Genotoxicity and Teratogenicity of African Medicinal Plants

Genotoxicity and Teratogenicity of African Medicinal Plants

9 Genotoxicity and Teratogenicity of African Medicinal Plants Armel Jackson Seukep, Jaures A.K. Noumedem, Doriane E. Djeussi and Victor Kuete Faculty ...

390KB Sizes 10 Downloads 284 Views

9 Genotoxicity and Teratogenicity of African Medicinal Plants Armel Jackson Seukep, Jaures A.K. Noumedem, Doriane E. Djeussi and Victor Kuete Faculty of Science, Department of Biochemistry, University of Dschang, Dschang, Cameroon

9.1

Introduction

Genotoxicity is the ability of chemicals to damage the genetic information within a cell resulting in mutations, which may lead to malignancies. The genotoxic substances induce damage to the genetic material in the cells through interactions with the DNA sequence and structure. Most often, genotoxicity is confused with mutagenicity as all mutagenic chemicals are genotoxic; however, not all genotoxic compounds are mutagenic. Teratogenic substances induce abnormalities of physiological development and the study involves human congenital abnormalities, but it is much broader than that, taking in other nonbirth developmental stages, including puberty, and other nonhuman life forms, including plants. The developmental toxicity takes in account all manifestations of abnormal development by toxic substances and can either be caused toxic substances (xenobiotics including plantbased drugs), transmitted infection, lack of nutrients (e.g., lack of folic acid in nutrition during pregnancy for humans can result in spina bifida), physical restraint (e.g., Potter syndrome due to oligohydramnios in humans), and genetic disorders. Worldwide and especially in Africa, the population does not pay enough attention to the toxic potencies of medicinal plants contrary to other chemicals, believing that, if these products have been used so far, they should be devoid of toxicity [16]. However, compiled data highlighting the genotoxicity and teratogenicity of African medicinal plants are scarce, though there are several scientific publications documenting the effects of individual plants. This chapter aims not only to review the genotoxicity and teratogenicity of the medicinal plants used in African traditional medicine (ATM), but also to bring together information regarding the ability of plant used in the continent to display genoprotective effects.

Toxicological Survey of African Medicinal Plants. DOI: http://dx.doi.org/10.1016/B978-0-12-800018-2.00009-1 © 2014 Elsevier Inc. All rights reserved.

236

9.2

Toxicological Survey of African Medicinal Plants

Methods Used in Genotoxicity Assays

The investigation of DNA damage in cells exposed to the toxic substrates is the main way to assess the genotoxicity of a chemical. This DNA damage can be in the form of single- and double-strand breaks, loss of excision repair, cross-linking, alkali-labile sites, point mutations, and structural and numerical chromosomal aberrations [7]. Pronounced alteration or a compromised integrity of chromosome can lead to various types of cancers. Therefore, many sophisticated techniques including the Ames assay, in vitro and in vivo toxicology tests, and the Comet assay have been developed to assess the chemicals’ potential to cause DNA damage that may lead to cancer. Table 9.1 below summarizes the different validated nonanimal alternatives used in genotoxicity screening of chemicals as retrieved from AltTox.org database [7]. The genotoxicity testing is performed in bacterial, yeast, and mammalian cells and is aimed at determining if a substrate will influence genetic material or may cause cancer [7]. In laboratories, the test for gene mutation is based on the Bacterial Reverse Mutation Assay, also known as the Ames assay. In this assay, different bacterial strains are used in order to compare the different changes in the genetic material. This has led to the detection of the majority of genotoxic carcinogens and genetic changes and the types of mutations detected are frame shifts and base substitutions [8]. The in vitro toxicology testing is used to determine whether a substrate, product, or environmental factor induces genetic damage. This technique detects chromatid

Table 9.1 Validated Nonanimal Alternatives Methods in Genotoxicity Study of Chemicals [7] Methods

Test Purpose

In vitro mammalian cell micronucleus test

Alternative to the in vitro chromosome aberration assay for genotoxicity testing Genotoxicity testing—using bacterial cells Genotoxicity testing—using bacterial cells

Bacterial reverse mutation test (Ames Test) Genetic toxicology: Saccharomyces cerevisiae gene mutation assay Genetic toxicology: Saccharomyces cerevisiae mitotic recombination assay In vitro mammalian cell gene mutation test Genetic toxicology: in vitro sister chromatid exchange assay in mammalian cells Genetic toxicology: DNA damage and repair, unscheduled DNA synthesis in mammalian cells in vitro

Genotoxicity testing—using bacterial cells Genotoxicity testing—using mammalian cells Genotoxicity testing—using mammalian cells Genotoxicity testing—using mammalian cells

Genotoxicity and Teratogenicity of African Medicinal Plants

237

and chromosome gaps, chromosome breaks, chromatid deletions, fragmentation, translocation, complex rearrangements, and many more [8]. The in vivo testing determines the potential of DNA damage that can affect chromosomal structure or disturb the mitotic apparatus that changes chromosome number. Most often, the Comet assay is used in vivo to detect the genotoxicity of a substance. The technique involves cells lysis using detergents and salts. The DNA released from the lysed cells is electrophoresed in an agarose gel under neutral pH conditions. Cells containing DNA with an increased number of double-stranded breaks will migrate more quickly to the anode [9,10].

9.3

Methods Used in Teratogenicity Assays

Teratogenesis is the disturbed growth process involved in the production of a malformed neonate. There are six principles of teratology as defined by Wilson since 1959 [11]. These principles guide the study and understanding of teratogenic agents and their effects on developing organisms: G

G

G

G

G

G

Susceptibility to teratogenesis depends on the genotype of the conceptus and the manner in which this interacts with adverse environmental factors. Susceptibility to teratogenesis varies with the developmental stage at the time of exposure to an adverse influence. There are critical periods of susceptibility to agents and organ systems affected by these agents. Teratogenic agents act in specific ways on developing cells and tissues to initiate sequences of abnormal developmental events. The access of adverse influences to developing tissues depends on the nature of the influence. Several factors affect the ability of a teratogen to contact a developing conceptus, such as the nature of the agent itself, route, and degree of maternal exposure, rate of placental transfer and systemic absorption, and composition of the maternal and embryonic/ fetal genotypes. There are four manifestations of deviant development (death, malformation, growth retardation, and functional defect). Manifestations of deviant development increase in frequency and degree as dosage increases from the no observable adverse effect level (NOAEL) to a dose producing 100% lethality (LD100).

The screenings of the potential teratogenicity of chemicals or environmental agents use animal model systems such as rat, mouse, rabbit, dog, and monkey. In the past, pregnant animals were exposed to the chemical and their fetuses were observed for gross visceral and skeletal abnormalities. Though this is still part of the teratological evaluation procedures today, the field of teratology is moving to a more molecular level, seeking the mechanism(s) of action by which these agents act. Animals such as genetically modified mice are commonly used nowadays. The study of the teratogenicity of xenobiotics is important in preventing congenital abnormalities and also has the potential for developing new safe therapeutic drugs for use with pregnant women.

238

9.4

Toxicological Survey of African Medicinal Plants

Medicinal Plants with Genotoxic Effects

Pyrrolizidine alkaloids (PAs) are a well-known example of a genotoxic substance causing DNA damage. The transition metal chromium interacts with DNA in its high-valent oxidation state so to incur DNA lesions, leading to carcinogenesis. Medicinal plants worldwide containing PAs and several other metabolites were found to be genotoxic. Taylor et al. [12] demonstrated genotoxicity of many medicinal plants from South Africa. They used samples made of methanol and dichloreomethane extracts of the following plants: leaves of Balanites maughamii, Catharanthus roseus (Apocyanaceae), Catunaregam spinosa, Chaetacme aristata (Rubiaceae), Plumbago auriculata (Plumbaginaceae), Turraea floribunda (Meliaceae), Vernonia colorata (Asteraceae), Trichelia emetica (Meliaceae) and Rhamnus prinoides (Rhamnaceae), bark of Ocotea bulata (Lauraceae), Prunus africana (Rosaceae), Boophane disticha (Amaryllidaceae), Combretum mkhzense (Combretaceae), Crinum macowanii (Amaryllidaceae), Erythrina caffra (Leguminosae), Hypoxis colchicifolia, Hypoxis hemerocallidea and Ornithogalum longibracteatum (Hyacinthaceae), Ricinus comminus (Euphorbiaceae), Scilla natalensis (Liliaceae), Sclerocarya birrea (Anacardiaceae), Trichelia emetic (Meliaceae); twigs and leaves of Diospyros whyteana (Ebenaceae); twigs and bark of Spirostachys africana, Croton sylvaticus (Euphorbiaceae), and Gardenia volkensii (Rubiaceae). They demonstrated in Alkaline Comet/Single Cell Gel Electrophoresis (SCGE) Assay that these plants exert DNA damage in cells at concentration of 250 and 500 ppm. Some of the extracts also displayed genotoxicity by increasing the number of micronuclei in dividing human blood cells. It has been shown that Arictum lappa root extract induces chromosomal aberrations, micronuclei (MNC) formations in Allium cepa root tip cells. It also induced mitotic spindle disturbance and presented mitodepressive effects in Allium cepa by decreasing the mitotic index with more pronounced effects at 62.5 and 125 mg/mL [13]. It was shown that the aqueous extracts of Azadirachta indica (A. Juss), Morinda lucida (Benth.), Cymbopogon citratus (DC Stapf.), Mangifera indica (Linn.), and Carica papaya (Linn.) displayed mitodepressive effects on cell division and induced mitotic spindle disturbance in Allium cepa. Senna alata, already known for its toxicity on liver, intestine, and blood components [14], was shown recently as a potent mutagen [15] as it increased (in presence of metabolic activation system, namely S9 mixture and made up of microsome fraction of rats liver) the number of revertant strains of Salmonella typhimrium TA98 and TA1537 in the mutagenicity assay [10], while genotoxicity of Jatropha multifida, Sterculia africana, and Spirostachys Africana were demonstrated by vititox assay in absence of S9 [16]. A synopsis of African medicinal plants screened for their genotoxicity is summarized in Table 9.2. Some African medicinal plants also are known for their protective effects of the genome. These include Acacia salicina (Mimosoideae), Marrubium deserti de Noe´ (Lamiaceae), Myrtus communis (Myrtaceae), Pistacia lentiscus (Anacardiaceae), Pituranthos chloranthus (Apiaceae), Rosa roxburghii (Rosaceae), and Teucrium ramosissimum (Lamiaceae) (Table 9.3).

Table 9.2 African Medicinal Plants Screened for their Genotoxicity Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Antidesma venosum (Euphorbiaceae)

Abdominal pain, enema [17,18].

South Africa

DNA damage: The DCM leaf extract showed positive result in micronucleus test while the twigs extract was positive in micronucleus and Comet tests without S9 (human white blood cells in vitro) [12].

Balanites maughamii (Balanitacea)

Mulluscicidal [17,18].

South Africa

Alkaloids, phenols [19]; saponins, tannins, flavonoids [20]; 3,5,7,30 ,40 pentahydroxyflavanol (epicatechin), 5,7,8,20 ,30 ,4hexahydrodihydroflavanol-4O-β-D-glucoside, 5,7,40 trihydroxyl-isoflavone (genistein) [21]; γ-lactone ((3R,4R,5S)-4-hydroxy-5methyl-3-tetradecanyl γ-lactone), β-sitosterol, triterpenoids (friedelin, lupeol), phytosterols, stigmasterol [22]. Saponins (diogenin, sapogenins), steroidal glycosides [23], steroids, terpenoids, terpenes, flavonoids, organic acids (vanilic acid, syringic acid), furanocoumarins (marmesin, bergapten), hydroxy cholestenones (cryptopgenin) [24].

The DCM and MeOH extracts of leaves and twigs induced DNA damage according to micronucleus and Comet tests without S9 in human lymphocyte cultures [12].

(Continued)

Table 9.2 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Boophane disticha (Amaryllidaceae)

Dressing for cuts, boils, septic wounds, headaches, abdominal pain, weakness, eye conditions, sedative [17,18].

South Africa

Alkaloids (buphanadrine, buphanamine, distichamine [25], 6α-hydroxycrinamine, 6β-hydroxycrinamine) [26].

Catha edulis (Celastraceae)

Antalgic [27].

Ethiopia

Alkaloids (kathine, cathinine, cathidine, D-norpseudoephedrine, cathinone) [28], tannins (tannic acid), mannitol, flavonoids, volatile oils [29].

Catharanthus roseus (Apocyanaceae)

Diabetes, rheumatism [17,18] anti-inflammatory, antimalarial, antimitotic, antihypertensive, antifertility, antihypercholesterolemic, antimutagenic, antidiuretic,

South Africa

DCM-MeOH whole plant extract: alkaloids (vincristine vinblastine, ajmalicine, catharanthine, vindolinine), carbohydrates [32,33], saponins, flavonoids, anthraquinone glycosides [33].

The MeOH extract of bark induced micronuclei while the DCM extract was toxic suggesting an indirect effect on cell division, or other toxic processes according to micronucleus test without S9 in human lymphocyte cultures [12]. The MeOH leaves extract induced a significant increase in sister chromatid exchanges. Moreover, it induced various types of chromosomal aberrations in mice bone marrow cells which include: broken, sticky and ring chromosomes and disturbed metaphase and anaphase [30]. The MeOH leaf extract induced micronuclei in micronucleus and Comet tests without S9 while the DCM extract was found toxic in micronucleus test suggesting an (in)direct effect on cell division, or other

antifungal, antispasmodic, antiviral, cardio tonic, central nervous system depressant, antitumor, cytotoxic, antispermatogenic, anticancer [31]. Catunaregam spinosa (Rubiaceae)

Emetic, fever, aphrodisiac, gonorrhea, headaches, nausea, respiratory and febrile complaints, gynecological ailments, epilepsy, arthritis [17,18].

South Africa

Celtis africana (Ulmaceae)

Cancer, syphilis, rhumatism, pains, magical properties [17,18].

South Africa

Chaetacme aristata (Ulmaceae)

Hemorrhoids [17,18].

South Africa

Triterpenoid, alkaloids, glycosides, flavonoids, saponins, carbohydrates, tannins, phenols [35]. Triterpenoid saponins (Catunaroside, Catunaroside F, Catunaroside H, Mussaendoside J) [36]. Trans-N-coumaroyltyramine, trans-N-feruloyltyramine, trans-N caffeoyltyramine, lauric acid, oleic acid, palmitic acid, lupeol, β-sitosterol, oleanolic acid [37], polyphenols, flavonoids [38]. /

toxic processes in human lymphocyte cultures [12]. The MeOH and DCM leaf extracts induced mutant strains in Ames test with metabolic activation S9 (Salmonella typhimurium strain TA98) [34]. The DCM leaves extract induced micronuclei in micronucleus without exogenous metabolic activation S9 indicating DNA damage (human lymphocyte cultures) [12]. The MeOH root extract induced micronuclei in micronucleus test without S9 in human lymphocyte cultures [12].

DNA damage in micronucleus and Comet tests without S9 in human lymphocyte cultures [12]. The DCM leaves extract showed mutagenicity in Ames test with and without metabolic activation (Salmonella typhimurium strain TA98) [34]. (Continued)

Table 9.2 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Cleome amblyocarpa (Capparidaceae)

Colic, diabetes, pains [39].

Tunisia

Dammarane triterpenes, cleocarpanol [40], cabraleahydroxy lactone [41], stigma-4-en-3-one, lupeol, taraxasterol [42].

Crinum macowanii (Amaryllidaceae)

Scrofula, rheumatic fever, kidney and bladder diseases, fever, sores, glandular swellings [17,18].

South Africa

Lycorine, crinamine [43]; macowine, cherylline, pratorimine [44] buphanidrine [45].

Croton sylvaticus (Euphorbiaceae)

Abdominal and internal inflammations, uterine disorders, tonic, febrile conditions, purgative, pleurisy, indigestion, TB, rheumatism [17,18].

South Africa

/

Single and double strand DNA breaks as well as alkali-labile sites at 01 μg/mL concentrations of the plant extract according to the alkaline Comet assay [39]. The MeOH bark extract induced micronuclei in micronucleus test without metabolic activation S9 in human lymphocyte cultures [12]. The DCM bark extract increased the number of His1 revertants in Ames test (Salmonella typhimurium strain TA98) with and without metabolic activation [34]. The DCM and MeOH (leaves, twigs and bark) extracts induced micronuclei in micronucleus test without exogenous metabolic activation S9 in human lymphocyte cultures [12].

Diospyros whyteana (Ebenaceae)

Dysmenorrhea, irritating rashes, antibacterial [17,18].

South Africa

Isodiospyrin [46].

Erythrina caffra (Fabaceae)

Sores, wounds, arthritis, sprains, aches [17,18].

South Africa

Alkaloids [47], triterpe`nes [48], coumarins, steroids, flavonoids (isoflavones, pterocarpanes, flavanones and isoflavanones) [49].

Euclea divinorum (Ebenacea)

Purgative, for headache, toothache, constipation, antihelmintics, tonics, chest pain, pneumonia, stomach pain [17,18].

South Africa

alkaloids, saponins, diterpenes, tannins, phytosterols [50], DCM extract: triterpenoids, resins tannins, amino acids. DCM/MeOH extract: alkaloids, reducing sugar, saponins, triterpenoids, resins, phenols, tannins, amino acids, diterpenes. Water extract: alkaloids, reducing sugar, saponins, triterpenoids, phenols, flavonoids, amino acids [51].

Mutagenic effect of DCM twigs extract in Comet test and DNA damage with the MeOH extracts in micronucleus and Comet tests without S9 (human lymphocyte cultures) [12]. The DCM leaf extract induced mutant strains in Ames test with metabolic activation in Salmonella typhimurium strain TA98 [34]. The MeOH bark extract induced micronuclei in micronucleus test without exogenous metabolic activation S9 in human lymphocyte cultures [12]. The extract induced micronuclei in micronucleus test without S9 (human lymphocyte cultures) [12].

(Continued)

Table 9.2 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Gardenia volkensii (Rubiacea)

Emetic, sore eyes, headaches, asthma, dysmenorrhea, infertility, epilepsy, convulsions, earache [17,18].

South Africa

Irridoids (4-(2N-gardenamide)nbutanoic acid, genipin, genipin gentiobioside, pterocarpin medicarpin), coumarins, phenylpropanoids, benzenoids, triterpenes [52].

Heteromorpha trifoliata (Apiaceae)

Enemas, abdominal disorders, mental/nervous disorders, intestinal worms, headaches, antiscabies [17,18].

South Africa

Fulcarindiol, sarison [53].

Hypoxis colchicifolia (Hyacinthaceae)

Tonic, anti-HIV, antiinflammatory [17,18].

South Africa

/

Kigelia africana (Bignoniaceae)

Ulcers, sores, syphilis, rheumatism, enema [17,18].

South Africa

Irridoids (Verminoside, Specioside, Minecoside, Norviburtinal); flavonoids

Toxic effect with DCM leaves extract and DNA damage with DCM twigs/bark extracts in micronucleus test without S9. DNA damage of DCM and MeOH twigs/bark extracts in Comet assay (human white blood cells cultures) [12]. The DCM twigs/bark extracts and MeOH leaves extract induced micronuclei in micronucleus test without metabolic activation S9. Mutagenic effect of DCM leaves extract in Comet test without S9 in human lymphocyte cultures [12]. DNA damage of DCM and MeOH bark extracts in Comet and micronucleus tests, respectively (human lymphocyte cultures) [12]. The DCM fruits extract induced micronuclei in micronucleus test without exogenous

Marrubium alysson L (Lamiaceae)

Hypertension, rheumatics, cough, burns, intestinal troubles [54].

Peganum harmala L (Zygophylaceae)

Cancer, hypothermia, hallucinogen factor, antidepressant, antimicrobial, antinociceptive [56]. Headaches, emetics, warts, fractures, scrofula, edema, malaria, skin lesions [17,18].

Plumbago auriculata (Plumbaginaceae)

Tunisia

South Africa

(Luteolin, quercetin); naphtoquinone (Pinnatal, Isopinnatal, Kigelinone, Kigelinol, Isokigelinol, Lapachol); coumarins (Kigelin); phenylpropanoid (Caffeic acid, p-coumaric acid, Ferulic acid); fatty acids (Palmatic acid) [12]. MeOH extract: alkaloids, diterpenoids, saponosids [55]

alkaloids (harmaline, harmine), flavonoids, saponins, reducing compounds, tannins, volatile oils, triterpenes, sterols, anthraquinone [56]. Hexane:ethyl acetate:MeOH: water (40:10:10:2, v/v): naphthoquinones (plumbagin, epi-isoshinanolone), steroids (sitosterol, 3-Oglucosylsitosterol), plumbagic, palmitic acids [58].

metabolic activation S9 indicating DNA damage (human lymphocyte cultures) [12].

The ethyl acetate and MeOH extracts of aerials parts induced DNA damage according to the alkaline Comet assay in C3A cells [54]. The intercalation of P. harmala alkaloids into DNA has led to its mutagenic property which causes genotoxic effects [57]. DCM, MeOH extracts of leaves, fruits, twigs cause dependent DNA damage in micronucleus and Comet assays without S9 (human lymphocyte cultures) [12]. The DCM foliage extract induced mutant strains in Ames test with and without metabolic activation (Salmonella typhimurium tester strain TA98) [34]. (Continued)

Table 9.2 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Prunus Africana (Rosaceae)

Intercostal pain, prostate hypertrophy, hair tonics [17,18].

South Africa

Amygdalin, β-sitosterol [59].

Rhamnus prinoides (Rhamnaceae)

Sprains, blood purifiers, pneumonia, emetics, purgative, colic, stimulants [17,18].

South Africa

Naphthalene glycoside (Geshoidin) [12].

Scilla natalensis (Liliaceae)

Enema, sprains, fractures, purgative, boils, sores, infertility [17,18].

South Africa

/

Sclerocarya birrea (Anacardiaceae)

Diarrhea, dysentery, stomach problems, fever, malaria, tonic, diabetes [17,18].

South Africa

Flavonol glycoside, quercetin 3-O-α-L-(5”-galloyl)arabinofuranoside, phenols, epicatechin derivatives; sesquiterpenes [60].

DNA damage with DCM twigs extract in micronucleus and Comet assays without exogenous metabolic activation (S9). Mutagenic effect of MeOH leaves extract in Comet test (human lymphocyte cultures) [12]. Toxic effect of DCM twigs extract and DNA damage of DCM bark/twigs extracts in micronucleus assay without exogenous metabolic activation (S9). Mutagenic effect of MeOH extracts and DCM leaves extract in Comet test (human lymphocyte cultures) [12]. The DCM and MeOH bark extracts induced micronuclei in micronucleus test without S9 (human lymphocyte cultures) [12]. The MeOH bark induced micronuclei in micronucleus test without exogenous metabolic activation S9 indicating DNA damage (human lymphocyte cultures) [12].

Senecio serratuloides (Asteraceae)

Cuts, swellings, burns, sores, blood purifiers, headaches [17,18].

South Africa

/

Senna alata (Fabaceae)

Constipation, malaria, stomach pains, diabetes, purgative, skin, infectious diseases [15].

Nigeria

Flavonoid glycoside [61], alkaloids, Saponins, tannins, anthracionones, carbohydrates [62].

Spirostachys Africana (Euphorbiaceae)

Wood: stomach ulcers, acute gastritis, eye washes, headaches, rashes, boils, emetic, renal ailment, purgative, blood purifiers, diarrhea, dysentery [17,18].

South Africa

Diterpenes, 2-hydroxyketones, 2-Stachenone, stachenol, diosphenol [63].

The DCM leaves exhibit toxic effect in micronucleus test suggesting an (in)direct effect on cell division, or other toxic processes without exogenous metabolic activation S9 (human lymphocyte cultures) [12]. The extract dose-dependently increased the number of revertants in Salmonella typhymurium TA98 and 1537 strains according to the Ames Test [15]. The DCM and MeOH leaves/ bark/twigs extracts induced micronuclei in micronucleus test without addition of a metabolizing enzyme solution (S9 mix) at 500 and 100 mg/ mL (human lymphocyte cells) [12]. Mutagenic effect of MeOH leaves extract and DCM bark/ twigs extracts in Comet test without S9 (human lymphocyte cultures) [12]. (Continued)

Table 9.2 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Trichelia emetica (Meliaceae)

Stomach and intestinal complaints, dysentery, kidney problems, indigestion, parasites, fever, purgative, bruises, rheumatism [17,18].

South Africa

Steroids, triterpenoids, coumarins [64].

Tulbaghia violaceae (Alliaceae)

Fever, colds, asthma, enemas, tuberculosis, stomach problems, rheumatism, paralysis [17,18]. Emetic, rheumatism, dropsy, heart disease, swollen and painful joints [17,18].

South Africa

2,4-Dithiapentane, p-Xylene, Chloromethylmethyl sulfide, O-Xylene, Thiodiglycol, and p-xylol [65], Alliin [66].

South Africa

Limonoids and limonoid derivatives (turraflorins DI, turraflorins An and B) [67].

Appearance of micronuclei with DCM and MeOH bark extracts in micronucleus assay. DNA damage of MeOH leaves extract in Comet test without S9 (human lymphocyte cultures) [12]. The MeOH leaves/bark extracts induced micronuclei according to micronucleus assay without S9 (human lymphocyte cultures) [12]. Mutagenic effect in Comet test without exogenous metabolic activation (S9). Appearance of micronuclei with DCM leaves/bark extracts and MeOH leaves extract in micronucleus assay without S9 (human lymphocyte cultures) [12].

Turraea floribunda (Meliaceae)

Vernonia colorata (Asteraceae)

Abdominal pain, colic, rheumatism, dysentery, diabetes, ulcerative colitis [17,18].

South Africa

Aqueous and ethanolic extracts: reducing sugars, saponins, polyphenols, tannins, phlobatannins, alkaloids, sterols, Triterpe`nes [68]; coumarins, cardenolids, anthracenosids [69].

Ziziphus mucronata (Rhamnaceae)

Boils, sores, glandular swellings, diarrhea, dysentery, expectorant, emetic for coughs, chest problems, boils, sores, glandular swellings [17,18].

South Africa

Anthocyanins, quinones, tanins [70] Cyclopeptides alkaloids (mucronine), isoquinolone alkaloids (coclaurine, juziphine), porphine alkaloids (laurelliptine, asimilobine), flavonoids (swertish, apigenin glycosides), anthraquinones, triterpenoids (jujubogenin, betulinic acid) [71].

(/): not reported; MeOH: methanol; DCM: dichloromethane.

Toxic effect of DCM leaves/ roots extracts in micronucleus test suggesting an (in)direct effect on cell division, or other toxic processes. DNA damage with MeOH leaves extract in Comet test without exogenous metabolic activation (S9) (human lymphocyte cultures) [12]. Appearance of micronuclei with DCM and MeOH leaf extracts in micronucleus and Comet tests without exogenous metabolic activation (S9). DNA damage of DCM extract in Comet test (human lymphocyte cultures) [12]. The 90% MeOH leaves extract induced mutant strains in Ames test with metabolic activation (Salmonella typhimurium strain TA98) [34].

Table 9.3 African Medicinal Plants Screened for their Genoprotective Effects Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Acacia salicina (Mimosoideae)

Inflammatory diseases, febrifuge, cancer, promote human fertility [72]

Tunisia

Marrubium deserti de Noe´ (Lamiaceae)

Digestive disorders, scorpion stings, allergy [76].

Algeria

Chloroform and petroleum ether The extracts may adsorb the extracts: polyphenols, flavonoids, mutagens in a way similar to the tannins, sterols [73]. carcinogen adsorption which has been associated with pyrrole pigments (hemin and chlorophyllin) according to Salmonella typhimurium TA1535 and TA98 assay (Ames Test) [74,75]. They can induce also DNA glycosylase enzymes which are capable of repairing alkylating DNA bases [72]. The isolated compounds protect Dichloromethane extract: directly DNA strands from the β-stigmasterol 1 [77,78], electrophilic metabolite of the diterpenes, labdane diterpenes mutagen. The protective effect (cyllenin A 2), 15-epi-cyllenin A may correspond to a synergic 3 [79], marrubiin [80,81], participation of several of each marrulactone, marrulibacetal 7 component [76]. [82]. Ethyl acetate and n-BuOH phases from methanol extract: Apigenin7-O-b neohesperidoside, acteoside [83], forsythoside B [84], apigenin-7-O-glucoside, terniflorin (apigenin-7-O-(600-Ep-coumaroyl)-glucoside), apigenin [85], apigenin-7-Oglucuronide [86].

Myrtus communis (Myrtaceae)

Candidiasis, lung disorders [87].

Tunisia

Aqueous, ethyl acetate and methanol extracts: flavonoids, coumarins, tannins [87].

Pistacia lentiscus (Anacardiaceae)

/

Tunisia

Flavonols; galloyl derivatives (galloyl-glucosides, ellagitannins and galloyl-quinic acids) [88].

Pituranthos chloranthus (Apiaceae)

/

Tunisia

Essentials oils (monoterpenoid: thymol and carvacrol; sesquiterpenoid), alkanes, alcohols and aldehydes [91].

The extracts, according to Nifuroxazide and aflatoxine B assays, inhibit microsomal enzyme activation or protect DNA strands from the electrophilic metabolites of the mutagens [87]. An isolated compound (from fruits) Digallic Acid (DGA) is able to interact and neutralize electrophiles such as nitrofurantoine or may inhibit microsomal activation of B[a]p to electrophilic metabolite. It may act by inhibiting microsomal activation or by directly protecting DNA strands from the electrophilic metabolite of the mutagen. They may inhibit also several metabolic intermediates and Reactive Oxygen Species (ROS) formed during the process of microsomal enzyme activation which is capable of breaking DNA strands [89,90]. Essential oils showed a protective effect against damages induced by radicals, obtained from the photolysis of H2O2, on DNA plasmid through free radical scavenging mechanisms [91]. (Continued)

Table 9.3 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Rosa roxburghii (Rosaceae)

Detoxification and restoration of the liver, colon, kidney, lungs and skin, supports healthy cardiovascular function and reduces the risk of heart attacks and stroke [92]. Gastric ulcer, intestinal inflammation, cicatrisant.

South Africa

Ascorbic acid, polyphenols [92].

The fruit extract dose-dependently protected against the metabolically activated carcinogens, 2-AAF and AFB1 in Salmonella typhymurium T98 and TA100 strains, respectively [92].

Tunisia

Tannins, coumarins, sterols, flavonoids [93], sesquiterpene hydrocarbons, oxygenated sesquiterpenes, δ-Cadinene, α-cadinol, germacradien-4-α-1ol [94].

The total oligomer flavonoids (TOF), petroleum ether and aqueous extracts reduced the number of frameshift mutations induced by the direct genotoxicant NOPD (10 μg/plate) and the indirect genotoxicant B [a] P (5 μg/plate) in S. typhymurium TA98 strain, as well as the base pair substitution induced by the direct acting agent SA (1.5 μg/plate) and indirect acting agent AFB1 (0.5 μg/plate) in strains TA100 and TA1535 [93].

Teucrium ramosissimum (Lamiaceae)

Genotoxicity and Teratogenicity of African Medicinal Plants

9.5

253

Medicinal Plants with Teratogenic Effects

Several phytochemicals among which some are commonly used as food and medicinal plants are potential teratogenic agents. The use of laxatives containing anthraquinones should be avoided during pregnancy and especially in the first trimester (the period during which organogenesis occurs and during which malformations can arise from brief exposure), as they can induce uterine contractions [95], increase blood flow to the uterus and its attachments, increase the risk of fetal loss, and may pass into breast milk and cause unwanted effects such as spasms in the infant [96]. Caffeine, considered as the most widely consumed stimulant in the world, could also be the cause of teratogenic effects of plants. It crosses the placental barrier and decreases blood flow to the placenta, where it can be a cause of weight loss in newborns, though there is no consensus on this activity [97,98]. It is estimated that 75% of pregnant women in the United States consume drinks containing caffeine, including those from plants such as black tea (Camellia sinensis), green tea (made from the leaves from Camellia sinensis that have undergone minimal oxidation during processing), and guarana (Paullinia cupana) [97]. Zingiber officinalis (Zingiberaceae) commonly known as ginger, is effective in the treatment of nausea and vomiting during pregnancy [99,100] and the prevention of motion sickness. It is controversial, as there are many differences in relation to its teratogenic potential. It was reported to cause embryonic loss when administered to rats during pregnancy, but also to increase the weight of the remaining fetuses [101]. Ginger components are indicated as potential inducers of apoptosis in human cells [102], a process involved in the remodeling of the fetal brain and other organs. In contrast, Weidner and Sigwart [103] found that an ethanol extract of ginger administered to pregnant rats caused no harm to either mother or developing fetus. Cyanide contained in cassava (Manihot esculenta) could be responsible for its teratogenic effects as this was demonstrated in pregnant rats receiving cassava powder as 80% of their total diet during the first 15 days of gestation. Singh [104] demonstrated that there was a growth retardation of fetuses, embryonic death in 19%, and malformations in 28% of the total implantations, limb defects in 39% and microcephaly with open eyes in 5.5% of cases due to cassava. Garcinia kola (Guttiferae) was also shown to produce duration-dependent teratogenicity in fetal rats at 200 mg/kg body weight (bw) during the first 5 days of gestation, inducing a decrease in the weight and malformations of left upper limb in 7% of the fetuses from pregnant rats. The teratogenesis of Acanthus montanus (Acanthaceae) was demonstrated in another study where the following changes were observed on rat fetuses: decrease in body weight, crown-rump and tail length, placental weight, and poor ossification of bones extremities (forelimbs and hindlimbs) suggesting intrauterine growth retardation after the administration of 500 and 1000 mg/kg bw per day of the leaves methanol/methylene chloride (1:1) extract for 5 days [105]. African medicinal plants screened for their teratogenic potential are reported in Table 9.4.

Table 9.4 African Medicinal Plants Screened for their Teratotoxicity Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Rauwolfia vomitoria (Apocynaceae)

Snake and insect bites and stings, insomnia and insanity [106].

Nigeria

Alkaloids (rauwolfine, reserpine, Ethanolic root bark and leaf rescinnamine, serpentine, extracts induced adverse effects ajmaline), steroid-serposterol and on developing liver of fetal rats saponin [107]. [106]. β-sitosterol from MeOH/CH2Cl2 The MeOH/CH2Cl2 extract had an embryotoxic effect at higher extract of leaves [105]. doses (500 and 1000 mg/(kg day)) when it’s administered orally to Wistar pregnant rats during organogenesis. This effect disappeared within the first 5 days after parturition [105]. Biflavonoid (Kolaviron) The seed extract at 200 mg/kg bw administered alters estrous cycle [13,811], benzophenones, in rats, partly inhibits ovulation Garcinia biflavonones (GB-1, and may produce durationGB-2), kolaflavonone [109]. dependent teratogenicity in fetal Apigenin (flavonoids) [110]. female SpragueDawley rats [108].

Acanthus montanus Cough, epilepsy, (Acanthaceae) dysmenorrhea, pain, miscarriages, rheumatism, hypertension, skin infections [105].

Cameroon

Garcinia kola (Guttiferae)

Nigeria

Cough, purgative, antiparasitic, antimicrobial, bronchitis, throat infections, liver disorders, hypoglycemic, aphrodisiac, antioxidant [108].

Effects of the Plant

Manihot (esculenta), and Manihot dulcis (Euphorbiaceae)

/

Nigeria

Cyanide [104].

Treculia africana (Moraceae)

Malaria, worms, coughs and digestive disorders [111].

Nigeria

Polyphenols [111]. Methanolic extracts: 4hydroxybenzoic acid, caffeic acid, vanillic acid, syringic acid, p-coumaric acid, syringaldehyde, ferulic acid [112].

The milled cassava powder on albino rats at 80% of the diet showed a low incidence of limb defects, open eye, microcephaly, and growth retardation [104]. The extract induced significant numbers of malformations (gross and skeletal) in pregnant female rats treated by polyphenols obtained from outer coat of the fruit [111].

Table 9.5 Teratogenic Plants Reported in Other Parts of the World but also Found in Africa Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Luffa operculata L. Purgative effect, Cogn. treatment of parasitic (Cucurbitaceae) diseases [113].

Brazilia

Ceramides, triterpenoid, steroids [114].

Artemisia absinthium L (Asteraceae)

Brazilia

Thujone (terpenoid) [117], terpenes (limonene, myrcene, α and β thujone [118], sesquiterpene, caryophellene [119], sabinyl acetate, chrysanthenyl acetate [120], sesquiterpene lactone endoperoxide (Artemisinin) [121]. Alkaloid (swansonine) [122,123].

The decoction administered to female mice during the implantation of embryos caused a reduction in birth rate [115]. When ingested in large amounts, it can cause epileptiform seizures and even abortion due to the presence of toxic isolated terpenoid compound thujone [119].

It is used for their antiparasitic effects, and to treat anorexia and indigestion [116].

Oxytropis lambertii / Pursh. (Fabaceae)

India

/

India

Nicotiana glauca Grahm. (Solanaceae)

Alkaloids (piperidine Anatabine, anabaseine) [122,123]. Nicotine [124].

The isolated alkaloid compound swansonine is an alphamannosidase inhibitor which induces vascular resistance and vasoconstriction in the fetus of sheep and cattle. [122,123]. Contains piperidine alkaloids and cause congenital contracture-type skeletal malformations and cleft palates in the fetus. Anatabine and anabaseine are involved in the teratogenic effects on swine [122,123]. Moreover, these induced multiple congenital contractures

Lupinus formosus (Fabaceae)

/

India

Alkaloids (piperidine) [122,123].

(MCC) and palatoschisis in goat kids when their dams were gavaged with the plant during gestation. The skeletal abnormalities included fixed extension of the carpal, tarsal and fetlock joints, scoliosis, lordosis, torticollis and rib cage problems. The clinical signs of toxicity in sheep, cattle, and pigs included, ataxia, incoordination, muscular weakness, prostration, and death [125]. Contains piperidine alkaloids and cause congenital contracture-type skeletal malformations and cleft palates in the fetus [122,123]. Moreover, these induced MCCs and palatoschisis in goat kids when their dams were gavaged with the plant during gestation. The skeletal abnormalities included fixed extension of the carpal, tarsal and fetlock joints, scoliosis, lordosis, torticollis and rib cage problems. The clinical signs of toxicity in sheep, cattle, and pigs included, ataxia, incoordination, muscular weakness, prostration, and death [125]. (Continued)

Table 9.5 (Continued) Plant (Family)

Traditional Use

Treatment of high blood Veratrum pressure and cancer californicum [126]. Durr. (Melanthiaceae) Conium maculatum Antispasmodic, sedative, L. (Umbelliferae) analgesic [123].

Area of Plant Collection

Chemical Constituents

Effects of the Plant

India

Steroidal alkaloids (a cyclopamine, jervine) [123,126,127].

India

Piperidine alkaloids (coniine and gamma-coniceine) [122,123], 2-Pentylpiperidine, (conmaculatin) [128].

Contains steroidal alkaloids that cause craniofacial malformations including cyclopia in the fetus of sheep [123,127]. Contains piperidine alkaloids (coniine and gamma-coniceine) that cause congenital contracture-type skeletal malformations and cleft palates, restricted fetal movement, arthrogrypotic limb deformities in calves (cow) [122,123]. Moreover, these induced MCC and palatoschisis in goat kids when their dams were gavaged with the plant during gestation. The skeletal abnormalities included fixed extension of the carpal, tarsal and fetlock joints, scoliosis, lordosis, torticollis and rib cage problems. The clinical signs of toxicity in sheep, cattle and pigs included, ataxia, incoordination, muscular weakness, prostration and death [125].

Descurainia sophia / L. (Brassicaceae)

India

Astragalus mollisimus Torr. (Fabaceae) Astragalus pubentissimus Torr. (Fabaceae)

/

India

/

India

Hyoscyamine (alkaloid), quercetin3-O-β-D-glucopyranosyl-7-Oβ-gentiobioside, kaempferol-3O-β-D-glucopyranosyl-7-Oβ-gentiobioside, isorhamnetin-3O-β-D-glucopyranosyl-7-Oβ-gentiobioside, quercetin-7-Oβ-gentiobioside, kaempferol-7O-β-gentiobioside, isorhamnetin7-O-β-gentiobioside, quercetin3,7-di-O-β-D-glucopyranoside, kaempferol-3, 7-di-O-β-Dglucopyranoside, isorhamnetin-3, 7-di-O-β-D-glucopyranoside, kaempferol-3-O-β-Dglucopyranosyl-7-O-[(2-O-transsinnapoyl)-β-D-glucopyranosyl (1-- . 6)]-β-D-glucopyranoside), sinapic acid ethyl ester, 3, 4, 5trimethoxyl-cinnamic acid [129]. Coumarins, flavonoids, terpenes, fatty acids, amino acids [130]. Swainsonine alkaloids [131].

Alkaloids (swainsonine, indolizidine), [132,133].

Contains hyoscyamine (alkaloid) that cause deformed foals in the fetus [131].

Contains swainsonine alkaloids that cause neurological defects in the fetus [131]. Contains swainsonine, and indolizidine alkaloid that caused generalized stunting in the fetus of sheep and cattle [132,133]. (Continued)

Table 9.5 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Solanurn tuberosum L. (Solanaceae)

/

India

Solanidanes and spirosolanes

Solanum melongena L. (Solanaceae)

Ani-diabetic, liver complaints [136].

India

Lycopersicon esculentum Mill. (Solanaceae)

Chronic degenerative disease [137].

India

Solanidanes and spirosolanes [132134], alkaloids, saponins, steroids, tannins/ phenolics, flavonoids, proteins, carbohydrates, glycoalkaloids (solasodine), anthocyanin, glycosides (cardiac and cyanogenic glycosides), [136]. Solanidanes and spirosolanes [132134], lycopenes [137].

Contains solanidanes and spirosolanes those cause brain defects and cleft palate in the fetus of sheep and cattle [134,135]. Contains solanidanes and spirosolanes those cause brain defects and cleft palate in the fetus of sheep and cattle [132134].

Voacanga globosa / (Blco.) Merr. (Apocynaceae) / Baccaurea tetrandra (Baill.) Muell.Arg. (Euphorbiaceae) Purgative, emetic effects Ficus septica [140]. Burm.f. (Moraceae)

India

Contains solanidanes and spirosolanes those cause brain defects and cleft palate in the fetus of sheep and cattle [132134]. Alkaloids, saponins, 2-deoxysugars, Causes hypoxia in fetus showed by and hydrolysable tannins [138]. in vitro assay [139].

India

/

Causes hypoxia in fetus showed by in vitro assay [139].

India

Flavonoids, coumarins, alkaloids, steroids, ceramides, triterpe`nes, tannins [141].

Causes hypoxia in fetus showed by in vitro assay [139].

Uncaria perrottetii (A.Rich) Merr. (Rubiaceae) Aglaia loheri (Blco.) (Meliaceae)

Veratrum viride Aiton. (Melanthiaceae)

Caulophyllum thalictroides L. Michx. (Berberidaceae) Cortex cinnamom L. (Lauraceae) Cannabis sativa L. (Cannabaceae)

/

India

Alkaloids, tannins and leucoanthocyanin [142].

Causes hypoxia in fetus showed by in vitro assay [139].

insecticidal, antifungi, anti-inflammatory, neuroprotector, cardioprotector, anticancerous properties [143]. Nervous palpitations, epileptics convulsions, neuralgia, paralysis, whooping-cough, drop, hydropisy [146]. Abortive and contraceptive effects

India

Phenolic ester [144], spinasterol, trilinolein, phytyl fatty acid ester [145], flavaglines [143].

Causes hypoxia in fetus showed by in vitro assay [139].

India

Steroidal alkaloids cyclopamine and jervine [147,148].

The isolated compounds are teratogenic, studied in vitro by ELISA test [147,148].

United States of Alkaloids (N-methylcytisine, America baptifoline, anagyrine, magnoflorine) [149].

/

India

Antimicrobial, antiinflammatory and antiageing balances skin disorder, rheumatism, inflammation, diabetes, excessive epidermal water loss [150].

India

Essentials oil, Cinnamaldehyde [133]. Terpenes (cannabinoids) [151], 9-trans-Tetrahydrocannabivarin, sterols (campesterol, stigmasterol, and β-sitosterol), amino acid (L-proline) [150].

The isolated compound Nmethylcytisine exhibit teratogenicity, studied in vitro rat embryo culture [149]. Cinnamaldehyde is teratogenic in chick embryos [133]. Causes brain defects in the fetus [152].

(Continued)

Table 9.5 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Podophyllum hexandrum L. (Berberidaceae)

/

India

/

Nicotiana tobacum L. (Solanaceae)

India Hair treatment, dysmenorrheal, vermiguge, rheumatism, hoarseness, fungal diseases of the skin, wounds, ulcers, bruises, sores, mouth lesions, stomartitis and mucosa; leaf is orally taken for kidney diseases, bronchitis and pneumonia [125]. India Edema, rheumatism [128].

Causes absent right thumb and radius, a supernumerary left thumb, a probable septal defect of the heart, a defect of the right external ear, and skin tagsin the fetus in humans [153]. The compounds anatabine and anabaseine cause arthrogryposis in the fetus and skeleton defects in pigs and also in swines [122].

Goniothalamus amuyon (Annonaceae)

Anatabine and anabaseine [122], nicotine, anabasine (an alkaloid similar to the nicotine but less active), glucosides (tabacinine, tabacine), 2,3,6-trimethyl-1,4naphthoquinone, 2methylquinone, 2-napthylamine, propionic acid, anatalline, anthalin, anethole, acrolein, anatabine, cembrene, choline, nicotelline, nicotianine, and pyrene [125]. Styrylpyrones ((6R,7R,8R)-8methoxygoniodiol, (6R,7R,8R)8-chlorogoniodiol) [128], goniothalesacetate, goniothalesdiol A, goniodiol-7monoacetate, goniodiol-8monoacetate, leiocarpin C,

It induced morphological abnormalities in the fetus of mice [139].

Espinheira santa Klen. (Maytenus ilicifolia Mart. ex Reissek) (Celastraceae) Alstonia macrophylla Wall. (Apocynaceae) Asparagus racemosus Willd. (Asparagaceae)

Lupinus argenteus Pursh. (Fabaceae)

Wound, cancer [154]

Brazilia

/

India

Gastric ulcers, dyspepsia and as a galactogogue [157].

India

/

India

liriodenine, griffithazanone A, 4-methyl-2,9,10-(2H)-1azaanthracencetrione, velutinam, aristolactam BII, isoquinolones [128]. Maytansine [154].

Bisindole alkaloid (macralstonine, thungfaine), secoiridoid glycoside (sweroside, naresuanoside) [155]. Polycyclic alkaloid (Asparagamine A), steroidal saponins (shatavaroside A, shatavaroside B, shatavarins VI-X), saponin (filiasparoside C), shatavarin I, shatavarin IV (or asparinin B), shatavarin V, immunoside and schidigerasaponin D5 (or asparanin A), [158,159]. Isoflavone, 8-methoxy-5,6,4’trihydroxyisoflavone 7-O-β-Dglucopyranoside [160]. Anagyrine, quinolizidine [161].

The isolated compound maytansine shows teratogenic activity in mice [154].

It induced morphological abnormalities in the fetus of mice [156].

Cause reabsorption of fetuses and intrauterine growth retardation in rats [156].

The isolated compound anagyrine shows teratogenic effect in calf [161]. (Continued)

Table 9.5 (Continued) Plant (Family)

Traditional Use

Area of Plant Collection

Chemical Constituents

Effects of the Plant

Lupinus caudatus L. (Fabaceae)

/

India

Anagyrine [161].

Lupinus nootkatensis Donn ex sims (Fabaceae) Thermopsis montana Nutt. (Fabaceae) Trigonella foenumgraecum (Fabaceae)

/

India

Anagyrine [161].

The isolated compound anagyrine shows teratogenic effect in calf [161]. The isolated compound anagyrine shows teratogenic effect in calf [161].

/

India

Anagyrine [162].

It’s used to ease childbirth and to increase milk flow, for menstrual pain and as hilba tea to ease stomach problems [163].

India

Alkaloids: Trimethylamine, Neurin, Trigonelline, Choline, Gentianine, Carpaine and Bin. Amino acids: Isoleucine, 4-Hydroxyisoleucine, Histidine, Leucine, lysine, L-tryptophan, Argenine. Saponins: Graecunins, fenugrin B, fenugreekine, trigofoenosides A-G. Steroidal sapinogens: Yamogenin, diosgenin, smilagenin, sarsasapogenin, tigogenin, neotigogenin, gitogenin, neogitogenin, yuccagenin, saponaretin. Flavonoids:

The isolated compound anagyrine shows teratogenic effect in calf [162]. Cause decrease bone marrow cell proliferation and increase fetal mortality rate in rats [164].

Quercetin,rutin,vetixin isovetixin. Fibers: Gum, neutral detergent. Other: Coumarin, lipids, vitamins, minerals [163]. Trifolium hybridum / L. (Fabaceae) Indigofera spicata / Forssk. (Fabaceae)

Sorghum bicolori L. (Poaceae) Datura stramonium Linn. (Solanaceae) Boerhaavia diffusa Linn. (Nyctaginaceae) Rauwolfia serpentina Benth. Ex Kurz. (Apocynaceae) Aspilia africana (Pers.) C.D. Adams (Asteraceae)

India

/

India

Indospicine, (1)-5vdeacetylpurpurin, (1)-5methoxypurpurin, (2S)-2,3dihydrotephroglabrin, (2S)-2,3dihydrotephroapollin C, flavanones, rotenoids, chalcone [165]. cyanogenic glycosides

/

India

Antispasmodiques, sedatifs, asthma, neuralgia [131]. Dyspepsia, abdominal pain, inflammation, cancer, diabetes [166]. Cancer, liver disease, or mental illness [169].

India

Alkaloid (hyoscyamine, scopolamine) [131].

India

alkaloids (punarnavine), rotenoids (boeravinones A to J), flavones [167]. yohimbine, reserpine, ajmaline, deserpidine, rescinnamine, serpentinine [169].

Contraceptive and antifertility properties, antimalarial, rheumatic pain, gonorrhea [171].

MCCs, multiple congenital contractures.

India

India

Alkoloids, tannins, saponins, flavonoids, phenols [171].

Contains mycotoxin which is teratogenic [131]. Contains indospicine which is teratogenic [131].

Contains cyanogenic glycosides which are teratogenic [131]. Alkaloid hyoscyamine is responsible for teratogenicity. [131]. It induced congenital malformation in albino rats [168]. Teratogenicity in Wistar rats [170].

Teratogenicity in Wistar rats. [170].

266

Toxicological Survey of African Medicinal Plants

The teratogenicity of some plants found in Africa but collected in other parts of the world was also reported. Some of them include Goniothalamus amuyon (Annonaceae), Rauwolfia serpentina Benth. Ex Kurz. Voacanga globosa (Blco.) Merr., and Alstonia macrophylla Wall. (Apocynaceae), Aspilia africana (Pers.) C. D. Adams, and Artemisia absinthium L (Asteraceae), Asparagus racemosus Willd. (Asparagaceae), Podophyllum hexandrum L., and Caulophyllum thalictroides L. Michx. (Berberidaceae), Descurainia sophia L. (Brassicaceae), Cannabis sativa L. (Cannabaceae), Espinheira santa Klen. (Maytenus ilicifolia Mart. ex Reissek) (Celastraceae), Luffa operculata L. Cogn. (Cucurbitaceae), Baccaurea tetrandra (Baill.) Muell.Arg. (Euphorbiaceae), Oxytropis lambertii Pursh., Lupinus formosus, Astragalus mollisimus Torr. and Lupinus argenteus Pursh., and Lupinus caudatus L., Lupinus nootkatensis Donn ex sims, Thermopsis montana Nutt. (Fabaceae), Trigonella foenum-graecum, Trifolium hybridum L., Indigofera spicata Forssk. and Astragalus pubentissimus Torr. (Fabaceae), Cortex cinnamom L. (Lauraceae), Veratrum californicum Durr., and Veratrum viride Aiton. (Melanthiaceae), Aglaia loheri (Blco.) (Meliaceae), Ficus septica Burm.f. (Moraceae), Boerhaavia diffusa Linn. (Nyctaginaceae), Sorghum bicolori L. (Poaceae), Nicotiana glauca Grahm., Solanurn tuberosum L., Solanum melongena L., Lycopersicon esculentum Mill., Nicotiana tobacum, and Datura stramonium Linn. (Solanaceae), Conium maculatum L. (Umbelliferae), and Uncaria perrottetii (A.Rich) Merr. (Rubiaceae) (Table 9.5).

9.6

Conclusion

In this chapter, we reviewed African medicinal plants with ability to induce genotoxic or teratogenic effects. We also pointed out some of the plants available in the continent with such effects even though they were collected in other parts of the world, in order to warn African therapists of their potential harmful effects. Though most of the results relied on animal or bacterial systems, it is important to have this information in order to prevent as well as possible some cancers or congenital abnormalities by using or developing new drugs which can safely be used by pregnant women, as well as other patients.

References [1] Cosyns JP, Jadoul M, Squifflet JP, De Plaen JF, Ferluga D, van Ypersele de Strihou C. Chinese herbs nephropathy, a clue to Balkan endemic nephropathy? Kidney Int 1994;45:16808. [2] Lord GM, Tagore R, Cook T, Gower P, Pusey CD. Nephropathy caused by Chinese herbs in the UK. Lancet 1999;354(9177):4812. [3] Luyckx VA, Naicker S. Acute kidney injury associated with the use of traditional medicines. Nat Clin Pract Nephrol 2008;4:66471. [4] Stengel B, Jones E. End-stage renal insufficiency associated with Chinese herbal consumption in France. Nephrologie 1998;19:1520.

Genotoxicity and Teratogenicity of African Medicinal Plants

267

[5] Tanaka A, Nishida R, Sawai K, Nagae T, Shinkai S, Ishikawa M, et al. Traditional remedy-induced Chinese herbs nephropathy showing rapid deterioration of renal function. Nihon Jinzo Gakkai Shi 1997;39:7947. [6] Tanaka A, Shinkai S, Kasuno K, Maeda K, Murata M, Seta K, et al. Chinese herbs nephropathy in the Kansai area, a warning report. Nihon Jinzo Gakkai Shi 1997; 39:43840. [7] ,http://www.alttox.org/ttrc/existing-alternatives/genotoxicity.htmL.; 2011. Genotoxicity, validated non-animal alternatives. AltTox.org. 2011-06-20. [accessed on 12.12.13]. [8] Furman G. Genotoxicity testing for pharmaceuticals current and emerging practices, ,http://www.pharmatek.com/pdf/PTEKU/Apr172008.pdf.; 2008 [accessed on 16.12.13]. [9] Ames BN, Durston WE, Yamasaki E, Lee FD. Proc Natl Acad Sci 1973;70(8): 22815. [10] Tice RR, Agurell E, Anderson D, Burlinson B, Hartmann A, Kobayashi H, et al. Single cell gel/Comet assay, guidelines for in vitro and in vivo genetic toxicology testing. Environ Mol Mutagen 2000;35(3):20621. [11] Wilson JG. Environment and birth defects. Environmental science series. London: Academic Press; 1973. [12] Taylor JLS, Elgorashi EE, Maes A, Gorp UV, Kimpe ND, Staden JV, et al. Investigating the safety of plants used in south African traditional medicine, testing for genotoxicity in the micronucleus and alkaline Comet assays. Environ Mol Mutagen 2003;42:14454. [13] Fatemeh K, Khosro P. Cytotoxic and genotoxic effects of aqueous root extract of Arctium lappa on Allium cepa Linn root tip cells. Intl J Agron Plant Prod 2012; 3(12):6307. [14] Hennebelle T, Weniger B, Joseph H, Sahpaz S, Bailleuil F. Senna alata. Fitoterapia 2009;80:38593. [15] Hong C-E, Lyu S-Y. Genotoxicity detection of five medicinal plants in Nigeria. J Toxicol Sci 2011;36(1):8793. [16] van den Bout-van den Beukel CJ, Hamza OJ, Moshi MJ, Matee MI, Mikx F, Burger DM, et al. Evaluation of cytotoxic, genotoxic and CYP450 enzymatic competition effects of Tanzanian plant extracts traditionally used for treatment of fungal infections. Basic Clin Pharmacol Toxicol 2008;102(6):51526. [17] Hutchings A, Scott AH, Lewis G, Cunningham AB. Zulu medicinal plants, an inventory. Natal, South Africa: University of Natal Press; 1996. [18] Van Wyk B-E, van Oudtshoorn B, Gericke N. Medicinal plants of South Africa. Pretoria, South Africa: Briza; 1997. [19] Fawole OA. Pharmacology and phytochemistry of South African traditional medicinal plants used as antimicrobials. Master of science in the research centre for plant growth and development school of biological and conservation sciences. Pietermaritzburg: University of KwaZulu-Natal; 2009. p. 135. [20] Tor-Anyiin TA, Terseer YD. Phytochemical screening and antimicrobial activity of stem bark extracts of Antidesma Venosum. J Nat Prod Plant Res 2012;2(3):42730. [21] Gitu LM. Biological and phytochemical studies of medicinal plants, Antidesma venosum (Euphorbiaceae) and Kotschya africana (Fabaceae) used in traditional medicine in Kenya. JKUAT Abstracts of PostGraduate Thesis; 2009. [22] Magadula JJ, Mwangomo DT, Moshi MJ, Heydenreich M. A novel γ-lactone and other constituents of a Tanzanian Antidesma venosum. Spatula DD Peer Rev J Complementary Med Drug Dis 2013;3(1):712.

268

Toxicological Survey of African Medicinal Plants

[23] Yadov JP, Panghal M. Balanites aegyptiaca (L.) Del. (Hingot), a review of its traditional uses, phytochemistry and pharmacological properties. Int J Green Pharm 2010; 4(3):1406. [24] van Wyck B-E, Van Oudtshoorn MCB, Gericke N. Medicinal plants of South Africa. Pretoria: Briza Publications; 2009. p. 320. [25] Sandager M, Nielsen ND, Stafford GI, van Staden J, Jager AK. Alkaloids from Boophane disticha with affinity to the serotonin transporter in rat brain. J Ethnopharmacol 2005;98:36770. [26] Adewusi EA, Fouche G, Steenkampa V. Cytotoxicity and acetylcholinesterase inhibitory activity of an isolated crinine alkaloid from Boophane disticha (Amaryllidaceae). Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa; 2012. p. 130. [27] Hautefeuille M, Ve´le´a D. Les drogues de synthe`se. Paris: Presses Universitaires de France, coll; 2002. [28] Al-Ahdal MN, McGarry TJ, Hannan M. Cytotoxicity of Khat (Catha edulis) extract on cultured mammalian cells. Mutat Res 1988;204(3):17322. [29] Krikorian AD, Getahun A. Chat, Coffee’s Rival from Harar, Ethiopia. II. Chemical composition. Econ Bot 1973;27:37889. [30] Abderrahman SM, Modallal N. Genotoxic effects of Catha edulis (Khat) Extract on Mice Bone Marrow cells. JJBS 2008;1(4):16572. [31] Huxley A. New RHS dictionary of gardening. London: Macmillan; 1992. ISBN 0-33347494-5. [32] Ibrahim M, Mehjabeen SS, Mangamoori LN. Pharmacological evaluation of Catharanthus roseus. Int J Pharm Appl 2011;2(3):16573. [33] Yadav PD, Bharadwaj NSP, Yedukondalu M, Methushala CH, Ravi KA. Pytochemical evaluation of Nyctanthes arbortristis, Nerium oleander and Catharathnus roseus. IJRPB 2013;1(3):3338. [34] Elgorashi EE, Taylor JLS, Maes A, Van Staden J, De Kimpe N, Verschaeve L. Screening of medicinal plants used in South African traditional medicine for genotoxic effects. Toxicol Lett 2003;143:195207. [35] Senthamarai R, Kirubha SV, Gayathri S. Pharmacognostical and Phytochemical studies on fruits of Catunaregam spinosa Linn. J Chem Pharm Res 2011;3(6):82938. [36] Guang-Chun G, Zhong-Xian L, Shu-Hong T, Si Z, Fa-Zuo W, Qing-Xin L. Triterpenoid saponins from the stem bark of Catunaregam spinosa. Can J Chem 2011;89(10):127782. [37] Al-Taweel AM, Shagufta P, Azza ME-S, Ghada AF, Abdul M, Nighat A, et al. Bioactive phenolic amides from Celtis africana. Molecules 2012;17:267582. [38] Adedapo AA, Jimoh FO, Afolayan AJ, Masika PJ. Antioxidant properties of the methanol extracts of the leaves and stems of Celtis africana. RNP 2009;3(1):2331. [39] Edziri H, Mastouri M, Aouni M, Anthonissen R, Verschaeve L. Investigation on the genotoxicity of extracts from Cleome amblyocarpa Barr. and Murb, an important Tunisian medicinal plant. S Afr J Bot 2013;84:1023. [40] Tsichritzis F, Abdel-Mogip M, Jakupovic J. Dammarane triterpenes from Cleome africana. Phytochemistry 1993;33:4235. [41] Cascon SC, Brown KS. Biogenetically significant triterpenes in a species of meliaceae, Cabralea polytricha A. Juss. Tetrahedron 1972;28:31523. [42] Jente R, Jakupovic J, Olatunji GA. A Cembranoid diterpene from Cleome viscosa. Phytochemistry 1990;29:6667.

Genotoxicity and Teratogenicity of African Medicinal Plants

269

[43] Davey MW, Persiau G, De Bruyn A, van Damme J, Bauw G, Van Montau M. Purification of the alkaloid lycorine and simultaneous analysis of ascorbic acid and lycorine by micellar electrokinetic capillary chromatography. Anal Biochem 1998;257:808. [44] Waller GR, Nowachi EK. Alkaloids biology and metabolism in plants. Warsaw, Poland: Polish Academy of Sciences; 1978. p. 3467. [45] Neuwinger HD. African ethnobotany poisons and drugs. Germany: Chapman & Hall; 1996. p. 106. [46] Mallavadhani UV, Panda AK, Rao YR. Review article number 134 pharmacology and chemotaxonomy of Diospyros. Phytochemistry 1998;49(4):90151. [47] Ghosal S, Singh S, Bhattacharya SK. Alkaloids of Mucuna pruriens, chemistry and pharmacology. Planta Med 1971;19:279. [48] Nkengfack AE, Vouffo W, Vardamides JC, Kouam J, Fomum ZT, Meyer M, et al. Phenolic metabolites from Erythrina species. Phytochemistry 1997;46(3):5738. [49] Chacha M, Bojase-Moleta G, Majinda RRT. Antimicrobial and radical scavenging flavonoids from the stem wood of Erythrina latissima. Phytochemistry 2005;66:99104. [50] Mbanga J, Ncube M, Magumura A. Antimicrobial activity of Euclea undulata, Euclea divinorum and Diospyros lycioides extracts on multi-drug resistant Streptococcus mutans. J Med Plants Res 2013;7(37):27416. [51] Ngari FW, Gikonyo NK, Wanjau RN, Njagi EM. Safety and Antimicrobial properties of Euclea divinorum Hiern, ChewiSticks used for management of oral health in Nairobi County, Kenya. AJBPS 2013;3(3):18. [52] Juma BF, Majinda RRT. Constituents of Gardenia volkensii, their brine shrimp lethality and DPPH radical scavenging properties. Nat Prod Res 2007;21(2):1215. [53] Villegas M, Vargas D, Msonthi JD, Marston A, Hostettmann K. Isolation of the antifungal compounds falcarindiol and sarisan from Heteromorpha trifoliata. Planta Med 1988;54:367. [54] Edziri H, Mastouri M, Mahjoub A, Anthonissen R, Mertens B, Cammaerts S, et al. Toxic and mutagenic properties of extracts from Tunisian traditional medicinal plants investigated by the neutral red uptake, VITOTOX and alkaline Comet assays. S Afr J Bot 2011;77:70310. [55] Edziri H, Samia A, Groh P, Mahjoub MA, Mastouri M, Gutmann L, et al. Antimicrobial and cytotoxic activity of Marrubium alysson and Retama raetam grown in Tunisia. Pakistan J Biol Sci 2007;10:175962. [56] Benbott A, Bahri L, Boubendir A, Yahia A. Study of the chemical components of Peganum harmala and evaluation of acute toxicity of alkaloids extracted in the Wistar albino mice. J Matern Environ Sci 2013;4(4):55865. [57] Moloudizargari M, Mikaili P, Aghajanshakeri S, Asghari MH, Shayegh J. Pharmacological and therapeutic effects of Peganum harmala and its main alkaloids. Pharmacogn Rev 2013;7(14):19919212. [58] De Paiva SR, Figueiredo MR, Kaplan MAC. Isolation of secondary metabolites from roots of Plumbago auriculata Lam. by countercurrent chromatography. Phytochem Anal 2005;16(4):27881. [59] Van Wyk BE, Gericke N. People’s plants. A guide to useful plants of Southern Africa. Pretoria: Briza Publications; 2000. p. 351. [60] Kpoviessi DSS, Gbaguidi FA, Kossouoh C, Agbani P, Yayi-Ladekan E, Sinsin B, et al. Chemical composition and seasonal variation of essential oil of Sclerocarya birrea (A. Rich.) Hochst subsp birrea leaves from Benin. J Med Plants Res 2011;5(18):46406.

270

Toxicological Survey of African Medicinal Plants

[61] Aowoyale JA, Olatunji GA, Oguntoye SO. Antifungal and antibacterial activities of an alcoholic extract of Senna alata leaves. J Appl Sci Environ Manage 2005;9(3):1057. [62] Okonko IO, Sule WF, Joseph TA, Ojezele MO, Nwanze JC, Alli JA, et al. In vitro antifungal activity of Senna Alata Linn. Crude leaf extract. Adv Appl Sci Res 2010;1 (2):1426. [63] Baarschers WH, Horn DH, Johnson LRF. The structure of some diterpenes from tambooti wood, Spirostachys africana Sond. J Chem Soc 1962;404655. [64] Traore M, Zhai L, Chen M, Olsen CE, Odile N, Pierre GI. Cytotoxic kurubasch aldehyde from Trichilia emetica. Nat Prod Res 2007;21:137. [65] Soyingbe OS The chemical composition, antimicrobial and antioxidant properties of the essential oils of Tulbaghia violacea Harv. and Eucalyptus grandis W. Hill ex Maiden. Thesis of Master of Science in the Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, South Africa; 2012. p. 108. [66] Van Wyk B-E, Van Oudtshoorn B, Gericke N. Medicinal plants of South Africa. Pretoria, South Africa: Briza; 1997. pp. 304. [67] McFarland K, Mulholland DA, Fraser L-A. Limonoids from Turraea floribunda (Meliaceae). Phytochemistry 2004;65(14):20317. [68] Adebayo OL, Asamoah B, Mills-Robertson E, Charles F. In vitro evaluation of aqueous and ethanolic extracts of Vernonia colorata as an antibacterial agent. Int J Curr Res Rev 2012;04(01):217. [69] Guenne´ S, Hilou A, Ouattara N, Nacoulma OG. Anti-bacterial activity and phytochemical composition of extracts of three medicinal Asteraceae species from Burkina Faso. Asian J Pharm Clin Res 2012;5(2):09742441. [70] Karola DO The ethnobotany and chemistry of South African traditional tonic plants. Thesis of Doctorate in Botany, University of Johannesburg; 2012. p. 481. [71] Dictionary of natural products. London: Chapman & Hall; 1996. [72] Steele VE, Kellov GJ. Development of cancer chemopreventive drugs based on mechanistic approaches. Mutat Res 2005;591:1623. [73] Chatti IB, Jihed B, Skandrani I, Bhouri W, Ghedira K, Ghedira LC. Antioxidant and antigenotoxic activities in Acacia salicina extracts and its protective role against DNA strand scission induced by hydroxyl radical. Food Chem Toxicol 2011;49:17538. [74] Ferguson RL, Philpott M, Karunasinghe N. Dietary cancer and prevention using antimutagens. Toxicology 2004;198:14759. [75] Ikuma NEM, Passoni HM, Biso IF, Longo M. Investigation of genotoxic and antigenotoxic activities of Melampodium divaricatum in Salmonella typhimurium. Toxicol In Vitro 2006;20:3616. [76] Zaabat N, Hay A-E, Michalet S, Darbour N, Bayet C, Skandrani I, et al. Antioxidant and antigenotoxic properties of compounds isolated from Marrubium deserti de Noe´. Food Chem Toxicol 2011;49:332835. [77] Marsan MP, Warnock W, Muller I, Nakatani Y, Ourisson G, Milon A. Synthesis of deuterium-labeled plant sterols and analysis of their side-chain mobility by solid state deuterium NMR. J Org Chem 1996;61:42527. [78] Pereira PS, Franca SC, Anderson O, Paulo VB, Camila MS, Pereira SIV, et al. Chemical constituents from Tabernaemontana catharinensis root bark, a brief NMR review of indole alkaloids and in vitro cytotoxicity. Quim Nova 2008;31:204. [79] Karioti A, Heilmann J, Skaltsa H. Labdane diterpenes from Marrubium velutinum and Marrubium cylleneum. Phytochemistry 2005;66:10606.

Genotoxicity and Teratogenicity of African Medicinal Plants

271

[80] Kno¨ss W, Reuter B, Zapp J. Biosynthesis of the labdane diterpene marrubiin in Marrubium vulgare via a non-mevalonate pathway. Biochem J 1997;326:44954. [81] Hussein AA, Meyer MJJ, Rodriguez B. Complete H and C NMR assignments of three labdane diterpenoids isolated from Leonotis ocymifolia and six other related compounds. Magn Reson Chem 2003;41:147. [82] Rigano D, Aviello G, Bruno M, Formisano C, Rosselli S, Capasso R, et al. Antispasmodic effects and structure-activity relationships of labdane diterpenoids from Marrubium globosum ssp. Libanoticum. J Nat Prod 2009;72:147781. [83] Li L, Tsao R, Liu Z, Liu S, Yang R, Young JC, et al. Isolation and purification of acteoside and isoacteoside from Plantago psyllium L. by high-speed counter-current chromatography. J Chromatogr A 2005;1063:1619. [84] Endo K, Takahashi K, Abe T, Hikino H. Structure of forsythoside B, an antibacterial principle of Forsythia koreana stems. Heterocycles 1982;19:2614. [85] Van LP, De Bruyne T. Reinvestigation of the structural assignment of signals in the 1 H and 13 C NMR spectra of the flavone apigenin. Magn Reson Chem 1986;24:87982. [86] Agrawal PK. Carbon-13 NMR of Flavonoids. Studies in organic chemistry, Carbon-13 NMR of flavonoids. Amsterdam: Elsevier; 1989. [Magn Reson Chem 28(6):56263]. [87] Hayder NA, Abdelwahed S, Kilani R, Ben Ammar A, Mahmoud K, Ghedira L, et al. Anti-genotoxic and free-radical scavenging activities of extracts from (Tunisian) Myrtus communis. Mutat Res 2004;564:8995. [88] Bhouri W, Derbel S, Skandrani I, Boubaker J, Bouhlel I, Mohamed B, et al. Study of genotoxic, antigenotoxic and antioxidant activities of the digallic acid isolated from Pistacia lentiscus fruits. Toxicol In Vitro 2010;24:50915. [89] Shon MY, Choi SD, Kahng GG, Nam SH, Sung NJ. Antimutagenic antioxidant free radical scavenging activity of ethyl acetate extracts from white yellow and red onions. Food Chem Toxicol 2004;42:65966. [90] De flora S. Mechanisms of inhibitors of mutagenesis and carcinogenesis. Mutat Res 1998;402:1518. [91] Neffati I, Bouhlel M, Sghaiera B, Boubakera J, Limema I, Kilani S, et al. Antigenotoxic and antioxidant activities of Pituranthos chloranthus essential oils. Environ Toxicol Phar 2009;27:18794. [92] Westhuizen FH, Van D, Catharina S, van Rensburg J, George S, Rautenbach JL, et al. In vitro antioxidant, antimutagenic and genoprotective activity of Rosa roxburghii fruit extract. Phytother Res 2008;22:37683. [93] Sghaier MB, Bhouri W, Bouhlel I, Skandrani I, Boubaker J, Chekir-Ghedira L, et al. Inhibitory effect of Teucrium ramosissimum extracts on aflatoxin B1, benzo[a]pyrene, 4-nitro-o-phenylenediamine and sodium azide induced mutagenicity, Correlation with antioxidant activity. S Afr J Bot 2011;77:73040. [94] Hachicha SF, Skanji T, Barrek S, Ghrabi ZG, Zarrouk H. Composition of the essential oil of Teucrium ramosissimum Desf. (Lamiaceae) from Tunisia. Flavour Frag J 2007;22(2):1014. [95] Conover EA. Herbal agents and over-the-counter medications in pregnancy. Best Pract Res Clin En 2003;17(2):23751. [96] Shulz V, Ha¨nsel R, Tyler VE. Fitoterapia Racional, um guia de fitoterapia para as Ciˆencias da Sau´de. Barueri, Ed. Manole, 2002. p. 386. [97] Bicalho GC, Filho AAB. Peso ao nascer e influˆencia do consumo de cafeı´na. Rev Sau´de Pu´bl 2002;36(2):1807.

272

Toxicological Survey of African Medicinal Plants

[98] Moreira LMA, Dias AL, Ribeiro HBS, Falca˜o CL, Felı´cio TD, Stringuetti C, et al. Associac¸a˜o entre o uso de abortificantes e defeitos congˆenitos. Rev Bras Ginecol Obstet 2001;23(8):51721. [99] Marcus DM, Snodgrass WR. Do no harm, avoidance of herbal medicine during pregnancy. Obstet Gynecol 2005;105(5):111922. [100] Vutyavanich T, Kraisarin T, Ruangsri R. Ginger for nausea and vomiting in pregnancy, randomized, double-masked, placebo-controlled trial. Obstet Gynecol 2001; 97(4):57782. [101] Wilkinson JM. Effect of ginger tea on the fetal development of Sprague-Dawley rats. Reprod Toxicol 2000;14(6):50712. [102] Myioshi N, Yoshimasa N, Yasuhiro U, Masako A, Yoshio O, Koji U, et al. Dietary ginger constituents, galanals A and B, are potent apoptosis inducers in human T lymphoma Jurkat cells. Cancer Lett 2003;199(2):1139. [103] Weidner MS, Sigwart K. Investigation of the teratogenic potential of a Zingiber officinale extract in the rat. Reprod Toxicol 2000;15(1):7580. [104] Singh JD. The teratogenic effects of dietary Cassava on the pregnant albino rat: a preliminary report. Teratology 1981;24:2892917. [105] Nana P, Asongalem EA, Foyet HS, Folefoc GN, Dimo T, Kamtchouing P. Maternal and developmental toxicity evaluation of Acanthus montanus leaves extract administered orally to Wistar pregnant rats during organogenesis. J Ethnopharmacol 2008;116:22833. [106] Eluwa MA, Ekanem TB, Udoh PB, Akpantah AO, Ekong MB, Asuquo OR, et al. Teratogenic effect of crude ethanolic root bark and leaf extracts of Rauwolfia vomitoria (Apocynaceae) on the liver of albino Wistar rat fetuses. Asian J Med Sci 2013; 4(1):304. [107] Gill LS. Ethnomedical uses of plants in Nigeria. Benin: UNIBEN Press; 1992. p. 204. [108] Akpantah AO, Oremosu AA, Noronha CC, Ekanem TB, Okanlawon AO. Effects of Garcinia kola seed extract on ovulation, oestrous cycle and foetal development in cyclic female Sprague-Dawley rats. Niger J Physiol Sci 2005;20(12):5862. [109] Cotterih P, Scheinmenn F, Stenhuise I. Composition of G. kola seeds. J Chem Soc 1978;1:5323. [110] Iwu MM, Igboko O. Flavonoids of Garcinia kola seeds. J Nat Prod 1982;45:6501. [111] Lawal RO. Effects of dietary protein on teratogenicity of polyphenols obtained from the outer coat of the fruit of Treculia Africana. Food Chem 1997;60(4):4959. [112] Smith JL, Stanley DW. Toughening in blanched asparagus, identification of phenolic compounds. Food Chem 1989;1314:27187. [113] Sanseverino MTV, Spritzer DT, Schuler-Faccini L. Manual de Teratogˆenese. Porto Alegre, Ed. da UFRGS; 2001. p. 42350. [114] Feitosa CRD-S, Da Silva RC, Braz-Filho R, De Menezes JESA, Siqueira SMC, Monte FJQ. Characterization of chemical constituents of Luffa operculata (Cucurbitaceae). Am J Anal Chem 2011;2:98995. [115] Barilli SLS, Santos ST, Montanari T. Efeito do decocto dos frutos de buchinha-donorte (Luffa operculata Cogn.) sobre a reproduc¸a˜o feminina e o desenvolvimento embriona´rio e fetal. In, sala˜o de iniciac¸a˜o cientı´fica (17, 2005, Porto Alegre). Livro de resumos. Porto Alegre, UFRGS; 2005. p. 539. [116] Lachenmeier DW. Wormwood (Artemisia absinthium L.), a curious plant with both neurotoxic and neuroprotective properties. J Ethnopharmacol 2010;131:2247. [117] Robbers JE, Tyler VE. Tyler’s Herbs of choice, the therapeutic use of phytomedicinals. New York, NY: Haworth Herbal; 1999. p. 287.

Genotoxicity and Teratogenicity of African Medicinal Plants

273

[118] Vostrowsky O, Brosche T, Ihm H, Zintl R, Knobloch K. The essential components from Artemisia absinthium. Z Naturforsch 1981;36:36977. [119] Tucker AO, Maciarello MJ. The essential oil of Artemisia “Powis Castle” and its putative parents, A. absinthium and A. arborescens. J Essent Oil Res 1993;5: 22439. [120] Chilava FP, Lidlle AP, Doglia G. Chemotaxonomy of wormwood (Artemisia absinthium L.). A. Lebensm Unters Forech 1983;176:3636. [121] Klayman DL, Lin AJ, Acton N, Scovill N, Hock JM, Milhous WK, et al. Isolation of artemisinin (qinghaosu) from Artemisia annua growing in the United States. J Nat Prod 1984;47:7157. [122] Bush LP, Crowe MW. Nicotiana alkaloids. Toxicants of plant origin. Alkaloids, vol. I. Boca Raton, FL: CRC Press Inc.; 1989. 335, p. 87107. [123] Schmidt SP, Forsythe WB, Cowgill HM, Myers RK. A case of congenital occipito atlantoaxial malformation (OAAM) in a lamb. J Vet Dia Inv 1993;5:45862. [124] MacMahon JA. (1997). Deserts, New York, NY, National Audubon Society Nature Guides, Knopf A.A. Inc, 9e e´d. [125] Panter KE, Keeler RF, Bunch TD, Callan RJ. Congenital skeletal malformations and cleft palate induced in goats by ingestion of Lupinus, Conium and Nicotiana species, USDA/ARS/Poisonous Plant Research Laboratory, Logan, UT. Toxicology 1990;28 (12):137785. [126] Beachy PA, Chen JK, Cooper MK, Wang B, Mann RK, Milenkovic L, et al. Effects of oncogenic mutations in smoothened and patched can be reversed by cyclopamine. Nature 2000;406(6799):10059. [127] Kimberling CY. Jensen and Swift’s diseases of sheep. 3rd ed. Philadelphia/Paris: Lea and Febiger; 1988. p. 37273. [128] Lan Y-H, Chang F-R, Yang Y-L, Wu Y-C. New constituents from stems of Goniothalamus amuyon. Chem Pharm Bull 2006;54(7):10403.Changwichit K, Khorana N, Suwanborirux K, Waranuch N, Limpeanchob N, Wisuitiprot W, et al. Bisindole alkaloids and secoiridoids from Alstonia macrophylla Wall. ex G. Don. Fitoterapia 2011;82(6):798804. [129] Xue Y, Xue B. Isolation and structure identification of chemical constituents from the seeds of Descurainia sophia (L.) Webb ex Prantl. Yao Xue Xue Bao 2004;39(1):4651. [130] Mohamed NH, Mahrous AE. Chemical constituents of Descurainia sophia L. and its biological activity. Rec Nat Prod 2009;3(1):5867. [131] Hanson G. The toxicity of plants in equines, a modern three-point approach to disseminating information. A thesis presented in partial fulfillment of the requirements for the degree of Master of Science in Rangeland ecology and management in the college of graduate studies. Moscow: University of Idaho; 2008. p. 839. [132] James LF, Keeler RF, Binns W. Sequence in the abortive and teratogenic effects of locoweed fed to sheep. Am J Vet Res 1969;30:377. [133] Keller K. Cinnamomum species, adverse reactions of herbal drugs. Berlin: SpringerVerlag; 1992. p. 10514. [134] Warkany J. Congenital malformations. Chicago, IL: Year Book Medical Publishers Inc.; 1971. p. 1309. [135] Gaffield W, Keeler RF. Structure-activity relations of teratogenic natural products. Pure Appl Chem 1994;66:240710. [136] Tiwari A, Rajesh SJ, Tiwari P, Nayak S. Phytochemical investigations of crown of Solanum melongena fruit. Int J Phytomed 2009;1:911.

274

Toxicological Survey of African Medicinal Plants

[137] Cano A, Acosta M, Arnao M. Hydrophilic and lipophilic antioxidant activity changes during on-vine ripening of tomatoes (Lycopersicon esculentum Mill). Postharvest Biol Tec 2003;28:5965. [138] Vital PG, Rivera WL. Antimicrobial activity, cytotoxicity, and phytochemical screening of Voacanga globosa (Blanco) Merr. leaf extract (Apocynaceae). Asian Pac J Trop Med 2011;4(10):8248. [139] Herrera AA. In vivo evaluation of the potent angiosuppressive activity of some indigenous plants from Bataan, Philippines. Asia Life Sci 2010;19(1):18390. [140] Liao JC. In Flora of Taiwan, vol. 2 (2nd ed.), Taipei: Editorial Committee of the Flora of Taiwan, 1996. p. 17780. [141] Simo CCF, Kouam SF, Poumale HMP, Simo KI, Ngadjui BT, Green RI, et al. Biochem Syst Ecol 2008;36:23843. [142] Pierangeli GV, Rivera WL. Antimicrobial activity and cytotoxicity of Chromolaena odorata (L. f.) King and Robinson and Uncaria perrottetii (A. Rich) Merr. Extracts. J Med Plants Res 2009;3(7):5118. [143] Ebada SS, Lajkiewicz N, Porco JA, Li-Weber M, Proksch P. Chemistry and biology of rocaglamides (5flavaglines) and related derivatives from aglaia species (meliaceae). Fortschr Chem Org Naturst 2011;94:158. [144] Dapat E, Jacinto S, Efferth T. A phenolic ester from Aglaia loheri leaves reveals cytotoxicity towards sensitive and multidrug-resistant cancer cells. BMC Complement Altern Med 2013;13:286. [145] Ragasa CY, Torres OB, Shen C-C, Mejia MGR, Ferrer RJ, Jacinto SD. Chemical constituents of Aglaia loheri. Pharmacogn J 2012;4(32):2931. [146] Durand, L., Morissette, F. and Lamoureux, G. (1981). Plantes sauvages comestibles, Le groupe Fleurbec. [147] Campbell MA, Brown KS, Hassell JR, Horigan EA, Keeler RF. Inhibition of limb chondrogenesis by a Veratrum alkaloid, temporal specificity in vivo and in vitro. Dev Biol 1985;111:46470. [148] Lee ST, Panter KE, Gaffield W, Stegelmeier BL. Development of an enzyme-linked immunosorbent assay for the veratrum plant teratogens, cyclopamine and jervine. J Agr Food Chem. 2003;51(3):5826. [149] Dugoua JJ, Perri D, Seely D, Mills E, Koren G. Safety and efficacy of blue cohosh (Caulophyllum thalictroides) during pregnancy and lactation. Can J Clin Pharmacol 2008;15(1):e6673. [150] Mole MLJ, Turner CE. Phytochemical screening of cannabis sativa L. I, constituents of an Indian variant. J Pharm Sci 1974;63(1):1546. [151] Mechoulam R, Gaoni Y. Recent advances in the chemistry of hashish. Fortschr Chem Org Naturst 1967;25:175213. [152] Reece AS. Chronic toxicology of cannabis. Clin Toxicol (Phila) 2009;47(6):51724. [153] Cullis JE. Congenital deformities and herbal slimming tablets. Lancet 1962;2:5112. [154] Sieber SM, Whang-Peng J, Botkin C, Knutsen T. Teratogenic and cytogenetic effects of some plant-derived antitumor agents (vincristine, colchicine, maytansine, VP-16213 and VM-26) in mice. Teratology 1978;18(1):3147. [155] Changwichit K, Khorana N, Suwanborirux K, Waranuch N, Limpeanchob N, Wisuitiprot W, et al. Bisindole alkaloids and secoiridoids from Alstonia macrophylla Wall. ex G. Don. Fitoterapia 2011;82(6):798804. [156] Goel RK, Prabha T, Kumar MM, Dorababu M, Prakash, Singh G. Teratogenicity of Asparagus racemosus Willd. Root, an herbal medicine. Indian J Exp Biol 2006;44:5703.

Genotoxicity and Teratogenicity of African Medicinal Plants

275

[157] Goyal RK, Singh JLH. Asparagus racemosus--an update. [Review]. Indian J Med Res 2003;57(9):40814. [158] Sharma PC, Yelne MB, Dennis TJ. New Delhi, central council for research in Ayurveda and Siddha, department of ISM&H, Ministry of Health and Family Welfare (Govt. of India). Database on medicinal plants used in Ayurveda 2005;3:7687. [159] Hayes PY, Jahidin AH, Lehmann R, Penman K, Kitching W, De Voss JJ. Steroidal saponins from the roots of Asparagus racemosus. Phytochemistry 2006;69(3): 796804. [160] Saxena VK, Chourasia SA. New isoflavone from the roots of Asparagus racemosus. Fitoterapia 2001;72(3):3079. [161] Keeler RF. Teratogens in plants. J Anim Sci 1984;58(4):102939. [162] Chase RL, Keeler RF. Mountain thermopsis toxicity in cattle. Utah Sci 1983;44:2831. [163] Yadav R, Kaushik R, Ggupta D. The health benefits of Trigonella foenum-graecum: a review. IJCEA 2009;1(1):0325. [164] Araee M, Norouzi M, Habibi G, Sheikhvatan M. Toxicity of Trigonella foenumgraecum (fenugreek) in bone marrow cell proliferation in rat. Pak J Pharm Sci 2009;22(2):12630. [165] Pe´rez LB, Li J, Lantvit DD, Pan L, Ninh TN, Chai H-B, et al. Bioactive constituents of Indigofera spicata. J Nat Prod 2013;76(8):1498504. [166] Kapoor LD. Handbook of Ayurvedic medicinal plants. Boca Raton, FL: CRC Press; 1990. p. 200. [167] Singh A, Singh RG, Singh RH, Mishra N, Singh N. An experimental evaluation of possible teratogenic potential in Boerhaavia diffusa in Albino rats. Planta Med 1991;57(4):3156. [168] Leyon PV, Lini CC, Kuttan G. Inhibitory effect of Boerhaavia diffusa on experimental metastasis by B16F10 melanoma in C57BL/6 mice. Life Sci 2005;76:133949. [169] Oudhia, P. and Tripathi, R.S. (2002). Identification, cultivation and export of important medicinal plants. In: Proc. National Seminar on Horticulture Development in Chhattisgarh, Vision and Vistas. Indira Gandhi Agricultural University, Raipur (India), p. 7885. [170] Eweka AO. Histological studies of the teratogenic effects of oral administration of Aspilia africana (Asteraceae) leaf extract on the developing kidney of Wistar rats. Int J Toxicol 2008;4:2. [171] Abii TA, Onuoha EN. The chemical constituents of the leaf of Aspilia africana as a scientific backing to its tradomedical potentials. Ag J 2011;6(1):2830.