34(6), 1212–1236 Search PubMed .
  42. Y. J. Moon, X. Wang and M. E. Morris, Toxicol. in Vitro, 2006, 20, 187–210 Search PubMed .
  43. A. Kozłowska and D. Szostak-Węgierek, in Sweeteners, eds. J.-M. Merillon and K. G. Ramawat, Springer International Publishing, Cham, 2018, pp. 1–27 Search PubMed .
  44. C. M. M. Santos and A. M. S. Silva, Molecules, 2020, 25, 696 Search PubMed .
  45. L. Bourassa, https://thefitbay.com/best-flavonoid-supplements-reviewed/, 2024, accessed on 2024-01-12.
  46. V. S. Lemos, S. F. Côrtes, M. H. dos Santos, J. Ellena, M. E. C. Moreira and A. C. Doriguetto, Chem. Biodiversity, 2006, 3, 635–645 Search PubMed .
  47. Supersmart, https://www.supersmart.com/en/shop/anti-ageing/xanthohumol-supplement-0784, 2024, accessed on 2024-08-12.
  48. M. Sandasi, G. Kamatou, W. Chen and N. Mulaudzi, in The South African Herbal Pharmacopoeia, Elsevier, 2023, pp. 41–56 Search PubMed .
  49. Z. Y. Desta and R. R. T. Majinda, Nat. Prod. Commun., 2014, 9, 817–820 Search PubMed .
  50. S. El-Masry, M. H. Hala, H. H. Zaatout and M. S. Abdel-Kader, Nat. Prod. Sci., 2010, 16, 211–216 Search PubMed .
  51. M. Na, J. Jang, D. Njamen, J. T. Mbafor, Z. T. Fomum, B. Y. Kim, W. K. Oh and J. S. Ahn, J. Nat. Prod., 2006, 69, 1572–1576 Search PubMed .
  52. J. Jang, M. Na, P. T. Thuong, D. Njamen, J. T. Mbafor, Z. T. Fomum, E.-R. Woo and W. K. Oh, Chem. Pharm. Bull., 2008, 56, 85–88 Search PubMed .
  53. R. F. Tchokouaha, X. Alexi, E. Chosson, T. Besson, A.-L. Skaltsounis, E. Seguin, M. N. Alexis and J. Wandji, J. Enzyme Inhib. Med. Chem., 2010, 25, 228–233 Search PubMed .
  54. M. S. Ali, M. I. Ali, G. Ahmed, N. Afza, M. Lateef, L. Iqbal, A. F. K. Waffo and Z. Ahmed, Z. Naturforsch., B: J. Chem. Sci., 2012, 67, 98–102 Search PubMed .
  55. E. Talla, E. Yankep and J. Tanyi Mbafor, Bull. Chem. Soc. Ethiop., 2014, 28, 155 Search PubMed .
  56. L. A. Mitscher, S. K. Okwute, S. R. Gollapudi, S. Drake and E. Avona, Phytochemistry, 1988, 27, 3449–3452 Search PubMed .
  57. M. I. Ali, Z. Ahmed, A. F. K. Waffo and M. S. Ali, Nat. Prod. Commun., 2010, 5, 889–892 Search PubMed .
  58. K. K. Atindehou, E. F. Queiroz, C. Terreaux, D. Traore and K. Hostettmann, Planta Med., 2002, 68, 181–182 Search PubMed .
  59. A. K. Waffo, G. A. Azebaze, A. E. Nkengfack, Z. T. Fomum, M. Meyer, B. Bodo and F. R. Van Heerden, Phytochemistry, 2000, 53, 981–985 Search PubMed .
  60. Z. T. Fomum, J. F. Ayafor, J. T. Mbafor and C. M. Mbi, J. Chem. Soc., Perkin Trans. 1, 1986, 33–37 Search PubMed .
  61. R. Promsattha, M. S. Tempesta, Z. T. Fomum, J. F. Ayafor and J. T. Mbafor, J. Nat. Prod., 1986, 49, 932–933 Search PubMed .
  62. R. Promsattha, M. S. Tempesta, Z. T. Fomum and J. T. Mbafor, J. Nat. Prod., 1988, 51, 611–613 Search PubMed .
  63. A. E. Nkengfack, J. Kouam, W. T. Vouffo, Z. T. Fomum, E. Dagne, O. Sterner, L. M. Browne and G. Ji, Phytochemistry, 1993, 32, 1305–1311 Search PubMed .
  64. D. Djamen, Planta Med., 2004, 70, 104–107 Search PubMed .
  65. J. Kouam, X. Siewe Noundou, L. B. Kouitcheu Mabeku, A. Meli Lannang, M. Iqbal Choudhary and Z. T. Fomum, Bull. Chem. Soc. Ethiop., 2007, 21, 373–378 Search PubMed .
  66. J. Kouam, A. L. Meli, V. Kuete and Z. T. Fomum, Nat. Prod. Commun., 2008, 3, 1934578X0800301 Search PubMed .
  67. M. S. Ali, M. I. Ali, Z. Ahmed and P. A. Onocha, Nat. Prod. Commun., 2011, 6, 1099–1102 Search PubMed .
  68. A. E. Nkengfack, T. W. Vouffo, J. C. Vardamides, J. Kouam, Z. T. Fomum, M. Meyer and O. Sterner, Phytochemistry, 1997, 46, 573–578 Search PubMed .
  69. A. E. Nkengfack, A. G. B. Azebaze, A. K. Waffo, Z. T. Fomum, M. Meyer and F. R. Van Heerden, Phytochemistry, 2001, 58, 1113–1120 Search PubMed .
  70. T. T. Dao, P. H. Nguyen, P. T. Thuong, K. W. Kang, M. Na, D. T. Ndinteh, J. T. Mbafor and W. K. Oh, Phytochemistry, 2009, 70, 2053–2057 Search PubMed .
  71. S. El-Masry, M. E. Amer, M. S. Abdel-Kader and H. H. Zaatout, Phytochemistry, 2002, 60, 783–787 Search PubMed .
  72. B. F. Juma and R. R. T. Majinda, Nat. Prod. Commun., 2006, 1, 1934578X0600100 Search PubMed .
  73. A. Yenesew, J. O. Midiwo, M. Heydenreich and M. G. Peter, Phytochemistry, 1998, 49, 247–249 Search PubMed .
  74. A. Yenesew, J. O. Midiwo, M. Heydenreich, D. Schanzenbach and M. G. Peter, Phytochemistry, 2000, 55, 457–459 Search PubMed .
  75. A. W. Andayi, A. Yenesew, S. Derese, J. O. Midiwo, P. M. Gitu, O. J. Jondiko, H. Akala, P. Liyala, J. Wangui, N. C. Waters, M. Heydenreich and M. G. Peter, Planta Med., 2006, 72, 187–189 Search PubMed .
  76. J. O. Ombito, R. R. T. Majinda, I. B. Masesane, G. Bojase, A. Schüffler and T. Opatz, Phytochem. Lett., 2018, 26, 110–114 Search PubMed .
  77. E. Bae, M. Na, D. Njamen, J. Mbafor, Z. Fomum, L. Cui, D. Choung, B. Kim, W. Oh and J. Ahn, Planta Med., 2006, 72, 945–948 Search PubMed .
  78. P. H. Nguyen, G. Sharma, T. T. Dao, M. N. Uddin, K. W. Kang, D. T. Ndinteh, J. T. Mbafor and W. K. Oh, Bioorg. Med. Chem., 2012, 20, 6459–6464 Search PubMed .
  79. W. Wätjen, A. Kulawik, A. K. Suckow-Schnitker, Y. Chovolou, R. Rohrig, S. Ruhl, A. Kampkötter, J. Addae-Kyereme, C. W. Wright and C. M. Passreiter, Toxicology, 2007, 242, 71–79 Search PubMed .
  80. W. Wätjen, A. K. Suckow-Schnitker, R. Rohrig, A. Kulawik, J. Addae-Kyereme, C. W. Wright and C. M. Passreiter, J. Nat. Prod., 2008, 71, 735–738 Search PubMed .
  81. C. M. Passreiter, A.-K. Suckow-Schnitker, A. Kulawik, J. Addae-Kyereme, C. W. Wright and W. Wätjen, Phytochemistry, 2015, 117, 237–244 Search PubMed .
  82. M. Chacha, G. Bojasemoleta and R. Majinda, Phytochemistry, 2005, 66, 99–104 Search PubMed .
  83. C. C. W. Wanjala and R. R. T. Majinda, Fitoterapia, 2000, 71, 400–405 Search PubMed .
  84. Y. Zarev, K. Foubert, V. Lucia De Almeida, R. Anthonissen, E. Elgorashi, S. Apers, I. Ionkova, L. Verschaeve and L. Pieters, Phytochemistry, 2017, 141, 140–146 Search PubMed .
  85. E. Dagne, A. A. L. Gunatilaka, D. G. I. Kingston, M. Alemu, G. Hofmann and R. K. Johnson, J. Nat. Prod., 1993, 56, 1831–1834 Search PubMed .
  86. F. Machumi, G. Bojase-Moleta, R. Mapitse, I. Masesane and R. R. T. Majinda, Nat. Prod. Commun., 2006, 1, 1934578X0600100 Search PubMed .
  87. M. Moriyasu, M. Ichimaru, Y. Nishiyama, A. Kato, S. G. Mathenge, F. D. Juma and J. N. Nganga, J. Nat. Prod., 1998, 61, 185–188 Search PubMed .
  88. A. Yenesew, D. Solomon, I. Beatrice, J. O. Midiwo, N. C. Waters, P. Liyala, H. Akala and M. G. Peter, Planta Med., 2003, 69, 658–661 Search PubMed .
  89. M. Ichimaru, M. Moriyasu, Y. Nishiyama, A. Kato, S. G. Mathenge, F. D. Juma and J. N. Nganga, J. Nat. Prod., 1996, 59, 1113–1116 Search PubMed .
  90. L. Cui, P. T. Thuong, H. S. Lee, D. T. Ndinteh, J. T. Mbafor, Z. T. Fomum and W. K. Oh, Bioorg. Med. Chem., 2008, 16, 10356–10362 Search PubMed .
  91. L. Cui, H. Lee, D. Ndinteh, J. Mbafor, Y. Kim, T. Le, P. Nguyen and W. Oh, Planta Med., 2010, 76, 713–718 Search PubMed .
  92. P. H. Nguyen, T. T. Dao, J. Kim, D. T. Phong, D. T. Ndinteh, J. T. Mbafor and W. K. Oh, Bioorg. Med. Chem., 2011, 19, 3378–3383 Search PubMed .
  93. K. G. Bedane, S. Kusari, D. Eckelmann, I. B. Masesane, M. Spiteller and R. R. T. Majinda, Fitoterapia, 2015, 105, 113–118 Search PubMed .
  94. G. T. Gurmessa, S. Kusari, H. Laatsch, G. Bojase, G. Tatolo, I. B. Masesane, M. Spiteller and R. R. T. Majinda, Phytochem. Lett., 2018, 25, 37–42 Search PubMed .
  95. A. Yenesew, B. Irungu, S. Derese, J. O. Midiwo, M. Heydenreich and M. G. Peter, Phytochemistry, 1998, 63, 445–448 Search PubMed .
  96. E. Talla, N. Sidoine, N. Jacqueline, N. Gisele, Y. Euml, L. N. Elisabeth, M. Joseph and N. Nicolas, J. Appl. Pharm. Sci., 2015, 001–005 Search PubMed .
  97. K. G. Bedane, S. Kusari, A. Bullach, I. B. Masesane, S. O. Mihigo, M. Spiteller and R. R. T. Majinda, Phytochem. Lett., 2017, 20, 84–88 Search PubMed .
  98. R. B. Taylor, D. G. Corley, M. S. Tempesta, Z. T. Fomum, J. F. Ayafor, J. Wandji and P. N. Ifeadike, J. Nat. Prod., 1986, 49, 670–673 Search PubMed .
  99. V. Kuete, L. P. Sandjo, D. E. Djeussi, M. Zeino, G. M. N. Kwamou, B. Ngadjui and T. Efferth, Invest. New Drugs, 2014, 32, 1053–1062 Search PubMed .
  100. W. Oh, H. Lee, S. Ahn, J. Ahn, J. Mbafor, J. Wandji, Z. Fomum, H. Chang and Y. Kim, Phytochemistry, 1999, 51, 1147–1150 Search PubMed .
  101. J. Lee, W. Oh, J. Ahn, Y. Kim, J. Mbafor, J. Wandji and Z. Fomum, Planta Med., 2009, 75, 268–270 Search PubMed .
  102. Z. T. Fomum, J. F. Ayafor and J. Wandji, J. Nat. Prod., 1987, 50, 921–922 Search PubMed .
  103. J. Wandji, A. E. Nkengfack, Z. T. Fomum, R. Ubillas, K. B. Killday and M. S. Tempesta, J. Nat. Prod., 1990, 53, 1425–1429 Search PubMed .
  104. J. Wandji, Z. T. Fomum, F. Tillequin, E. Seguin and M. Koch, Phytochemistry, 1994, 35, 245–248 Search PubMed .
  105. Z. T. Fomum, J. Foyere Ayafor and J. Wandji, Phytochemistry, 1985, 24, 3075–3076 Search PubMed .
  106. J. H. Donfack, F. N. Nico, B. Ngameni, A. Tchana, P. D. Chuisseu, P. V. Finzi, B. Ngadjui and P. F. Moundipa, Asian J. Tradit. Med., 2008, 3, 172–179 Search PubMed .
  107. A. Togola, B. Hedding, A. Theis, H. Wangensteen, F. Rise, B. Smestad Paulsen, D. Diallo and K. Egil Malterud, Planta Med., 2009, 75, 1168–1170 Search PubMed .
  108. V. Kuete, L. P. Sandjo, G. M. N. Kwamou, B. Wiench, A. E. Nkengfack and T. Efferth, Phytomedicine, 2014, 21, 682–688 Search PubMed .
  109. M. N. Ombuna, A. K. Machocho, M. M. Ng’ang’a, G. K. Keru, A. W. Wanyonyi and O. Amuka, J. Med. Plants Res., 2016, 10, 686–694 Search PubMed .
  110. K. Koch, G. Schulz, W. Döring, C. Büchter, S. Havermann, P. C. Mutiso, C. Passreiter and W. Wätjen, J. Pharm. Pharmacol., 2019, 71, 1007–1016 Search PubMed .
  111. W. Hauschild, P. B. C. Mutiso and C. M. Passreiter, Nat. Prod. Commun., 2010, 5, 1934578X1000500 Search PubMed .
  112. S. S. Nyandoro, J. J. E. Munissi, M. Kombo, C. A. Mgina, F. Pan, A. Gruhonjic, P. Fitzpatrick, Y. Lu, B. Wang, K. Rissanen and M. Erdélyi, J. Nat. Prod., 2017, 80, 377–383 Search PubMed .
  113. V. S. Kamat, K. Nakanishi, F. Y. Chuo and I. Kubo, Heterocycles, 1981, 15, 1163 Search PubMed .
  114. L. Biyiti, D. Pesando and S. Puiseux-Dao, Planta Med., 1988, 54, 126–128 Search PubMed .
  115. R. Promsattha, J. T. Mbafor, M. S. Tempesta and Z. T. Fomum, J. Nat. Prod., 1989, 52, 1316–1318 Search PubMed .
  116. J. Kouam, F.-X. Etoa, L. B. Mabeku and Z. T. Fomum, Nat. Prod. Commun., 2007, 2, 1934578X0700201 Search PubMed .
  117. A. E. Nkengfack, D. R. Sanson, M. S. Tempesta and Z. T. Fomum, J. Nat. Prod., 1989, 52, 320–324 Search PubMed .
  118. Z. Y. Desta, N. Sewald and R. R. T. Majinda, Nat. Prod. Res., 2014, 28, 667–673 Search PubMed .
  119. M. S. Ali, M. I. Ali, P. A. Onocha and Z. Ahmed, J. Asian Nat. Prod. Res., 2011, 13, 182–187 Search PubMed .
  120. J. Takashima and A. Ohsaki, J. Nat. Prod., 2002, 65, 1843–1847 Search PubMed .
  121. K. G. Bedane, S. Kusari, I. B. Masesane, M. Spiteller and R. R. T. Majinda, Fitoterapia, 2016, 108, 48–54 Search PubMed .
  122. L. Cui, D. T. Ndinteh, M. Na, P. T. Thuong, J. Silike-Muruumu, D. Njamen, J. T. Mbafor, Z. T. Fomum, J. S. Ahn and W. K. Oh, J. Nat. Prod., 2007, 70, 1039–1042 Search PubMed .
  123. P. H. Nguyen, T. T. Dao, J. Kim, D. T. Phong, D. T. Ndinteh, J. T. Mbafor and W. K. Oh, Bioorg. Med. Chem., 2011, 19, 3378–3383 Search PubMed .
  124. A. Yenesew, B. Irungu, S. Derese, J. O. Midiwo, M. Heydenreich and M. G. Peter, Phytochemistry, 2003, 63, 445–448 Search PubMed .
  125. E. Talla, A. J. Y. Gbaweng, L. Mokale, J. P. Abdou and J. T. Mbafor, Nat. Prod.: Indian J., 2016, 12, 1–9 Search PubMed .
  126. A. F. K. Waffo, P. Coombes, D. Mulholland, A. Nkengfack and Z. Fomum, Phytochemistry, 2006, 67, 459–463 Search PubMed .
  127. A. Yenesew, M. Induli, S. Derese, J. O. Midiwo, M. Heydenreich, M. G. Peter, H. Akala, J. Wangui, P. Liyala and N. C. Waters, Phytochemistry, 2004, 65, 3029–3032 Search PubMed .
  128. L. Cui, P. Thuong, H. Lee, D. Njamen, J. Mbafor, Z. Fomum, J. Lee, Y. Kim and W. Oh, Planta Med., 2008, 74, 422–426 Search PubMed .
  129. A. E. Nkengfack, T. W. Vouffo, Z. T. Fomum, M. Meyer, O. Bergendorff and O. Sterner, Phytochemistry, 1994, 36, 1047–1051 Search PubMed .
  130. Z. Y. Desta, N. Sewald and R. R. T. Majinda, Bull. Chem. Soc. Ethiop., 2016, 30, 427 Search PubMed .
  131. E. F. Queiroz, K. K. Atindehou, C. Terreaux, S. Antus and K. Hostettmann, J. Nat. Prod., 2002, 65, 403–406 Search PubMed .
  132. A. E. Nkengfack, T. W. Vouffo, J. C. Vardamides, Z. T. Fomum, O. Bergendorff and O. Sterner, J. Nat. Prod., 1994, 57, 1172–1177 Search PubMed .
  133. H. Tanaka, T. Tanaka, A. Hosoya, Y. Kitade and H. Etoh, Phytochemistry, 1998, 48, 355–357 Search PubMed .
  134. J. Wandji, Z. T. Fomum, F. Tillequin, F. Libot and M. Koch, J. Nat. Prod., 1995, 58, 105–108 Search PubMed .
  135. A. E. Nkengfack, D. R. Sanson, Z. T. Fomum and M. S. Tempesta, Phytochemistry, 1989, 28, 2522–2526 Search PubMed .
  136. A. E. Nkengfack, Z. T. Fomum, R. Ubillas and M. S. Tempesta, J. Nat. Prod., 1990, 53, 1552–1556 Search PubMed .
  137. A. E. Nkengfack, Z. T. Fomum, R. Ubillas, D. R. Sanson and M. S. Tempesta, J. Nat. Prod., 1990, 53, 509–512 Search PubMed .
  138. A. Nkengfack, M. Meyer, M. Tempesta and Z. Fomum, Planta Med., 1991, 57, 488–491 Search PubMed .
  139. J. Wandji, S. Awanchiri, Z. Tanee Fomum, F. Tillequin and F. Libot, Phytochemistry, 1995, 39, 677–681 Search PubMed .
  140. J. Wandji, Z. T. Fomum, F. Tillequin, G. Baudouin and M. Koch, Phytochemistry, 1994, 35, 1573–1577 Search PubMed .
  141. K. G. Bedane, I. B. Masesane and R. R. T. Majinda, Phytochem. Lett., 2016, 17, 55–58 Search PubMed .
  142. S. M. Nassief, M. E. Amer, E. Shawky, K. Sishtla, E. Mas-Claret, A. Muniyandi, T. W. Corson, D. A. Mulholland and S. El-Masry, J. Nat. Prod., 2023, 86, 759–766 Search PubMed .
  143. A. Yenesew, J. O. Midiwo, S. M. Guchu, M. Heydenreich and M. G. Peter, Phytochemistry, 2002, 59, 337–341 Search PubMed .
  144. A. Nkengfack, Phytochemistry, 1995, 40, 1803–1808 Search PubMed .
  145. P. H. Nguyen, T. V. T. Le, P. T. Thuong, T. T. Dao, D. T. Ndinteh, J. T. Mbafor, K. W. Kang and W. K. Oh, Bioorg. Med. Chem. Lett., 2009, 19, 6745–6749 Search PubMed .
  146. P. H. Nguyen, T. N. A. Nguyen, K. W. Kang, D. T. Ndinteh, J. T. Mbafor, Y. R. Kim and W. K. Oh, Bioorg. Med. Chem., 2010, 18, 3335–3344 Search PubMed .
  147. N. M. Fahmy, E. Al-Sayed, M. El-Shazly and A. Nasser Singab, Nat. Prod. Res., 2020, 34, 1891–1912 Search PubMed .
  148. A. F. Parsons and M. J. Palframan, in The Alkaloids: Chemistry and Biology, Elsevier, 2010, vol. 68, pp. 39–81 Search PubMed .
  149. D. Zofou, F. Ntie-Kang, W. Sippl and S. M. N. Efange, Nat. Prod. Rep., 2013, 30, 1098 Search PubMed .
  150. H. Tanaka, I. Atsumi, M. Hasegawa, M. Hirata, T. Sakai, M. Sato, R. Yamaguchi, Y. Tateishi, T. Tanaka and T. Fukai, Nat. Prod. Commun., 2015, 10, 499–501 Search PubMed .
  151. H. Tanaka, H. Hattori, T. Oh-Uchi, M. Sato, R. Yamaguchi, M. Sako and Y. Tateishi, J. Asian Nat. Prod. Res., 2008, 10, 983–987 Search PubMed .
  152. H. Tanaka, T. Oh-Uchi, H. Etoh, M. Sako, F. Asai, T. Fukai, M. Sato, J. Murata and Y. Tateishi, Phytochemistry, 2003, 64, 753–758 Search PubMed .
  153. T. L. Adetunji, M. A. Acho, V. O. Samuel, C. R. Ohoro and M. Ramulondi, J. Ethnopharmacol., 2024, 319, 117273 Search PubMed .
  154. S. Kumar, A. S. Pathania, A. K. Saxena, R. A. Vishwakarma, A. Ali and S. Bhushan, Chem.-Biol. Interact., 2013, 205, 128–137 Search PubMed .
  155. H. Tanaka, T. Tanaka and H. Etoh, Phytochemistry, 1997, 45, 835–838 Search PubMed .
  156. H. Tanaka, M. Hirata, H. Etoh, M. Sako, M. Sato, J. Murata, H. Murata, D. Darnaedi and T. Fukai, Heterocycles, 2003, 60(12), 2767–2773 Search PubMed .
  157. H. Tanaka, M. Sudo, M. Hirata, H. Etoh, M. Sato, R. Yamaguchi, E. Sakai, I.-S. Chen and T. Fukai, Nat. Prod. Commun., 2010, 5, 1781–1784 Search PubMed .
  158. J. Morante-Carriel, H. Nájera, A. Samper-Herrero, S. Živković, M. J. Martínez-Esteso, A. Martínez-Márquez, S. Sellés-Marchart, A. Obrebska and R. Bru-Martínez, Int. J. Mol. Sci., 2024, 25, 13036 Search PubMed .
  159. A. Yenesew, S. Derese, J. O. Midiwo, C. C. Bii, M. Heydenreich and M. G. Peter, Fitoterapia, 2005, 76, 469–472 Search PubMed .
  160. A. Yenesew, J. O. Midiwo, M. Miessner, M. Heydenreich and M. G. Peter, Phytochemistry, 1998, 48, 1439–1443 Search PubMed .
  161. A. Yenesew, H. Twinomuhwezi, B. Kiremire, M. Mbugua, P. Gitu, M. Heydenreich and M. Peter, Bull. Chem. Soc. Ethiop., 2009, 23(2), 205–210 Search PubMed .
  162. B. Tsakem, B. T. Toussie, X. Siwe-Noundou, B. K. Ponou, R. B. Teponno, S. G. Musharraf and L. A. Tapondjou, Magn. Reson. Chem., 2023, 61(8), 497–503 Search PubMed .
  163. Y.-M. Hsu, W.-C. Lai, C.-Y. Li, Y.-H. Lan, Y.-C. Tsai, A. Hunyadi, M.-F. Hou, S. S. Yuan, Y.-C. Wu and F.-R. Chang, Nat. Prod. Commun., 2016, 11, 1099–1102 Search PubMed .
  164. G. Galati and P. J. O'Brien, Free Radical Biol. Med., 2004, 37, 287–303 Search PubMed .
  165. B. Botta, A. Vitali, P. Menendez, D. Misiti and G. Monache, Curr. Med. Chem., 2005, 12, 713–739 Search PubMed .
  166. A. Sychrová, G. Škovranová, M. Čulenová and S. Bittner Fialová, Molecules, 2022, 27, 4491 Search PubMed .
  167. T. Rukachaisirikul, P. Innok, N. Aroonrerk, W. Boonamnuaylap, S. Limrangsun, C. Boonyon, U. Woonjina and A. Suksamrarn, J. Ethnopharmacol., 2007, 110, 171–175 Search PubMed .
  168. D. E. Djeussi, L. P. Sandjo, J. A. K. Noumedem, L. K. Omosa, B. T. Ngadjui and V. Kuete, BMC Complementary Altern. Med., 2015, 15, 453 Search PubMed .
  169. N. Cotelle, J.-L. Bernier, J.-P. Catteau, J.-P. Henichart and D. Pesando, Nat. Prod. Lett., 1993, 3, 79–86 Search PubMed .
  170. K. Šmejkal, Phytochem. Rev., 2014, 13, 245–275 Search PubMed .
  171. L. Molčanová, D. Janošíková, S. Dall'Acqua and K. Šmejkal, Phytochem. Rev., 2019, 18, 1051–1100 Search PubMed .
  172. S. A. Mir, A. Dar, L. Hamid, N. Nisar, J. A. Malik, T. Ali and G. N. Bader, Curr. Res. Pharmacol. Drug Discov., 2024, 6, 100167 Search PubMed .
  173. R. Mashima and T. Okuyama, Redox Biol., 2015, 6, 297–310 Search PubMed .
  174. S. Habtemariam and E. Dagne, Planta Med., 2010, 76, 589–594 Search PubMed .
  175. M. C. B. Nde, D. Njamen, S. Tanee Fomum, J. Wandji, E. Simpson, C. Clyne and G. Vollmer, Eur. J. Pharmacol., 2012, 674, 87–94 Search PubMed .
  176. B. Wang, Q. Wang, R. Yuan, S. Yang, M. Lu, F. Yuan, Z. Dong, M. Mo, Q. Pan and H. Gao, Chin. Med., 2023, 18, 153 Search PubMed .
  177. C. Bailly, Asian J. Nat. Prod. Biochem., 2021, 19(2), 45–57 Search PubMed .
  178. R. Mukai, Biosci., Biotechnol., Biochem., 2018, 82, 207–215 Search PubMed .
  179. A. Hisanaga, R. Mukai, K. Sakao, J. Terao and D. Hou, Mol. Nutr. Food Res., 2016, 60, 1020–1032 Search PubMed .
  180. P. Basu and A. Basu, Molecules, 2020, 25, 1171 Search PubMed .
  181. S. Sokeng, E. Talla, V. Jeweldai, J. Koubé and F. Dongmo, J. Drugs Med., 2013, 5, 121–128 Search PubMed .
  182. A. Yenesew, H. M. Akala, H. Twinomuhwezi, C. Chepkirui, B. N. Irungu, F. L. Eyase, M. Kamatenesi-Mugisha, B. T. Kiremire, J. D. Johnson and N. C. Waters, Acta Trop., 2012, 123, 123–127 Search PubMed .
  183. T. Behl, A. Gupta, A. Sehgal, A. Albarrati, M. Albratty, A. M. Meraya, A. Najmi, S. Bhatia and S. Bungau, Biomed. Pharmacother., 2022, 153, 113405 Search PubMed .
  184. C. Barba-Ostria, S. E. Carrera-Pacheco, R. Gonzalez-Pastor, J. Heredia-Moya, A. Mayorga-Ramos, C. Rodríguez-Pólit, J. Zúñiga-Miranda, B. Arias-Almeida and L. P. Guamán, Molecules, 2022, 27, 4490 Search PubMed .
  185. Y. Yarden, Oncology, 2001, 61, 1–13 Search PubMed .
  186. A. Citri and Y. Yarden, Nat. Rev. Mol. Cell Biol., 2006, 7, 505–516 Search PubMed .
  187. C. Gutierrez and R. Schiff, Arch. Pathol. Lab. Med., 2011, 135, 55–62 Search PubMed .
  188. N. Iqbal and N. Iqbal, Mol. Biol. Int., 2014, 2014, 1–9 Search PubMed .
  189. J. Jeong, W. Kim, L. K. Kim, J. VanHouten and J. J. Wysolmerski, PLoS One, 2017, 12, e0174849 Search PubMed .
  190. L. N. Klapper, S. Glathe, N. Vaisman, N. E. Hynes, G. C. Andrews, M. Sela and Y. Yarden, Proc. Natl. Acad. Sci. U. S. A., 1999, 96, 4995–5000 Search PubMed .
  191. M. Christgen, S. Bartels, A. Luft, S. Persing, D. Henkel, U. Lehmann and H. Kreipe, Virchows Arch., 2018, 473, 577–582 Search PubMed .
  192. R. Roskoski, Pharmacol. Res., 2014, 79, 34–74 Search PubMed .
  193. A. Kumar, E. T. Petri, B. Halmos and T. J. Boggon, J. Clin. Oncol., 2008, 26, 1742–1751 Search PubMed .
  194. S. E. D. C. Jorge, S. S. Kobayashi and D. B. Costa, Braz. J. Med. Biol. Res., 2014, 47, 929–939 Search PubMed .
  195. W. Yan, Y. Zhao and J. He, Mol. Med. Rep., 2018, 18, 4175–4184 Search PubMed .
  196. C.-H. Yun, K. E. Mengwasser, A. V. Toms, M. S. Woo, H. Greulich, K.-K. Wong, M. Meyerson and M. J. Eck, Proc. Natl. Acad. Sci. U. S. A., 2008, 105, 2070–2075 Search PubMed .
  197. M. S. Zubair, S. Anam, S. Maulana and M. Arba, Indones. J. Chem., 2021, 21, 310 Search PubMed .
  198. A. N. J. Imanuddin, I. Wiani, A. Hardianto and T. Herlina, Trends Sci., 2023, 20, 5350 Search PubMed .
  199. S. Beyerstedt, E. B. Casaro and É. B. Rangel, Eur. J. Clin. Microbiol. Infect. Dis., 2021, 40, 905–919 Search PubMed .
  200. W. Ni, X. Yang, D. Yang, J. Bao, R. Li, Y. Xiao, C. Hou, H. Wang, J. Liu, D. Yang, Y. Xu, Z. Cao and Z. Gao, Crit. Care, 2020, 24, 422 Search PubMed .
  201. J. M. Penninger, M. B. Grant and J. J. Y. Sung, Gastroenterology, 2021, 160, 39–46 Search PubMed .
  202. T. Herlina, A. W. R. Akili, V. Nishinarizki, A. Hardianto and J. Latip, Adv. Appl. Bioinf. Chem., 2024, 17, 61–70 Search PubMed .

Footnote

Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d5ra03457d

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.