Academia.eduAcademia.edu
Biomedicine & Pharmacotherapy 144 (2021) 112264 Contents lists available at ScienceDirect Biomedicine & Pharmacotherapy journal homepage: www.elsevier.com/locate/biopha Hydrolyzable tannins (ellagitannins), flavonoids, pentacyclic triterpenes and their glycosides in antimycobacterial extracts of the ethnopharmacologically selected Sudanese medicinal plant Combretum hartmannianum Schweinf Enass Y.A. Salih a, d, *, 1, Riitta Julkunen-Tiitto b, Olavi Luukkanen c, Mustafa K.M. Fahmi e, Pia Fyhrquist a a Faculty of Pharmacy, Division of Pharmaceutical Biosciences, Viikki Biocenter, University of Helsinki, P.O. Box 56, FIN-00014, Finland Faculty of Science and Forestry, Department of Environmental and Biological Sciences, University of Eastern Finland, 80101 Joensuu, Finland c Viikki Tropical Resources Institute (VITRI), Viikki Campus, University of Helsinki, FIN-00014, Finland d Department of Forest Products and Industries, Shambat Campus, University of Khartoum, SUD-13314, Sudan e Department of Forest Management, Shambat Campus, University of Khartoum, SUD-13314, Sudan b A R T I C L E I N F O A B S T R A C T Keywords: Combretum hartmannianum Traditional medicine Tuberculosis Ellagitannins Flavonoids Terpenoids In Sudanese traditional medicine, decoctions, macerations, and tonics of the stem and root of Combretum hartmannianum are used for the treatment of persistent cough, a symptom that could be related to tuberculosis (TB). To verify these traditional uses, extracts from the stem wood, stem bark, and roots of C. hartmannianum were screened for their growth inhibitory effects against Mycobacterium smegmatis ATCC 14468. Methanol Soxhlet and ethyl acetate extracts of the root gave the strongest effects (MIC 312.5 and 625 µg/ml, respectively). HPLC-UV/ DAD and UHPLC/QTOF-MS analysis of the ethyl acetate extract of the root led to the detection of 54 compounds, of which most were polyphenols and many characterized for the first time in C. hartmannianum. Among the major compounds were terflavin B and its two isomers, castalagin, corilagin, tellimagrandin I and its derivative, (S)flavogallonic acid dilactone, punicalagin, and methyl-ellagic acid xylopyranoside. In addition, di-, tri- and tetragalloyl glucose, combregenin, terminolic acid, cordifoliside D, luteolin, and quercetin-3-O-galactoside-7-Orhamnoside-(2→1)-O-β-D-arabinopyranoside were characterized. Luteolin gave better growth inhibition against M. smegmatis (MIC 250 µg/ml) than corilagin, ellagic acid, and gallic acid (MIC 500–1000 µg/ml). Our study justifies the use of C. hartmannianum in Sudanese folk medicine against prolonged cough that could be related to TB infection. This study demonstrates that C. hartmannianum should be explored further for new anti-TB drug scaffolds and antibiotic adjuvants. 1. Introduction Tuberculosis (TB) is a bacterial infectious disease caused mainly by Mycobacterium tuberculosis and spreads by coughing and/or sneezing [1]. TB was discovered in 1882 by the German scientist Robert Koch [2, 3]. Although TB mainly infects the lungs, also the bones and the central nervous system can be affected [4]. Recently osteoarticular TB, a chronic inflammatory disease affecting the bones and joints, has been reported to become a serious problem in China [5]. Moreover, in Africa Buruli ulcer, caused by Mycobacterium ulcerans, is still a significant mycobacterial disease in rural remote regions, leading to amputations if untreated [6]. In 2020, the WHO estimated that more than one-third of the global population is infected with TB [7]. However, TB deaths occur mainly in developing countries, and especially in sub-Saharan Africa [8–10]. The high burden of TB in some countries in Africa is due to poverty and consequently shared and crowded living conditions, which facilitate the spread of TB. Moreover, malnutrition worsens the TB burden in some regions of Africa. Besides, latent M. tuberculosis is known to become activated in patients with severe immunosuppression due to HIV [11]. In addition, in South Africa, TB patients have been observed to * Corresponding author at: Faculty of Pharmacy, Division of Pharmaceutical Biosciences, Viikki Biocenter, University of Helsinki, P.O. Box 56, FIN-00014, Finland. E-mail addresses: enass.salih@helsinki.fi, eyabdelkareem@uofk.edu (E.Y.A. Salih). 1 Permanent address (E. Salih): Department of Forest Products and Industries, Shambat Campus, SUD-13314, University of Khartoum, Sudan. https://doi.org/10.1016/j.biopha.2021.112264 Received 9 June 2021; Received in revised form 14 September 2021; Accepted 27 September 2021 Available online 5 October 2021 0753-3322/© 2021 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 delay their treatments as they cannot afford to travel to the treatment centers daily for observation by health workers to take their TB drugs [12]. Also, a low antibiotic treatment compliance and even treatment interruption due to the complicated and long treatment regimens contribute to increase the pool of more resistant TB strains (multi-drug resistant, MDR-TB and extremely-drug resistant, XDR-TB). These TB strains are resistant to one or more of the available conventional TB drugs, such as rifampicin, isoniazid, thioacetazone, streptomycin, ethambutol, or pyrazinamide [13–16]. Due to the relatively high cost and limited availability to synthetic antibacterial drugs, communities in Africa have largely relied on traditional healers and plant-based drugs to treat infections, including tuberculosis [17,18]. Higher plants, and especially those used in traditional medicine for infections, are rich in a variety of defense compounds against microbial intrusion and could contain valuable new anti-TB drug lead compounds [19,20]. Despite this, there are still no modern antibacterial drugs developed from plant compounds. African medicinal plants could be a valuable source for new antimicrobial drug scaffolds and/or antibiotic adjuvants [18,19,21]. Combretum hartmannianum (Schweinf) (in Sudanese vernacular language: Al-Habeel ‫ )ﺍﻝﻩﺏﻱﻝ‬occurs in the horn of Africa where it grows mainly in Eritrea, Ethiopia, South Sudan, and Sudan Fig. 1F [22,23]. In addition, it is common in the whole Sahel region and West Africa [24, 25]. C. hartmannianum is a shrub or small tree and grows mainly in wooded grassland, high-rainfall savannas, and savannah woodland habitats on well-drained alluvial soils [25]. The fruits need fire to open and release their seeds and fire is also needed for new sprouting from the stem, and thus regular bushfires are needed for the regeneration of this species [26,27]. The crown is very dense and broader when compared to other species of Combretum (Fig. 1A), and the leaves are glabrous with characteristic, extremely extended leaf tips (Fig. 1D). In Sudanese traditional medicine C. hartmannianum is used habitually against a wide range of diseases including asthma, malaria, fever, jaundice, fungal and bacterial infections, including tuberculosis [26,28–34]). Accordingly, some in vitro studies confirm that C. hartmannianum extracts are antibacterial [30,33,35–37]. However, only three papers to date are available on the phytochemistry of C. hartmannianum [38–40]. Based on our ethnopharmacological survey in Sudan, C. hartmannianum is used for the treatment of persistent and prolonged coughing, that could be symptoms of tuberculosis (Table 1). However, only two previous investigations [13,32] reported on the antimycobacterial effects of C. hartmannianum. According to the other study [32], dichloromethane, ethyl acetate, and ethanol extracts of the stem bark were growth inhibitory against M. aurum A+ with MIC values of 0.78, 3.12, and 0.19 mg/ml, respectively. Moreover, a leaf extract of C. hartmannianum inhibited the growth of M. tuberculosis. However, the root and stem wood parts were not studied for their antimycobacterial potential, although we found that decoctions, macerations, and ethanolic tonics made from these plant parts are used for the alleviation of prolonged coughing. Therefore, in this present screening we have chosen to study the in vitro antimycobacterial activity of the root, stem bark, and stem wood extracts of various polarities, including traditional medicinal preparations, against a fast-growing model bacterium for tuberculosis, Mycobacterium smegmatis. To date, the chemical constituents of the root of C. hartmannianum have not been studied. Thus, HPLC-DAD and UHPLC/QTOF-MS were used to decipher the molecular structures of constituents in a root ethyl acetate extract, with special emphasis on polyphenols. 2. Material and methods 2.1. Ethnobotanical survey and collection of plant material To study plants of the genus Combretum that are used for infectious diseases in Sudan, ethnobotanical surveys were performed in seven localities in the Blue Nile and Kordofan regions, in South Eastern and South Western Sudan in 2006 and 2014. The surveys were performed by the first author in collaboration with Associate Professor Haytham H. Geebril from the Faculty of Forestry, Department of Silviculture, University of Khartoum. A questionnaire was used for the interviews and focus was put on questions relating to symptoms that could be a sign of bacterial infection and TB, such as persistent coughing, fever, fatigue, and chest pain (Appendix). Among the seven Combretum spp. that were growing naturally in the study area, C. hartmannianum was the most commonly used species for the treatment of TB and its symptoms, and it was used in the same way in all of the villages we visited (Table 1). Fig. 1. (A), C. hartmannianum occurs in savannah woodland and wooded grassland in Sudan; (B) rough and finely fissured bark; (C) samara fruits; (D) leaves with extremely long tips that are almost as long as the leaf blade; (E), flowers borne in axillary spikes. Photos by: E. Y. A. Salih and H. H. Gibreel 2006. (F) Occurrence of C. hartmannianum in Africa; (G), Study site in the Blue Nile region in Sudan is marked in red color on the map. Source: African Plant Database. 2 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Table 1 Ethnobotanical uses, occurrence, phenology, and phytochemistry of Combretum hartmannianum. Results from our ethnobotanical survey in Blue Nile State, Sudan, and the phytochemical analysis reported in this present paper are marked with an asterisk *. The species name, synonyms and Sudanese vernacular name Botanical data Gum exudate and its traditional/commercial uses and chemical constituents * Chemical constituents Traditional uses Combretum aculeatum Syn. Combretum ovale R.Br. ex G. Don; Combretum alternifolium Spreng. Sudanese vernacular name: Shuheit 4–5 m in height *[41]. Flowering/fruiting time: March to June/July to October *[41] Gum was observed from the stem * No reports on the chemical constituents of the gum Palmitoleic acid, Ethyl linoleate, α-Linolenic acid, Stearic acid, Omega-6 (Linoleic acid), Behenic acid, Dotriacontanol, Squalene, Lycoxanthin from the aerial parts[42]. Combretun ghasalense Engl. & Diels. Syn. Combretum ternifolium Engl. & Diels. Combretum fragrans F.Hoffm. Combretum adenogonium Steud. ex A. Rich. Sudanese vernacular name: Habil Um-ismaeel Combretum collinum Fresen. Syn. Combretum bongense Engl. & Diels. Sudanese vernacular name: Habil el-gouruz 12–15 m in height *[44]. Flowering/fruiting time: November to February /February to March *[45] Tapping produces a long line of gum * Gum contains arginine, aspartic acid, glutamic acid, glycine, histidine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, valine[46] Arjunolic acid, Velutin (3′ ,7-dimethoxy-4′ ,5dihydroxyflavone), Asiatic acid, Belamcanidin (5-hydroxy6,7,3′ ,4′ -tetramethoxyflavone), Cirsilineol (5,4′ -dihydroxy6,7,3′ -trimethoxyflavone), Combretin B, Cirsimaritin (5,4′ dihydroxy-6,7-dimethoxyflavone), 3β-Acetoxy-20,24-epoxy11,25-hydroxy-dammarane, Combretin A[44]. 10 m in height *[48]. Flowering/fruiting time: January to February /January to March *[41]. Myricetin-3-O-rhamnoside, Myricetin-3-O-glucoside, Tetracosanoic acid, Squalene, Campestrol, Palmitic acid[49]. Combretum glutinosum Perr. Ex DC. Syn. Combretum glutinosum var. relictum (Hutch & Dalziel) Aubrév. Sudanese vernacular name: Habil el gebel 12 m in height *[50]. Flowering/fruiting time: December to March / October to December *[51]. The exudate is an edible gum * Gum contains sarginine, aspartic acid, glutamic acid, glycine, histidine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, valine[46] Gum exudate observed * Gum contains arginine, aspartic acid, glutamic acid, glycine, histidine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, valine[46]. Leaves decoctions as laxatives and for venereal disease; stem against skin infections, including leprosy; root decoction against flu * [42]. Decoction of the aerial parts against TB symptoms *[43]. Bark for treating stomach diseases; smoke from the wood as perfume * . Leaves and stem cold and hot water extracts are used against gastrointestinal infectious diseases * Stem bark used as a tea against malaria * Root decoction is used against cough *[47]. The gum is used against toothache; root and leaves decoctions are used against diarrhea and cough* Root and stem water extracts are used against intestinal pain * . Leaves infusions are used against cough[49]. Combretum hartmannianum Schweinf. Syn. Poivrea hartmanniana Schweinf. Sudanese vernacular name: Al-Habil, Subagh Shrub-tree grows up to 25 m in height * . Characteristic leaf morphology with long, pointed leaf tips that differs from all other species of Combretum. Bark rough and finely fissured. Crown rounded *[23]. Occurrence: Horn of Africa (Erithrea, Ethiopia, Sudan, Somalia) and Sahel region to Senegal. High rainfall savanna woodland and wooded grasslands on well-drained alluvial soils [23,25]. Flowering/fruiting time: Flowering in January to February and fruiting from April to May * Water-soluble and edible gum exudate, called kaakol* . The gum contains the sugars galactose, arabinose, mannose, xylose, rhamnose, glucuronic acid, 4-Omethylglucuronic acid, and galacturonic acid[58] and the amino acids arginine, aspartic acid, glutamic acid, glycine, histidine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, valine[46]. Apigenin, isorhamnetin derivatives, phenantherenes, kaempferol and its derivatives, quercetin and its derivatives, chrysoeriol and its dervatives[39]. Flavogallonic acid dilactone and terchebulin[40]. Ursolic acid, pomolic acid, corosolic acid, arjunic acid, arjunglucoside I, trachelosperoside E-1, combreglucoside, chebuloside II, and 2α,3β,6β,19α-tetrahydroxyoleanolic acid 28-O-β-D-glucopyranoside[38]. In this present study, we have tentatively characterized the following compounds from a root ethyl acetate extract of C. hartmannianum: Terflavin B and its isomer, Castalagin, Corilagin, S- flavogallonic acid dilactone, Tellimagrandin I and its derivatives, Punicalagin, Terchebulin, Combregenin, Isorhamnetin, Cordifoliside D, Luteolin, Ellagic acid, and its derivatives, Quercetin-3-O-galactoside-7-O-rhamanoside-(2–1)-O-Darabinopyranoside, Terminolic acid, mono-, di-, tri-, and tetra-galloyl glucose * . Combreglutinin, 2,3-(S)-hexahydroxydiphenoyl-D-glucose [53], punicalagin, punicalin[52,54]. Root, stem, and leaves decoction for hypertension, as laxatives, for stomach pain, hepatitis, respiratory symptoms, and fever *[55,56]. Leaves extracts are used to cure cough[57]. The traditional medicinal preparation called “Dokhan” is made from burning the root and stem and a smoke bath is used to cure sexually transmitted infectious diseases * . Leaves decoction and maceration are used to cure ascites (that can be caused by TB), stem bark, leaves, or fruits are mixed with gum against jaundice, for skin infections and wounds; root, leaf, and bark water extracts are used as a mouth wash and against inflamed gums; water extracts and decoctions, as well as, ethanolic tonics of stem bark, and root, are used to slow down a persistent cough (TB), and against eye infections *[39,59]. (continued on next page) 3 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Table 1 (continued ) The species name, synonyms and Sudanese vernacular name Combretum nigricans Guill. & Perr. Syn. Combretum elliotii Engl. & Diels; Combretum lecananthum Engl. & Diels. Sudanese vernacular name: Habil Combretum molle R. Br. Ex G. Don. Syn: Combretum deserti Engl.; Combretum reticulatum Fresen.; Combretum tetragonum M.A. Lawson; Combretum pisoniiflorum (Klotzsch) Engl. Sudanese vernacular name: Habil Khrisha Botanical data Gum exudate and its traditional/commercial uses and chemical constituents * Chemical constituents 12 m in height *[63]. Flowering/fruiting time: February to March /April to July *[64]. The bark exudates an edible gum* , that contains rhamnose and uronic acid[65]. Arjunglucoside I, arjungenin, combreglucoside, mollic acid, combregenin, arcapitin A, 11α-acetoxy-20,24-epoxy-25hydroxy-dammar-3-one, 20,24-epoxy-11α,25-dihydroxydammar-3-one, 20,24-epoxy-12β-25-dihydroxy-dammar-3-one [53]. 15 m in height *[48]. Flowering/fruiting time: February to May / May to August *[45]. Exudates a gum * that contains arginine, aspartic acid, glutamic acid, glycine, histidine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, and valine[46]. Punicalagin, punicalin[54,66], combretene A and B, combregenin, arjungenin, arjunglucoside I, combreglucoside, sericoside, mollic acid and its glycosides; mollic acid glucoside, mollic acid arabinoside, mollic acid xyloside[53]. Traditional uses Water extracts of the barks and infusion of the leaves for jaundice[60]. Paste and ointments from crushed and powdered leaves against skin infections, and leprosy *[55]. Stem bark used as a tea against Malaria * . “Dokhan”, a smoke bath from the burning roots and stem is used to cure sexually transmitted infectious diseases. The smoke of bark and wood against arthritis rheumatism and to treat dryness of skin in Sudan *[28]. For bacterial diseases * [61,62]. Leaf decoctions for jaundice, external infections on the skin, malaria, and febrile diseases[39]. Leaves decoction is used for intestinal disorders, against fever, acne, jaundice, respiratory symptoms, and rheumatism * . Paste and ointments from leaves and stem wood are used against skin infections * . The burnt leaves produce an aromatic smoke that is used to induce abortion and enhance constipation, and as an antianthelmintic especially against hookworms; leaves decoction against cough *[66]. Paste and ointments from leaves are used against skin infections and leprosy * . African plant database and The Plant List were used as sources for the scientific name synonyms. http://www.theplantlist.org/tpl1.1/record/kew-2732601 http://www.villege.ch/musinfo/bd/cjb/africa/details.php?langue=an&id=189625 Therefore, C. hartmannianum was selected for further analysis on its antimycobacterial extracts and compounds, and bulk collections were done for this species. Root and stem bark material were collected from four individuals of C. hartmannianum growing in savannah woodland and wooded grassland habitats in the Blue Nile and Kordofan regions. Specimens of C. hartmannianum were botanically identified by the first author and the taxonomists, Dr. Haytham H. Gebreel and Dr. El-Sheikh Abd Alla El-Sheikh. Voucher specimens were authenticated and deposited at the Faculty of Forestry, University of Khartoum, Sudan. The collected plant parts were dried in the shade, grinded, and kept in paper bags for further use. Plant material from different plant individuals was kept separate. 2.2. Extraction techniques 2.2.1. Hot (Soxhlet) and cold methanol extractions A hot extraction method was employed by using a Soxhlet apparatus (Sigma-Aldrich, Germany). Fifty grams of dry and powdered plant samples were loaded into a Soxhlet thimble in the glass chamber of a Soxhlet apparatus. The extraction solvent, 100% methanol, was poured into a round-bottom distillation flask that was connected to the Soxhlet apparatus. The solvent was brought to a boil and the extraction process was continued for 6 h whereafter the resulting extracts were cooled down and transferred to a rotary evaporator (Heidolph VV2000). After rotary evaporation, the extracts were further dried to complete dryness in a freeze drier for 2–3 days (lyophilizer, HETO LyoPro 3000, Denmark). The resulting dry extracts were dissolved in methanol for antimycobacterial testing (50 mg/ml stock solutions). 4 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Cold methanol extraction was made to compare the composition of the extracts with the hot methanol extracts since hot extraction could lead to the loss of some compounds due to molecular degradation. For the cold methanol extractions, 20 g of various parts of C. hartmannianum were extracted at room temperature for 24 h with 500 ml methanol and using a magnetic stirrer (RCT, digital). The extracts were filtered with Whatman filter paper (150 mm, Germany), whereafter they were dried using a rotary evaporator and a water bath at + 40 ◦ C. These extracts were then further dried for three days using a lyophilizer (HETO LyoPro 3000, Denmark). Pasteur pipette (P 4518–5X, Accupipette ™ Pipets/ DADE and DURAN®, ring Caps, Germany). The mobile phase consisted of methanol, water, and orthophosphoric acid (50:50:1, v/v/v). Fluorescent and quenching spots were observed at the wavelengths of 366 and 254 nm, respectively, using a Camaq Reprostar UV-detector (3 TLC Visualizer). Retardation factors (Rf values) of the fractions and pure compounds were calculated according to the formula 2: Rf value = Distance moved by the fractions or compounds / Distance moved by the solvent (2) For the analysis of the antioxidant effects of extracts and compounds of C. hartmannianum, the 2,2-diphenyl-1-picrylhydrazyl (DPPH) reagent (C18H12N5O6, Sigma-Aldrich, China), was prepared in methanol to a concentration of 0.2% (w/v). The developed thin layer plates containing the extracts, fractions, and pure compounds were sprayed with the DPPH reagent to qualitatively investigate the oxidative reactions of the separated spots in visible light. The compounds and fractions with antioxidant properties resulted in a change in the color of the stable free radical, DPPH, from dark purple (color of the DPPH in the form of a radical) to colorless. This color change is a result of the weak hydrogen bonds in the antioxidant compounds present in the extracts that react with the strong, stable DPPH radical to form the colorless DPPH-H complex. Retardation factors of standard compounds (ellagic acid, gallic acid, corilagin, and luteolin, (Sigma-Aldrich) were compared with the retardation factors of the spots present in the fractions and extracts, to localize these compounds in the extracts. 2.2.2. Sequential extraction and solvent partition Sequential extraction, starting with lipophilic solvents and gradually extracting with more polar solvents, was used according to the method described in Salih et al. [67,68]. A hundred grams dry powder of the stem bark, stem wood, and root of C. hartmannianum was extracted overnight, first with 1000 ml n-hexane, secondly with 1000 ml of dichloromethane or chloroform (for the stem wood and bark) and acetone (for the root part), and finally with 80% methanol. All extractions were performed at room temperature. A magnetic stirrer was used to facilitate the extraction. Solvent partition was performed using the 80% methanol extract that was combined with an equal volume of ethyl acetate in a separation funnel and mixed carefully (1000 ml, a Nalgene® FEP). This sequential extraction and solvent partition led to the separation of n-hexane, chloroform, dichloromethane, acetone, aqueous (80% methanol), and ethyl acetate fractions. A rotary evaporator was used to dry the fractions before they were freeze-dried for three days in a lyophilizer (HETO LyoPro 3000, Denmark). The dried fractions were dissolved in methanol (stock solutions of 50 mg/ml) for antimycobacterial testing. 2.3.2. HPLC-UV/DAD analysis Phytochemical screening of the extracts and fractions of C. hartmannianum was performed using an Agilent Chemstation HPLCUV/DAD system (Waters Corp., Milford, USA) and using the method described as the HPLC method II in Fyhrquist et al. [73]. A reversed-phase column, Hypersil Rp C-18 (length 10 mm; ID 2 mm) was used for phytochemical separation. A diode-array UV detector (a 991 PDA), an automatic sample controller, and a pump (Waters 600 E) were connected in the HPLC system. The mobile phase eluents comprised of solvent A (aqueous solvent consisting of 1.5% tetrahydrofuran and 0.25% orthophosphoric acid) and solvent B (100% methanol). The gradient elution was programmed to a flow rate of 2 ml/min and with a sample injection volume of 10 µl (2 mg/ml in 50% MeOH). The UV detector wavelengths were adjusted at 220, 270, 280, 320, 360, and 380 nm, to record the UVλ absorption maxima spectra for the identified peaks. The retention times and UVλ absorption maxima spectra of the detected compounds were compared with the compounds in the natural compounds library available in our computer system Agilent Chemstation library as well as with information obtained from SciFinder, PubChem, and other sources of literature [74,75]. 2.2.3. Water extracts Twenty grams of powdered plant materials were used for the hot (decoctions) and cold water (macerations) extractions. For the decoctions, the samples were first brought to a boil for 5 min and then cooled down whereafter extraction was continued at room temperature overnight and using a magnetic stirrer. No heating was used for the maceration method and the extraction was done overnight at room temperature with a magnetic stirrer facilitating extraction. After extraction, the water extracts were decanted into Eppendorf centrifuge tubes (50 ml v/v, Germany) and centrifuged in an Eppendorf centrifuge (AG, 5810 R, Germany) at 3000 rpm for 15 min. The supernatants of the centrifuged extracts were frozen at −20 ◦ C for 24 h and dried using a lyophilizer (HETO LyoPro 3000, Denmark). The dried extracts were dissolved in methanol to a concentration of 50 mg/ml for antimycobacterial testing [69]. The percentage extraction yields of the extracts and fractions were calculated according to the formula 1 [70]: 2.3.3. UHPLC/QTOF-MS method Ultra-high performance liquid chromatography coupled to quadrupole time of flight mass spectrometry (UHPLC/QTOF-MS) in negative mode was used to identify exact molecular masses [M] between 100 and 2000 m/z. A method described by Taulavuori et al. [76] and Julkunen-Tiitto and Sorsa [77], was employed to identify the various compounds, with emphasis on polyphenols, in a root ethyl acetate extract of C. hartmannianum. Shortly, the Agilent Technologies UPLC-DAD (Model 1200 Agilent Technologies)-JETSTREAM/QTOF-MS (Model 6340 Agilent Technologies) and a reverse-phase column C18 (2.1 ×60 mm, 1.7 µm, Agilent technologies) were set into a UHPLC/Q-TOF-MS system. Eluent (A) was aqueous 1.5% tetrahydrofuran and 0.25% acetic acid, and eluent (B) was 100% methanol. Gradient elution was used as follows: from 0 to 1.5 min, B 0%, from 1.5 to 3 min, 0–15% B, from 3 to 6 min, 10–30% B, from 6 to 12 min, 30–50% B, from 12 to 20 min, 50–100%, and from 20 to 22 min, 100–0% B. Percentage extraction yield (%) = weight of the dry extract/weight of dry plant material before extraction × 100 (1) 2.3. Antioxidant analysis and phytochemical profiling 2.3.1. Thin-layer chromatography and qualitative antioxidant analysis A reversed-phase thin layer chromatography method combining TLC separation with DPPH-antioxidant analysis was used according to Wang et al. [71] and Sethiya et al. [72] to detect the antioxidant compounds in C. hartmannianum root methanolic Soxhlet extracts. A TLC was prepared with the plates sized at 10 × 20 cm (RP-18 F 254 S TLC-plates, Darmstadt, Germany). Ten microliters (10 µl) of corilagin, gallic acid, luteolin, and ellagic acid (1 mg/ml in methanol, Sigma-Aldrich, Germany) and 20 µl of the extracts and fractions (50 mg/ml in methanol) were applied manually 1.5 cm from the bottom of the TLC plate using a glass 5 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 The mass accuracy (PPM) was calculated according to formula 3: Sigma-Aldrich, China), quercetin (Merk Art. 7546, Darmstadt, Germany), corilagin (Sigma-Aldrich, Darmstadt, Germany) and apigenin (Extrasynthese Genay, France) were two-fold diluted in Dubos broth at concentrations ranging from 0.030 to 1000 μg/ml. Preparation for the tests started by transferring a few colonies of M. smegmatis into 25 ml of Dubos broth and incubating this suspension for three days in a shaking incubator (Stuart® SI500, UK) at + 37 ◦ C, 200 rpm. The inoculum for the test was prepared by pipetting 2 ml from this suspension into a sterile test tube and mixing carefully. 1 ml from this suspension was pipetted into a disposable UV cuvette (BrandTech®, USA), to measure the turbidity at 625 nm using a UV-Visible Spectrophotometer (Pharmacia LKB-Biochrom 4060). The suspension that was left in the test tube was diluted following the guidelines certified by the Clinical and Laboratory Standards Institute [79] to reach an absorbance of 0.1 at 625 nm (consisting of approximately 1 × 108 CFU/ml). This suspension was diluted further with Dubos broth so that the inoculum used for the microdilution assay contained 5 × 105 CFU/ml. A hundred microliters from this prepared inoculum was added to each well of a 96-well microplate, except the sample control wells (Nunc, Nunclone, Denmark). In addition, 100 µl of C. hartmannianum extracts, pure compounds or rifampicin were added in the wells, so that the final volume in the wells was 200 µl and thus consisting of 2.5 × 105 colony-forming units (CFU)/ml. The wells containing extracts, or antibiotics and bacteria were called test wells (GT). Moreover, the microplate wells consisted of growth control (GC) wells that contained 100 µl of the bacterial suspension (containing 5 × 105 CFU/ml) and 100 µl of Dubos broth so that the final volume of 200 µl contained an inoculum of 2.5 × 105 CFU/ml. In addition, the sample control wells (SC), made individually for each two-fold dilutions of the extracts and rifampicin, contained a volume of 100 µl of the extracts, pure compounds or antibiotics, and 100 µl of Dubos broth without bacterium. Sample controls were used to subtract eventual absorption resulting from the color pigments or precipitates in the extracts and the antibiotic. Methanol and hexane were used as solvent controls at a maximum concentration of 5%, and at this concentration, no growth inhibition was detected. The 96-well microplates were incubated in a shaking incubator (Stuart® SI500, UK) at + 37 ◦ C at 100 rpm for four days. Subsequently, the turbidity of the wells was measured at 620 nm using a spectrophotometer (Victor 1420, Wallac, Finland). The percentage growth was calculated as the mean percentage of growth of triplicates in relation to the growth of the growth control (the growth control of freely growing bacteria was defined as growing 100%). Therefore, the percentage growth inhibition was obtained by subtracting the percentage growth value of the test sample from the growth control (100% growth). Calculations for percentage growth and % growth inhibition are explained by the formulas (5) and (6) below. All assays were done in triplicate, and the experiments were repeated three times. The lowest concentration that inhibited ≥ 90% of the mycobacterial growth (resulting in no visible growth) was defined as the minimum inhibitory concentration (MIC). Applied formulas for the percentage of growth and growth inhibition calculations: ppm (parts per million mass error) mass accuracy = (M measured –M calculated) × (3) 106/M calculated Where M measured is the measured mass in QTOF-MS and M calculated is the exact calculated mass according to the molecular formula of the compound. Since QTOF-MS was used in negative mode, the monoisotopic mass of the hydrogen atom (1.0078) was subtracted from the calculated masses. 2.4. Antimycobacterial assays 2.4.1. Agar diffusion assay Primary antimycobacterial testing was performed for all extracts using an agar diffusion method according to Fyhrquist et al. [73]. Before the test, Mycobacterium smegmatis ATCC 14468 was grown on Löwenstein–Jensen agar (Becton–Dickinson & Company, USA) at + 37 ◦ C for five days. For the test, Petri dishes (Ø 14 cm, Bibby Sterilin, UK), were used. A bottom layer that consisted of 25 ml of base agar (Antibiotic medium Number 2, Difco, UK) and a top layer of 25 ml Middlebrook 7H10 agar (Difco, UK), supplemented with oleic albumin dextrose catalase (OADC, Difco supplement) were added to the Petri dishes. The Petri dishes were inoculated with 200 µl of M. smegmatis suspension, containing approximately 1.0 × 108 CFU/ml. Six filter paper disks (Ø = 12.7 mm, Schleicher & Schuell) loaded with 200 µl of the tested extracts (50 mg/ml) and the positive control rifampicin (10 mg/ml, Sigma-Aldrich) were placed equidistantly on the inoculated Petri dishes. Methanol and hexane were used as negative controls. Before incubation, the Petri dishes were kept in the cold room (+4 ◦ C) for two hours where after the dishes were incubated for five days at + 37 ◦ C. The tests were performed in triplicates, with each replicate sample on a separate Petri dish. The results were calculated as the mean of the diameters of the inhibition zones (IZD in mm) of three replicates ± standard error of the mean (SEM), with the replicates placed on separate Petri dishes. In addition, also the activity indexes of the extracts were calculated in relationto rifampicin according to the formula 4 [70, 73]: AI (Activity index) = Inhibition zone of the plant extract / Inhibition zone of rifampicin (4) The agar diffusion method was also employed to obtain the approximate minimum inhibitory concentration (MIC) values for some extracts, such as methanol extracts of the wood and bark and acetone, cold and hot water extracts of the root. The mentioned extracts form excess precipitation in broth, making it impossible to use the turbidimetric microplate assay for measuring MIC. For these experiments, 200 µl of two-fold dilutions of the extracts (39–5000 µg/ml) and rifampicin (0.030–1000 µg/ml) were pipetted onto sterile filter papers. The approximate MIC was estimated as the smallest concentration still resulting in a visible inhibition zone (IZ ≤1 mm) around the filter paper [69,78]). Moreover, the inhibition zone diameter (IZD) of the two-fold dilutions were plotted against the concentrations of the extracts and compounds to investigate the linearity (dose-dependency) of the growth inhibition. Percentage (%) bacterial growth = [(x‾ GT A620 – x‾ SC A620) / x‾ GC A620) × 100] (5) Percentage (%) inhibition of growth = 100 (% growth of the growth control) – [(x‾ GT A620 – x‾ SC A620) / x‾ GC A620) × 100] (6) 2.4.2. Turbidimetric microplate assay C. hartmannianum extracts and rifampicin were tested for their minimum inhibitory concentration (MIC) and percentage growth inhibition against M. smegmatis ATCC 14468 using a turbidimetric microplate method as described by Salih et al. [70] and Fyhrquist et al. [73]. C. hartmannianum extracts were two-fold diluted from 39 to 5000 µg/ml and rifampicin from 0.030 to 1000 µg/ml in Dubos broth (Difco). Besides, standard compounds, that had been detected in extracts of C. hartmannianum, such as luteolin (Sigma-Aldrich, Darmstadt, Germany), ellagic acid (Sigma-Aldrich, England), gallic acid (G-7384, x‾ = the average of the triplicates of the examined wells; GT A620 = turbidity of the test well (plant extracts, compounds, or antibiotics incubated with the bacterium); SC A620 = turbidity of the sample control wells (controls containing the plant extracts, compounds, or rifampicin only with no bacteria); GC A620 = turbidity of the growth control. 2.4.3. Calculation of total activity The total antimycobacterial activities were calculated to assess the potential of the various solvents to extract antimycobacterial 6 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 compounds. The total activity indicates the maximum volume of solvent to which 1 g of an extract could be diluted into and still possess antimicrobial activity [80]. The total activity was calculated using formula (7) below as outlined by Eloff [80]): (7) Total activity (ml/g) = extraction yield of extract (mg/1000 mg) / MIC of the extract in mg/ml. Table 2 In vitro antimycobacterial activity of extracts and fractions of Combretum hartmannianum against M. smegmatis ATCC 14468. Results were obtained with an agar diffusion method and are presented as the diameters of inhibition zones (IZD) in mm. Activity indexes (AI) were calculated in relation to rifampicin. 2.5. Statistical analysis of data Data resulting from the antimycobacterial assays (diameters of inhibition zones, IZD, and percentage inhibition of growth) were expressed as the mean of replicates (n = 3) ± standard error of the mean (SEM), obtained from three independent experiments. For the extraction yields, the yield was expressed as the mean of two separate extractions ± SEM. 3. Results 3.1. Ethnopharmacological uses of Combretum spp., with special emphasis on C. hartmannianum in the villages of the Blue Nile state, Sudan According to our results, Combretum aculeatum, C. ghazalense, C. collinum, C. glutinosum, C. nigricans, C. hartmannianum and C. molle occur commonly in the Kordofan and Blue Nile state areas in Sudan and are used for the treatment of various infectious diseases, acne and infected wounds (Table 1). However, of these species, C. hartmannianum was the most frequently used among the traditional healers for the treatment of symptoms related to TB, including persistent cough. Traditional healers in all villages in Al-Azaza, A-Rosiers, A-Damazein, Baw, and Al-Ubied localities that we visited, use C. hartmannianum as a medicinal plant in several traditional applications to treat infectious diseases, and among them respiratory infections (Table 1). Water extracts (macerations and decoctions) and tonics in ethanol from the leaves, roots, and stems of C. hartmannianum are used to treat a persistent cough that could be due to TB. Moreover, macerations are used for skin ulcer infection, toothache, wound inflammations, and infections in general. In addition, tonics, pastes, and ointments of C. hartmannianum leaves, fruits, stem wood, and roots are frequently used to cure various types of acne, skin infections, and leprosy caused by Mycobacterium leprae and M. lepromatosis. The tonics are prepared by cooking the plant for 30–40 min in water or as overnight macerations in ethanol, prepared traditionally from the date palm or sorghum. For the pastes, dried plant powder is boiled in animal lipid called "Karkar" for 2–3 h and then mixed with clove and a little volume of water, whereafter the preparation is cooled down before use. The pastes are used to cover infected areas (wounds, fungal infections) of the skin. Ointments are prepared in the same way as the pastes, by boiling the plant material in "Karkar" for 2–3 h, cooled-down and filtrated whereafter the preparation solidifies. After solidification, the ointments are used as creams on the skin. Extracts and C. hartmannianum fractions IZD, Average SEM AI R MeSox* ** R acetone R hex R EtoAc R aqu R Dic R H2O‫٭٭‬ R Me‫٭٭‬ R HH2O* ** W MeSox W hex W H2O‫٭‬ W CHCl3 W HH2O W aqu W EtoAc W Me‫٭٭‬ B EtOAc B aqu B MeSox B hex B CHCl3 B H2O‫٭٭‬ B Me‫٭٭‬ B HH2O Rifampicin 31.50 25.67 19.73 24.67 20.50 13.00 21.00 20.00 22.00 20.17 19.00 18.00 14.67 18.17 19.67 20.67 24.33 23.00 19.00 24.83 15.00 16.00 20.00 24.67 20.33 47.5 0.29 0.33 0.15 0.17 0.29 0.00 0.00 0.00 0.17 0.33 0.00 0.00 0.17 0.17 0.17 0.17 0.17 0.00 0.00 0.17 0.00 0.00 0.17 0.17 0.17 0.29 0.66 0.54 0.42 0.52 0.43 0.27 0.44 0.42 0.46 0.42 0.40 0.38 0.31 0.38 0.41 0.44 0.51 0.48 0.40 0.52 0.32 0.34 0.42 0.52 0.43 1.00 W, stem wood; B, stem bark; R, root; L, leaves; Rb, root bark; Me* , cold methanol extracts; EtOAc, ethyl acetate extracts; hex, hexane extracts; Dic, dichloromethane extracts; CHCl3, chloroform extracts; aqu, aqueous extracts; HH2O, hot water extracts (decoctions); MeSox, methanol Soxhlet extracts; H2O* , Cold water extracts (macerations); NA, not active. NT, not tested; Filter paper disks (∅ 12.7 mm) were saturated with 200 µl extracts/fractions (50 mg/ ml) and rifampicin (10 mg/ml); AI, Activity index in relation to rifampicin; Diameter of inhibition zones (IZD) recorded in mm as mean of triplicates (n = 3) ± SEM of three experiments. Most promising results (IZD ≥20 mm) were indicated by bold text; the underlined extract was analyzed phytochemically with HPLC-DAD and UHPLC/QTOF-MS; * ** analyzed preliminary with HPLC-DAD. with a methanol Soxhlet and an ethyl acetate extract of the root with IZD of 31.50 and 24.67 mm, respectively, and MIC values of 312.5 and 625 µg/ml, respectively. Moreover, the growth inhibition of the methanol Soxhlet extract of the root was dose-dependent (Fig. 3), and displayed the highest total activity of 660 ml/g, resulting from a high extraction yield of 20.6% and the MIC of 312 µg/ml (Fig. 2 and Table 3). Also stem bark extracts, such as a cold methanol and a methanol Soxhlet extract, inhibited the growth of M. smegmatis with large inhibition zone diameter (IZD) of 24.33 and 24.83 mm, respectively. Acetone was found to be the best solvent by far to extract compounds from the root, resulting in an extraction yield of 35% (Fig. 2). Moreover, the acetone extract of the roots gave an IZD of 25.67 mm, and a MIC value of 2500 µg/ml. Therefore the total activity of the acetone extract was 140 ml/g (350/2.5 = 140). Applications used in traditional medicine, i. e. the decoctions and macerations of the root and stem bark, showed some growth inhibition (IZD 20–22 mm), but the MIC value for the decoction of the root was high (5000 µg/ml) and thus resulting in low total activity of 19.6 ml/g. In addition, some compounds that we detected in C. hartmannianum roots were evaluated for their antimycobacterial effects (Table 3). Of these compounds, luteolin showed the strongest effects (MIC 250 µg/ ml), while gallic acid and ellagic acid showed MIC values of 500 µg/ml and corilagin a MIC value of 1000 µg/ml. Moreover, apigenin and quercetin, of which apigenin and quercetin glycosides were previously 3.2. Antimycobacterial effects, extraction yields, and total activity A total of thirty-one extracts of various polarities from the stem wood, stem bark, and root of Combretum hartmannianum, were subjected to antimycobacterial screening (Tables 2 and 3). Alongside, also the total activities, which are dependent on the extraction yields (Fig. 2) and MIC values are shown in Table 3. The results revealed that hydrophilic extracts of C. hartmannianum were mostly more potent growth inhibitors than the lipophilic extracts, although this result could be due to the agar diffusion method that was used for the primary screening (Table 2). Moreover, MIC values were not estimated for the lipophilic extracts due to the small volumes of samples available. When compared to the more polar extracts, the hexane, dichloromethane, and chloroform extracts of C. hartmannianum showed low to moderate growth inhibitory activity against M. smegmatis with inhibition zone diameters (IZD) ranging from 13 to 19.7 mm (Table 2). The strongest growth inhibition was obtained 7 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 C. hartmannianum, all of them giving growth inhibitory effects against M. smegmatis in our analysis (Tables 2 and 3). The results of the antioxidant analysis are shown in Fig. 4. Accordingly, those spots (compounds or fractions) on the TLC plate that show antioxidant activity exhibit a light yellow to white color due to a color change of the DPPH reagent from a dark purple (violet) to yellow, as a result of the reaction and neutralization of the DPPH radical with the hydrogen atoms and other functional groups found in the antioxidant molecules of the extracts [81]. As can be seen in Fig. 4 (1C, 2C, and 3C) the phytochemical profiles of all three extracts look very similar. Strong antioxidant activity could be found among compounds within a wide range of polarities in all the studied extracts, and especially among the ellagic acid derivatives, the ellagitannins, and gallic acid, with luteolin being less active as judged by DPPH color change intensity (Fig. 4, 1 D and F). The results show that the root decoction is rich in antioxidant ellagitannins that could be important for its use to treat infections in traditional medicine. Table 3 Minimum inhibitory concentrations (MIC) in µg/ml of extracts and fractions of C. hartmannianum and pure compounds against Mycobacterium smegmatis ATCC 14468. The total activity was calculated as the relation between MIC and extraction yield. C. hartmannianum extracts MIC Extraction yield in mg from 1 g plant material (Percentage yield) Total activity (ml/g) W EtOAc W Me* B MeSox B EtOAc B Me‫٭‬ R MeSOx R acetone R aqu R EtOAc R HH2O Rifampicin 5000 (IC 97)* * 2500 2500 5000 (IC 90)* * 5000 312.5 (IC 95)* * 2500 5000 625 (IC 99)* * 5000 39.06 (3.90, IC 98)* * 45 (4.5%) 28 (2.8%) 189 (18.9%) 97 (9.7%) 21 (2.1%) 206 (20.6%) 351 (35.1%) 200 (20.0%) 115 (11.5%) 98 (9.8%) 9 11.2 75.6 19.4 4.2 660 140 40 184 19.6 Pure compounds Corilagin Gallic acid Luteolin Quercetin Apigenin Ellagic acid 3.4. HPLC-DAD and UHPLC/QTOF-MS results on the phytochemistry of a root extract An ethyl acetate extract of the root of C. hartmannianum was chosen for phytochemical analysis due to the fairly good antimycobacterial activity shown by this extract. Another criterium for the use of this extract for our phytochemical analysis was that, when compared to the methanol extract of the root, this extract had a cleaner profile, with the ellagitannins (ETs) better separated from each other. Moreover, according to our preliminary HPLC-DAD results, most of the ETs present in the methanol extract also occurred in the ethyl acetate extract. HPLCDAD and UHPLC/QTOF-MS analysis led to the identification of 54 compounds of which most were phenolic compounds, including hydrolyzable tannins and flavonoids (Fig. 5, Fig. 6, and Table 4). 1000 (IC 94)* * 500 (IC 98)* * 250 (IC 91)* * 250 (IC 94)* * 250 (IC 97)* * 500 (IC 98)* * W, stem wood; B, stem bark; R, root; L, leaves; Me* , cold methanol extracts; EtOAc, ethyl acetate extracts; HH2O, hot water extracts (decoctions); MeSox, methanolic Soxhlet extracts; H2O* , Cold water extracts (macerations); IC, the inhibitory concentration that indicates the percentage growth inhibition at the MIC concentration; * *, MIC measured with microdilution method and other MIC results obtained with an agar diffusion method. Corilagin, gallic acid, and luteolin were characterized from an ethyl acetate extract of the root of C. hartmannianum in this study. 3.4.1. Ellagitannins, ellagic acid and gallic acid derivatives Ten ellagitannins (ETs) with molecular weights ranging between 470 and 1084 Da were tentatively identified in an ethyl acetate extract of the root of C. hartmannianum based on their retention times and UVλ absorption maxima in HPLC-DAD (Figs. 5 and 7) and their [M-H]- ions resulting from UHPLC/QTOF-MS (Table 4, Figs. 5 and 7). Six other compounds were also identified as ellagitannins based on their UVλ absorption maxima spectra in HPLC-DAD, with three distinct absorption maxima (Fig. 7), although the molecular masses could not be determined for these compounds (Table 4). Among the tentatively identified ellagitannins were corilagin (7), terflavin B (6), and its two isomers (4 and C), castalagin (5), tellimagrandin I (9), a tellimagrandin I derivative detected in the leaves of C. hartmannianum [39], showed a MIC value of 250 µg/ml. 3.3. Antioxidant effects Since the antioxidant capacity of a plant extract could be an important second therapeutic ability in addition to its antimycobacterial effect, we assessed the qualitative antioxidant capacity for a methanol Soxhlet extract, a decoction, and an ethyl acetate extract of the root of Fig. 2. Percentage extraction yields resulting from (A), Sequential extraction and solvent partition extraction and (B), single solvent extraction from the roots, stem bark, and stem wood of Combretum hartmannianum. Red bars = root; blue bars = stem bark; green bars = stem wood. Ethyl acetate (EtOAc); aqueous (aqu, obtained from 80% MeOH); acetone (acet); hot water extracts (HH2O); hexane extracts (hex); dichloromethane extracts (Dic); cold water extracts (H2O*); methanolic Soxhlet extract (MeSox); and cold methanol extracts (Me*); (R), root; (W), stem wood and (B), stem bark. 8 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Fig. 3. Concentration-dependent growth inhibition of a methanolic Soxhlet extract of Combretum hartmannianum root against Mycobacterium smegmatis. Fig. 4. Thin layer chromatograms (RP 18-TLC) of a (1) methanolic Soxhlet extract, an (2) ethyl acetate extract, and a (3) decoction of the root of C. hartmannianum. The same extract is photographed at (A) 254 nm; (B) 366 nm; and (C) after treatment with the DPPH reagent. Rf = retardation factors for the standard compounds. The red braces in 1, 2, and 3 show the ellagitannin-zone (ETs). The dotted line shows the positions of the standard compounds in the extracts. D and F contain eluted standard compounds. (12), α-punicalagin (11), β-punicalagin or terchebulin (13) and (S)flavogallonic acid dilactone (8) (Fig. 5). Besides, gallic acid (1a), an ellagic acid derivative (14), methyl-3,4,5-trihydroxy benzoate (1b), methyl ellagic acid xyloside (18), and epigallocatechin gallate (1 C) were characterized from the root of C. hartmannianum. good agreement with compounds found in our natural compounds library (Agilent Chem Station software), as well as with triterpenes in SciFinder and PubChem databases, and with the literature [90–92]. Among these terpenes, the tetrahydroxylated pentacyclic triterpene, terminolic acid (S) (Syn. 2α,3β,4α,6β)− 2,3,6,23-tetrahydroxyolean-12-en-28-oic acid) was tentatively identified. Terminolic acid is also known as the aglycone of chebuloside II (Fig. 6 G) [93,94]. In addition, the saponin combregenin (J) and the norditerpene furan glycoside, cordifoliside D (Q) were tentatively identified in the root of C. hartmannianum. 3.4.2. Flavonoids Twelve peaks in the C. hartmannianum root ethyl acetate extract HPLC-DAD-chromatograms could be assigned to the group of flavonoids based on their retention times and UVλ absorption spectra, showing two main UV–visible absorption maxima bands; one at 240–285 nm for the B ring and another at 300–400 nm for the A & C rings [82–89]. Moreover, three flavonoids could be tentatively characterized as luteolin (H), isorhamnetin (L), and quercetin-3-O-galactoside-7-O-rhamnoside-(2→1)-O-β-D-arabinopyranoside (F) (Table 4, Fig. 5). 4. Discussion 4.1. Results from this present study on the antimycobacterial effects of extracts of C. hartmannianum in relation to previous studies of African species of Combretum 3.4.3. Triterpenes Our UHPLC/QTOF-MS analysis of the ethyl acetate extract of the C. hartmannianum roots revealed for the first time the presence of three terpenes in this plant (Table 4 and Figs. 5 and 6). Their UV–visible spectroscopic data and retention times (Table 4, Figs. 5 and 7) were in Various traditional medicinal preparations, such as decoctions, macerations, fumigations, tonics, ointments, and pastes of Combretum hartmannianum are used in Sudanese, West African, and Sahelian traditional medicine for the treatment of infectious diseases and 9 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Fig. 5. HPLC-DAD chromatogram at 270 nm of an ethyl acetate extract of the root of Combretum hartmannianum. Compound (1a), Gallic acid; (2), unknown ellagitannin; (3), unknown ellagitannin; (C), Terflavin B isomer I; (4), Terflavin B isomer II; (5), Castalagin; (6), Terflavin B; (7), Corilagin; (8), S-flavogallonic acid dilactone; (9), Tellimagrandin I; (1b), Methyl 3,4,5-trihydroxybenzoate; (10), Di-galloyl glucose; (11), α-Punicalagin; (12), Tellimagrandin I derivative; (13), β-Punicalagin or Terchebulin; (14), Ellagic acid derivative; (15), Tri-galloyl glucose; (16), Tetra-galloyl glucose; (17), ellagic acid derivative; (18), Methyl ellagic acid xyloside; (19), Unknown ellagitannin; (20), gallotannin; (1 C), Epigallocatechin gallate; (F), Quercetin 3-O-galactoside-7-O-rhamnoside – (2→1)-O-D-arabinopyranoside; (H), Luteolin; (J), Combregenin; (L), Isorhamnetin; (Q), Cordifoliside D; (S), Terminolic acid; (A, B, D, E, G, I, K, M, N, O, P, and R), are unknown compounds including unknown flavonoids. Their UV absorption maxima are shown in Fig. 7, Table 4. Fig. 6. Ellagitannins, pentacyclic triterpenes, and a diterpene were tentatively characterized in C. hartmannianum roots in this study. (A), Terchebulin; (B), α-Punicalagin R = H, R1 = OH and β-Punicalagin R = OH and R1 = H; (C), Castalagin, a c-glycosidic ellagitannin with an open chain glucose core; (D), Tellimagrandin I (1-Desgalloyleugeniin); (E), Terflavin B and its triphenyl (flavogallonyl) ester group in red color; (F), Corilagin; (G), Terminolic acid (Chebuloside II); (H), Combregenin; (I), Cordifoliside D. HHDP; hexahydroxydiphenic acid; NHTP, nonahydroxytriphenic acid in blue color. infections on the skin, including symptoms related to tuberculosis, as could be observed both in this present study and by other authors [61, 62,95]. In this present study, we found that in seven villages in Sudan, C. hartmannianum was the most commonly used species of the Combretum spp. included in our study for the treatment of symptoms related to TB. Despite the common use of decoctions, tonics and macerations of C. hartmannianum for bacterial infections and cough [28,61,62], there are only two studies to date that have confirmed that C. hartmannianum possesses antimycobacterial potential; Eldeen and Van Staden [32] showed that ethanol extracts of the leaves of C. hartmannianum gave good growth inhibitory effects against Mycobacterium aurum A+ with a MIC value of 190 µg/ml and Ahmed [13] reported that leaf and seed extracts of C. hartmannianum inhibit the growth of clinical strains of M. tuberculosis. These results are in agreement with our findings that 10 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Table 4 HPLC-DAD and UHPLC/ QTOF- MS data of polyphenolic compounds and derivatives in the root of C. hartmannianum. Combretum hartmannianum Root ethyl acetate extract Molecular formula Rt HPLCDAD Gallic acid (1a) Unknown (A) Unknown (B) Gallotannin Gallotannin Ellagitannin (2) Ellagitannin (3) Terflavin B isomer I (C) Castalagin (5) Terflavin B isomer II (4) Terflavin B (6) Corilagin (7) (S)-Flavogallonic acid dilactone (8) Flavonoid (D) Tellimagrandin I (9) Methyl gallate (Methyl 3,4,5-trihydroxybenzoate) (1b) Gallotannin Di-galloyl-β-D-glucose (10) together with α-Punicalagin (11) Tellimagrandin I derivative (12) Gallotannin β-Punicalagin or Terchebulin (13) Gallotannin Ellagic acid derivative (14) Tri-О-galloyol-β-D-glucose (15) Ellagitannin 1,2,3,6-Tetragalloylglucose (16) Ellagic acid derivative (17) Methyl ellagic acid xyloside (18) Ellagic acid derivative Flavonoid (E) Epigallocatechin gallate (1 C) Gallotannin Quercetin 3-O-galactoside-7-O-rhamnoside – (2→1)-O-D-arabinopyranoside (F) Flavonoid (G) C7H6O5 1.74 4.8 5.3 6.02 6.81 8.30 8.88 8.90 10.87 10.1 11.30 12.10 12.85 13.4 13.6 14.8 Luteolin (H) Flavonoid (I) Combregenin (2α,3β,6β,19α,23pentahydroxyolean-12-en-28-oic acid) (J) Ellagitannin (19) Unknown (K) Isorhamnetin (L) Flavonoid (M) Gallotannin (20) Unknown (N) Unknown flavonoid (O) Unknown (P) Cordifoliside D (Q) Unknown flavonoid (R) Terminolic acid (S) C34H24O22 C41H26O26 C34H24O22 C34H24O22 C27H22O18 C21H10O13 C34H26O22 C8H8O5 C20H20O14 C48H28O30 C34H26O22 C48H28O30 C27H24O18 C34H28O22 C20H16O12 C22H18O11 C32H38O20 14.9 15.1 15.6 15.9 16.5 17.6 17.7 18.6 18.9 19.8 19.9 20.9 21.9 22.0 22.1 24.3 24.3 25.4 Rt UHPLCDAD (min) [M-H]- Exact calculated mass (M+) PPM value UV λ absorbtion max. 216, 272 214, 225 214,225 216, 272 220, 282 214, 260, 380 218, 260, 380 218, 260, 380 215, 259, 377 218, 258, 380 216, 258, 378 216, 260, 382 214, 256, 380 216, 256, 280 216,256,380 216, 280 1.426 169.0147 170.0213 7.05 2.975 3.125 3.541 3.641 3.824 783.0709 933.0640 783.0699 783.0701 633.0725 469.0059 784.0750 934.0702 784.0750 784.0750 634.0798 470.0247 4.71 1.71 3.44 3.44 0.78 – 4.47 3.908 4.157 785.0860 183.0301 786.0906 184.0369 4.07 5.43 4.123 4.424 4.657 483.0801 1083.0574 785.0869 484.0846 1084.0654 786.0906 6.81 – 0.18 5.21 5.473 1083.0600 1084.0654 2.21 5.539 635.0899 636.0954 3.61 6.472 8.316 8.504 787.1030 788.1062 5.83 447.0659 448.0636 -4.44 9.940 457.0768 458.0843 0.65 10.502 741.4083 742.5644 -4.01 28.9 C15H10O6 30.2 12.933 285.0782 286.2070 -5.12 C30H48O7 30.8 31.9 13.050 519.3359 520.6374 -5.75 C16H12O7 C26H34O12 C30H48O6 33.0 33.9 34.9 36.2 36.9 37.1 38.9 39.9 40.6 41.1 43.2 14.982 315.1000 316.2306 -4.91 15.847 537.1568 538.4872 -4.90 16.599 503.3402 504.6384 -5.94 218, 286 216, 278 216, 256, 210, 258, 218, 274 216, 256, 210, 278 210, 254, 216, 278 218, 258, 224, 276 254, 362 254, 368 210, 254, 216, 262, 210, 276 216, 298 218, 258, 340 220, 270, 370 214, 264, 314 216, 280, 218, 260, 216, 256, 210, 230, 218, 274, 218, 278, 216, 298 230, 264, 216, 280, 210, 290 218, 276, 218, 280, 220, 266, Peak area (%) from HPLCDAD 1.1663 1.2229 1.2172 0.1446 0.0448 0.5014 1.6658 6.3051 2.5805 3.4495 7.0732 0.2506 0.2506 0.0372 284, 0.0426 0.9788 14.2385 0.1969 0.4149 0.9735 0.0622 1.5896 1.5234 0.3178 0.3025 2.3194 9.7547 0.5374 1.2071 0.1298 1.1498 1.6271 300, 1.6434 280, 2.9317 314 282 1.1143 3.5554 362 280 300 302 0.9248 0.7443 4.0821 0.9841 1.1489 1.5567 0.9987 1.2079 4.0442 0.3638 1.0830 380 364 278 380 360 366 280 372 314 300 302 370 The exact calculated mass was obtained from the molecular formula and is the sum of the monoisotopic masses and numbers of the atoms in the molecule; Compounds were detected at 270 nm; UVλ absorption maxima were obtained from HPLC-DAD. Peak area % was obtained from an HPLC-DAD chromatogram at 270 nm. methanol and ethyl acetate extracts of the root of C. hartmannianum possess fairly good antimycobacterial effects against M. smegmatis (MIC 312 and 625 µg/ml, respectively) (Table 3). However, although results from our ethnopharmacological survey of the traditional medicinal uses of C. hartmannianum in Sudan indicate that decoctions and macerations are used for the treatment of persistent cough that could be related to TB, we found that these preparations were not as effective as the methanol and ethyl acetate extracts against M. smegmatis (MIC 5000 µg/ml for the decoction) (Tables 1 and 3). Also, the most outstanding growth inhibitory effects in our study were shown by a methanol extract (MIC 312 µg/ml), which could imply that ethanol extracts (that would contain approximately the same composition of compounds as the methanol extract, both qualitatively and quantitatively) of the root could have better potential than decoctions and macerations as a traditional medicinal preparation for the treatment of TB. This view is also supported by the study of Eldeen and Van Staden [32], who found that ethanol extracts of C. hartmannianum were effective inhibitors of the growth of Mycobacterium aurum A+ . Therefore, the results of this present investigation and the results of Eldeen and Van Staden [32] could justify the use of ethanolic tonics of C. hartmannianum for the treatment of persistent cough (Table 1). Our results on the antimycobacterial effects of extracts of C. hartmannianum are in agreement with those of many authors who report that several African species of Combretum possess 11 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 Fig. 7. UVλ-visible absorption maxima spectra of ellagitannins and flavonoids in an ethyl acetate root extract of Combretum hartimannianum. (2), Unknown ellagitannin; (3), Unknown ellagitannin; (4), Terflavin B isomer II; (5), Castalagin; (8), S-flavogallonic acid dilactone; (10), Di-galloyl glucose; (11), α-punicalagin; (12), Tellimagrandin I derivative; (D, E, G, I, M, N, O, P and R), unknown flavonoids; (C), Terflavin B isomer I; (F), Quercetin 3-O-galactoside-7-O-rhamnoside – (2→1)-OD-arabinopyranoside; (H), Luteolin; (J), Combregenin; (L), Isorhamnetin; (S), Terminolic acid (Chebuloside II); (Q), Cordifoliside. antimycobacterial potential. For example, Eldeen and Van Staden [32] reported that ethyl acetate and ethanol extracts of the leaves, root, and bark of C. kraussii showed growth inhibitory effects against Mycobacterium aurum A+ , with MIC values ranging from 196 to 1500 µg/ml. In addition, Asres et al., [66] reported that acetone extracts of the stem bark of C. molle were mildly growth inhibitory against M. tuberculosis (MIC ˃ 1000 µg/ml) and Lall and Meyer [96] showed that an acetone extract of the bark of C. molle gave a MIC value of 500 µg/ml against M. tuberculosis H37Rv. Moreover, Fyhrquist et al. [97] found that stem bark and root extracts of Combretum padoides, C. zeyheri, and C. psidioides had good antimycobacterial potential against M. smegmatis, and a methanol extract of the stem bark of C. psidioides gave the lowest MIC of 625 µg/ml. Acetone extracts of the leaves of C. hereroense and an ethanol extract of the leaves of C. imberbe, gave good antimycobacterial effects (MIC 125 μg/ml and 470 μg/ml, respectively) against M. smegmatis [98, 99]. In addition, alkaloid enriched extracts from leaf extracts of C. zeyheri, C. molle, and C. platypetalum were antimycobacterial against M. smegmatis [100]. Interestingly, Queiroz et al., [101] demonstrated that water decoctions of the leaves, flowers, and stem of C. aculeatum, which are used for tuberculosis in Senegalese traditional medicine, gave good antimycobacterial effects in a host-pathogen assay using Mycobacterium marinum, and Acanthamoeba castellanii. This would imply that decoctions of Combretum aculeatum, and possibly also other Combretum spp., and amongst them C. hartmannianum, could contain compounds useful for the treatment of the more drug-resistant, dormant TB found in infected people without symptoms. different polarities with antioxidant activities, such as ellagitannins, ellagic acid derivatives, flavonoids, and gallic acid (Fig. 4). This would imply that the traditional preparations, such as ethanolic tonics and decoctions would be effective for the treatment of infections also via their antioxidant effects. To the best of our knowledge, there are no previous reports on the antioxidant effects of C. hartmannianum root extracts. However, Taha et al., [25], Mariod et al., [102], and Hassan et al., [103], demonstrated that methanol leaf and stem bark extracts of C. hartmannianum possess good antioxidant effects. Moreover, Hassan et al., [103] attributed the strong DPPH scavenging activity of the leaves of C. hartmannianum mostly to the high flavonoids content of this extract. However, we found that in the root, mainly ellagitannins, including corilagin and punicalagin and ellagic acid derivatives give good antioxidant effects (Fig. 4 C). In agreement with our results, corilagin and punicalagin were found to possess strong antioxidant effects [104,105]. We found that punicalagin was quantitatively the main ET in the root of C. hartmannianum (Table 4, Figs. 5 and 6), and could thus contribute to the antimycobacterial effects of the root extracts by stimulating the immune cells of the host via its antioxidant effects. Moreover, terchebulin and ellagic acid are good DPPH radical scavengers [106]. In our study another ET in the root of C. hartmannianum, which has the same MW as punicalagin, but a longer retention time in HPLC-DAD could be terchebulin, and could thus contribute to the antioxidant effect of the root extract. Interestingly, Seeram et al., [105] found that punicalagin alone was not as antioxidative as pomegranate juice, and therefore they implied that the ellagitannins in the juice potentiate the antioxidant effects of each other. Therefore, similarly, the combination of ETs in the root extracts of C. hartmannianum could be more effective than individual ellagitannins (and other polyphenols) from this extract. This warrants quantitative analysis of the antioxidant capacity of root extracts of C. hartmannianum in relation to that of single 4.2. Antioxidant effects of C. hartmannianum We found that a methanol, an ethyl acetate, and a hot water extract of the root of C. hartmannianum contain a range of compounds of 12 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 ETs separated from this extract. traditional medicinal preparations, such as decoctions and macerations of Combretum spp. are rich in ETs [97]. To date, there are six African species of Combretum that have been studied for their ETs [40,52,66,97, 101,122]. The ETs might have a greater impact than has previously been understood on the medical value of traditional preparations of Combretum spp. [101]. In our study, ten ellagitannins were tentatively characterized from the root of C. hartmannianum for the first time. The characteristic UV absorption with three maxima at 214–220, 254–256, and 370–380 nm revealed that these compounds contain gallic and ellagic acid units (Table 4, Figs. 5, 6, and 7; [123,124]. An ellagitannin at tR 15.6 min in HPLC-DAD was the quantitatively most abundant ellagitannin and is tentatively suggested to be the α-anomer of punicalagin (Table 4, Figs. 5, 6 and 7). Moreover, another ellagitannin at HPLC-DAD tR 17.6 min, with the same molecular mass as punicalagin (1084.0654), is suggested to be either the β-anomer of punicalagin or terchebulin, since both ellagitannins have longer retention times than α-punicalagin, and terchebulin has a mass identical to punicalagin [101,125]. Terchebulin differs from α-punicalagin in containing terchebulic acid, a gallic acid tetramer [126], whereas α-punicalagin contains hexahydroxydiphenic acid [127], (Fig. 6 A and B). Separations of the two anomers of punicalagin by HPLC-DAD have been described by many authors and the differences in their retention times is based on slight differences in the positions of the OH-group and the H-atom linked to the anomeric C1 atom (Fig. 6 B), affecting both the polarity and the UVλmax absorption spectra (Table 4) of these anomers [43,101,128–131]. Punicalagin has been found in some other African Combretum spp., such as C. psidioides and C. zeyheri [97], C. molle [66,122], C. glutinosum [52], and in C. aculeatum [43,101]. Terchebulin was earlier characterized from the stem bark of C. hartmannianum [40]. Moreover, punicalagin and terchebulin were reported from many Terminalia spp. [67,69,75,105,125]. Regarding the potential of ETs as inhibitors of mycobacterial growth, only a couple of studies have been made: For example, punicalagin from the stem bark of C. molle gave mild in vitro growth inhibitory activity against M. tuberculosis typus humanus ATCC 27294 with a MIC > 600 µg/ml [66]. In addition, Queiroz et al., [101], found that the two isomers of punicalagin, α- and β-punicalagin, isolated from a water decoction of Combretum aculeatum, effectively inhibited the growth of Mycobacterium marinum in Acanthamoeba castellanii host cells. In this same investigation, Queiroz et al., [101], also included the punicalagin metabolites, the urolithins, which were not as effective growth inhibitors as punicalagin. However, they concluded that the high consumption of tannins that would be achieved by taking the traditional decoction would lead to plasma levels of the urolithins that would be relevant for their antimycobacterial effects. Therefore, intake of decoctions and ethanolic tonics of C. hartmannianum root, preparations that are used to treat persistent cough in Sudanese traditional medicine, could likewise lead to the same in vivo antimycobacterial effect, as we have found that these preparations are rich in punicalagin. Punicalagin and terchebulin were the principle components in an aqueous ethanolic extract of the root of Terminalia macroptera and were suggested to be responsible for the anti-Helicobacter pylori effects of this extract [132]. In addition, terchebulin from Terminalia laxiflora showed good growth inhibitory effects against Propionibacterium acnes [106]. Recently, punicalagin was shown to give moderate growth inhibitory effects against Staphylococcus aureus (MIC 600 µg/ml) and to inhibit the growth of Escherichia coli as one of the more effective ETs among 23 tested ellagitannins [133]. Besides, punicalagin A (α-) and B (β-) inhibited the growth of Streptococcus spp. [30,130] and MRSA [134–136]. In our study, we could tentatively confirm the presence of the monomeric ellagitannin corilagin ([M-H]- 633.0725) in small quantities in the root of C. hartmannianum (Table 4, Figs. 5 and 6). Previously, corilagin was characterized in one other Combretum spp., C. psidioides [97], in many Terminalia species [125,132,137,138], and Lumnitzera spp. (also a genus within the Combretaceae family) [139]. Corilagin 4.3. Phytoconstituents characterized in this study in a root extract of C. hartmannianum and their possible contributions to its antimycobacterial activity Limited data is available on the phytochemistry of C. hartmannianum, and to the best of our knowledge, no research has been performed on the phytoconstituents of the roots of this species. 4.3.1. Flavonoids Among the 12 flavonoids that were identified in this present study in the root of C. hartmannianum, luteolin (H), isorhamnetin (L), and a quercetin glycoside; quercetin-3-O-galactoside-7-O-rhamnoside-(2→1)O-β-D-arabinopyranoside (F) could be tentatively identified. Luteolin and quercetin-3-O-galactoside-7-O-rhamnoside-(2→1)-O-β-D-arabinopyranoside were not described earlier in C. hartmannianum. Luteolin and isorhamnetin have been found in other Combretum spp. such as the African species, C. dolichopetalum, and C. aculeatum, and the Asian and South-American species, C. latifolium, and C. lanceolatum [42,107–109]. Earlier, Ali et al., [39] studied the polyphenol composition in a leaf extract of C. hartmannianum, resulting in the characterization of 16 flavonoids, of which many were either glycosides or methylated, methoxylated and hydroxylated flavonoids, including methoxylated and glucosylated isorhamnetin derivatives, glycosylated quercetin derivatives, kaempferol ([M+H]+ 287) and its glycosylated and methoxylated derivatives, chrysoeriol ([M+H]+ 301) and its glycosylated derivative, apigenin ([M+H]+ 271), and hydroxylated and methoxylated flavone derivatives. The methoxylated and hydroxylated flavonoids have been reported to be common within the genus Combretum [107]. The flavonoids that we have characterized in the ethyl acetate extract of the root of C. hartmannianum could contribute to the antimycobacterial effect of this extract presented in our study (Table 3, Fig. 4), since flavonoids in general are considered to form part of the plant defense system against pathogenic bacteria [110,111]. However, we found that luteolin and apigenin gave rather high MIC values of 250 µg/ml against M. smegmatis (Table 3). Accordingly also Lechner et al., [112] found that flavonoids gave weak to mild growth inhibitory effects against mycobacteria, with quercetin, isorhamnetin, and luteolin inhibiting the growth of M. smegmatis mc2 155 with MIC values ranging from 128 to 256 µg/ml. Flavonoids could act as antimycobacterial agents via their ability to act as protein chelators and thus affect the function of many important bacterial enzymes and proteins including the aminoglycoside-modifying enzymes (AME:s), efflux pumps, enzymes involved in the respiratory chain as well as enzymes involved in membrane and cell wall synthesis (Fatty acid synthase complex I and II, FASI and II) [113,114]. Moreover, flavonoids could also exert their antimicrobial effects by complexing with the bacterial cell wall, and by disrupting the microbial cell membrane [113–115]. For example, more lipophilic flavonoids, such as apigenin and quercetin are known to disrupt bacterial membranes [116, 117]. Interestingly, some flavonoids, for example, quercetin, are known to act as inhibitors of the mycobacterial proteasome, a viable and relatively newly discovered drug target for new anti-TB drugs [118]. The proteasome is involved in the maintenance of the non-replicating form of mycobacteria, for which there are no available effective conventional drugs [118,119]. Moreover, the structure-activity relationship (SAR) studies have revealed that for the flavonoids the presence of a C-ring, as well as, the number, and positions of free 5-, and 7- hydroxyl groups, and the position, and number of sugar residues are important for their antimycobacterial activity and bioactivity in general [120,121]. 4.3.2. Ellagitannins Ellagitannins (ETs) are generally not well studied within the genus Combretum compared to the genus Terminalia [53,75], although 13 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 consists of one hexahydroxydiphenoyl group (HHDP) connected with a di-ester bond to 3-O and 6-O of D-glucose (Fig. 6 F). According to our result, corilagin gave a weak growth inhibitory effect against M. smegmatis (MIC 1000 µg/ml) as compared to the crude methanol or ethyl acetate extracts showing MIC-values of 312 and 625 µg/ml, respectively (Table 3). Therefore, corilagin and the other ETs in the C. hartmannianum extracts are suggested to act synergistically with each other and other compounds to enhance the antimycobacterial effect of the extract. To the best of our knowledge, corilagin has not been investigated for its effects against mycobacteria previously. Thus, more testing of this and other ETs in C. hartmannianum, alone and in combinations, should be performed against M. tuberculosis, with dormant phenotypes included. Other authors have reported that corilagin possesses mostly weak antibacterial effects. For example, Puljula et al., [133], found that many other ETs, such as castalagin and tellimagrandin I, of which both are present in the root of C. hartmannianum according to our result, were much more antibacterial than corilagin. However, in another study, corilagin gave moderate growth inhibitory effects against S. aureus, E. coli, and C. albicans (MIC 31.25–62.5 µg/ml) and the effect was stronger than for erythromycin against E. coli [140]. Moreover, corilagin was found to affect the membrane permeability of E. coli and C. albicans in a dose-dependent manner, but did not affect S. aureus [140]. Interestingly, corilagin had potentiating effects on conventional antibiotics, and reduced the MIC of various β-lactams by 100- to 2000-fold against MRSA, and worked synergistically together with oxacillin to produce a bactericidal effect [141]. This warrants further research in the antimycobacterial potency of corilagin and its combinations effects with rifampicin and other antibiotics used for the treatment of TB. Corilagin could have a good potential as a new antibacterial (and anti-TB) agent since it is a small enough molecule to be bioavailable and was found to occur in rat and mice blood plasma after oral intake and showed no toxicity in acute toxicity tests [142]. Our results showed that the small molecular hydrolyzable tanninrelated tri-galloyl compound, (S)-flavogallonic acid dilactone ([M-H]469.0059) was present in the root of C. hartmannianum in high concentrations (Table 4; Fig. 5). (S)-flavogallonic acid dilactone was reported to occur also in the stem bark of C. hartmannianum [40]. To the best of our knowledge, no other species of Combretum has been found to contain (S)-flavogallonic acid dilactone. Moreover, a tannin-related to (S)-flavogallonic acid dilactone, that possesses an open ring instead of the lactone ring, (S)-flavogallonic acid (MW 458.32), was found in Terminalia laxiflora and showed good growth inhibitory effects against Propionibacterium acnes [106]. In addition, (S)-flavogallonic acid, isolated from the root of T. sericea, showed broad-spectrum antibacterial activity with MIC values ranging from 110 to 750 µg/ml [143]. We found that the root of C. hartmannianum contains castalagin. To the best of our knowledge castalagin has not been reported to occur in the genus Combretum before. However, castalagin was found to be one of the major ETs in the stem bark of Anogeissus leiocarpa, also a species belonging to the Combretaceae family [123]. Castalagin ([M-H]- ion at m/z 933.0640) is a rare C-glycosidic ET having an open-chain glucose core with the bond configuration of (C-O-C) and shows an S-S stereochemical arrangement of the triphenyl moiety (Fig. 6 C) [144–146]. In a recent study, Araú jo et al., [147] reported that castalagin gave bactericidal effects against methicillin-resistant S. aureus, and affected the assembly of peptidoglycan units in the bacterial cell wall. In agreement with this study, also Puljula et al., [133] found that castalagin effectively inhibits the growth of S. aureus, and was among the most effective ETs in this respect compared to 22 other ETs. Moreover, castalagin was found to suppress the growth of E. coli with a MIC value of 705 µg/ml, as well as to inhibit the growth of Pseudomonas aeruginosa, Clostridia, and Staphylococcus aureus [148,149]. We found that the monomeric ET, tellimagrandin I ([M-H]785.0860) and its derivative were present in the root extract of C. hartmannianum. Tellimagrandin I is a carbon-carbon bond ET (C-C) and has one hexahydroxydiphenyl group and two galloyl groups connected with the glucose core molecule (Fig. 6 D) [144–146]. Tellimagrandin I was found to potentiate the activity of β-lactams against MRSA [150]. Besides, in a large screening of the antimicrobial activities of 23 ETs, tellimagrandin I was among the more antibacterial ETs against S. aureus and E. coli [133]. Tellimagrandin I was earlier characterized in Syzygium aromaticum (Myrtaceae) [151]) and many Terminalia spp. [138]), but has not been reported in a Combretum species before. In this present study, we found that the root of C. hartmannianum contains terflavin B and two isomers. Terflavin B was not reported before in C. hartmannianum. Terflavin B, which is known to be the biosynthetic precursor of punicalagin, has a unique chemical structure, where the triphenyl (flavogallonoyl) ester group is attached to a glucopyranose part (Fig. 6 E) [152,153]. Terflavin B was found in several Terminalia spp. [138]. 4.3.3. Ellagic acid derivatives We found that the roots of C. hartmannianum contain several ellagic acid derivatives (Table 4, Fig. 5), of which methyl ellagic acid-xyloside was present in a high concentration, and could be important for the antimycobacterial activity we have observed for this extract. Besides, we studied the antimycobacterial effect of pure ellagic acid (EA), which had a weak inhibitory effect on the growth of M. smegmatis with a MIC value of 500 µg/ml. However, di-methyl ellagic acid-xyloside and 4-galloyl-dimethyl ellagic acid-xyloside from Terminalia superba were found to be potent growth inhibitors of M. smegmatis and M. tuberculosis, with MIC values of 4.88 µg/ml [154]. Thus, it seems that methylation and glycosylation are important for the antimycobacterial activity of EA derivatives, whereas EA itself is not very active. 4.3.4. Terpenoids We have tentatively identified two oleanolic acid-based pentacyclic triterpenes in the root of C. hartmannianum, namely combregenin (tR HPLC-DAD 31.9 min) and terminolic acid (tR HPLC-DAD 43.2 min) (Table 4, Fig. 6 G). In agreement with many authors, we found that an increase in the polarity of the terpenoids resulted in shortened retention times in reversed-phase HPLC-DAD [155]. Consequently, we found that combregenin, which contains six hydroxyl-groups, was eluted earlier in the HPLC-DAD system than terminolic acid, containing five hydroxyl-groups (Figs. 5 and 6). Terminolic acid is the aglycone of chebuloside II [94]. Terminolic acid and combregenin were identified in the leaves of Combretum zeyheri, the root of C. racemosum, stem bark of C. nigricans, and C. molle, the wood of Terminalia brassii, T. complanata, T. ivorensis, and T. impediens, and bark of T. macroptera [52,74,91,92, 156–160]. To the best of our knowledge, there are no previous reports on the occurrence of combregenin and terminolic acid in the root of C. hartmannianum. Morgan et al., [161], found eight pentacyclic triterpenes in the stem bark extract of C. hartmannianum, including ursolic acid, pomolic acid, corosolic acid, arjunic acid, arjunglycoside 1, trachelosperoside E-1, and combreglucoside. Terminolic acid, isolated from C. racemosum root, was antibacterial against E. faecalis, E. coli, and S. aureus, with MIC values ranging from 64 to 128 μg/ml [157]. Moreover, six triterpenes, including terminolic acid from a Centella asiatica extract were patented due to their antimycobacterial effect against M. avium, the causative agent of paratuberculosis in cattle [161]. In addition, ursolic acid that was also found in Combretum racemosum [157], inhibited the growth of M. tuberculosis H37Ra with a minimum inhibitory concentration of 20 μg/ml [162]. Imberbic acid, isolated from Combretum imberbe, was found to be a powerful inhibitor of the growth of M. fortuitum (MIC 1.56 μg/ml) [163]. Given this, it is possible that the triterpenes, terminolic acid and combregenin, which we have found in the root extract of C. hartmannianum, could contribute to its antimycobacterial effects. In addition to the triterpenes, we tentatively identified the norditerpene glycoside, cordifoliside D in the root of C. hartmannianum (Fig. 5). Although terminolic acid and cordifoliside D have the same 14 E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 number of hydroxyl groups (Fig. 6), cordifoliside D had a shorter retention time in HPLC-DAD compared to terminolic acid due to its glucose sugar residue. Moreover, the shorter retention time could also be due to the presence of a methoxy group in cordifoliside D, which decreases its polarity compared to terminolic acid [155]. To the best of our knowledge, cordifoliside D has not been identified before from the genus of Combretum. Cordifoliside D was originally found in Tinospora cordifolia (Menispermaceae) [164,165]. Moreover, an extract from Tinospora cordifolia was found to inhibit the growth of M. tuberculosis at 1:50000 fold dilutions [166]. Interestingly, the combination of powders obtained from Terminalia chebula, T. bellerica, and Tinospora cordifolia has a long history in Asian and African traditional medicine for the treatment of cough [167–169]. The presence of a glycosidic part and the beta (β)-configurations of the hydroxyl and methyl groups in cordifoliside D (Fig. 6 I), as well as the third carbon atom (C-6) with a γ-hydroxyl substitution that makes it sensitive to steric effects [170], might be important molecular features to impair the function of tyrosine kinase, an important virulence modulator in the mycobacterial cell [171–173]. Conflict of interest statement 5. Conclusion Appendix A. Supporting information In this present study, extracts of various polarities of the stem bark, stem wood, and root of C. hartmannianum showed growth inhibitory activity against Mycobacterium smegmatis. In general, the polar extracts were more active, and this could be due to the high diversity of polyphenols in these extracts. The study can justify the use of C. hartmannianum in Sudanese traditional medicine to cure persistent cough, a symptom that could be related to TB infection. However, the traditional medicinal preparations, decoctions, and macerations, were not as active as a methanol extract of the root that gave the best growth inhibitory effect of the tested extracts. Therefore, this study indicates that for the traditional medicinal use to treat persistent cough (that could be related to TB), ethanolic tonics would have a superior effect over water extracts and decoctions. This study confirmed that C. hartmannianum contains a large number of polyphenolic compounds, with ETs as prominent constituents. Some of the ETs could play a role in the antimycobacterial effects of the C. hartmannianum extracts in which they are suggested to enhance the effects of each other and other compounds. In addition, several flavonoids, gallic acid, and ellagic acid derivatives, including methyl ellagic acid-xyloside, and terpenoids were present in the root of C. hartmannianum. A large number of potentially active antimycobacterial compounds in the root of C. hartmannianum warrant for the further isolation of the compounds and testing against various mycobacterial strains, including clinical strains of M. tuberculosis. Moreover, studies on the impact of extracts and compounds from C. hartmannianum on mycobacterial biofilms could be relevant. Supplementary data associated with this article can be found in the online version at doi:10.1016/j.biopha.2021.112264. The authors declare no conflict of interest. Acknowledgment The first author is grateful for the initial funding of this research provided by the Ministry of Higher Education and the University of Khartoum, Sudan. The financial support provided by the Ekhaga Foundation, Stockholm, Sweden (2017-7) and the Swedish Cultural Foundation in Finland (Grant number 159102, 2020) is greatly acknowledged by the first and the last author. Sincere thanks go to Dr. Haytham Hashim Gibreel and Dr. El-Sheikh Abd Alla El-Sheikh for assisting the first author in plant identification during the fieldwork, as well as to Bashir Abaker, Hateel Al Qasim and Balla Alfadel for their support during the fieldwork. Finally, many thanks to Helsinki University Library (HuLib) in Finland, for funding the article open access processing charge (APC). References [1] World Health Organization (WHO). Tuberculosis and Air Travel: Guidelines for Prevention and Control, third ed., WHO/HTM/TB/2008.399,, Geneva, Switzerland, 2008. [2] E. Cambau, M. Drancourt, Steps towards the discovery of Mycobacterium tuberculosis by Robert Koch, 1882, Clin. Microbiol Infect. 20 (3) (2014) 196–201. [3] T. Kaura, P. Sharmaa, G.K. Guptac, F. Ntie-Kangd, D. Kumara, Treatment of Tuberculosis by natural drugs: a review, Plant Arch. 19 (2) (2019) 2168–2176. [4] I. Smith, Mycobacterium tuberculosis pathogenesis and molecular determinants of virulence, Clin. Microbiol. Rev. 16 (3) (2003) 463–496. [5] W. Liu, J. Zhou, F. Niu, F. Pu, Z. Wang, M. Huang, X. Zhao, L. Yang, P. Tao, P. Xia, J. Feng, Mycobacterium tuberculosis infection increases the number of osteoclasts and inhibits osteoclast apoptosis by regulating TNF-α-mediated osteoclast autophagy, Exp. Ther. Med. 20 (3) (2020) 1889–1898. [6] P.V. Tsouh Fokou, A.K. Nyarko, R. Appiah-Opong, L.R. Tchokouaha Yamthe, M. Ofosuhene, F.F. Boyom, Update on medicinal plants with potency on Mycobacterium ulcerans, Biomed. Res Int. 2015 (2015), 917086, https://doi.org/ 10.1155/2015/917086. [7] World Health Organization (WHO) , 2020. Tuberculosis. https://www.who.int/ news-room/fact-sheets/detail/tuberculosis. [8] C. Dye, K. Floyd, Tuberculosis, in: D.T. Jamison, J.G. Breman, A.R. Measham, G. Alleyne, M. Claeson, D.B. Evans, P. Jha, A. Mills, P. Musgrove (Eds.), Disease Control Priorities in Developing Countries, Oxford University Press, Washington, DC, 2006, pp. 289–309. ISBN-10: 0-8213-6179-1. [9] D.M. Castañeda-Hernández, A.J. Rodriguez-Morales, Epidemiological burden of tuberculosis in developing countries, in: Alfonso J. Rodriguez-Morales (Ed.), Current Topics in Public Health, IntechOpen, 2013, https://doi.org/10.5772/ 53363 (https://www.intechopen.com/books/current-topics-in-public-health/ epidemiological-burden-of-tuberculosis-in-developing-countries). [10] H. Mohajan, Tuberculosis is a fatal disease among some developing countries of the world, Am. J. Infect. Dis. Microbiol. 3 (1) (2015) 18–31. https://mpra.ub. uni-muenchen.de/82851/1/MPRA_paper_82851.pdf. [11] P. Habibi, H. Daniell, C.R. Soccol, M.F. Grossi-de-Sa, The potential of plant systems to break the HIV-TB link, Plant Biotechnol. J. 17 (10) (2019) 1868–1891. [12] S. Russell, The economic burden of illness for households in developing countries: a review of studies focusing on malaria, tuberculosis, and human immunodeficiency virus/acquired immunodeficiency syndrome, Am. J. Trop. Med. Hyg. 71 (2_suppl) (2004) 147–155. [13] Ahmed E.O. Ibnouf, Susceptibility of Mycobacterium tuberculosis isolates to six local medicinal plants. Thesis Submitted to the Faculty of Pharmacy, University of Khartoum, Sudan, 2005. https://khartoumspace.uofk.edu/handle/12345 6789/8207. [14] Centers for Disease Control and Prevention (CDC), Estimates of future global tuberculosis, MMWR Morb. Mortal. Wkly. Rep. 42 (1993) 49. https://pubmed. ncbi.nlm.nih.gov/8246861/. [15] P. Gopal, T. Dick, The new tuberculosis drug Perchlozone® shows cross-resistance with thiacetazone, Int. J. Antimicrob. Agents 45 (4) (2015) 430–433. [16] B. Katende, T.M. Esterhuizen, A. Dippenaar, R.M. Warren, Rifampicin resistant tuberculosis in lesotho: diagnosis, treatment initiation and outcomes, Sci. Rep. 10 (1) (2020) 1–8, https://doi.org/10.1038/s41598-020-58690-4. [17] World Health Organization (WHO) . 2003. Calls On African Governments to Formally Recognize Traditional Medicine. Johannesburg, South Africa. htt Funding This research was funded by the Ekhaga Foundation, project grant 2017-7 (Stockholm, Sweden), the Swedish Cultural Foundation in Finland (2020, Grant number 159102), and the Ministry of Higher Education and University of Khartoum, Sudan at the earlier stage of this research work. Open access was funded by Helsinki University Library (HuLib), Finland. Authors’ contributions E.S. conducted ethnobotanical fieldwork in Sudan in 2006 and 2014. E.S. and P.F. designed the research and wrote the manuscript. E.S. conducted the experiments. E.S., P.F., and R.T. contributed to phytochemical analysis and analytical tools. O.L and M.F. contributed to the writing process. All authors read and approved the manuscript. 15 E.Y.A. Salih et al. [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] Biomedicine & Pharmacotherapy 144 (2021) 112264 ps://www.afro.who.int/news/who-calls-african-governments-formally-reco gnize-traditional-medicine. S.B. Obakiro, A. Kiprop, I. Kowino, E. Kigondu, M.P. Odero, T. Omara, L. Bunalema, Ethnobotany, ethnopharmacology, and phytochemistry of traditional medicinal plants used in the management of symptoms of tuberculosis in East Africa: a systematic review, Trop. Med. Health 48 (1) (2020) 1–21. M.J. Cheesman, A. Ilanko, B. Blonk, I.E. Cock, Developing new antimicrobial therapies: are synergistic combinations of plant extracts/compounds with conventional antibiotics the solution? Pharmacogn. Rev. 11 (22) (2017) 57–72, https://doi.org/10.4103/phrev.phrev_21_17. H. Chandra, P. Bishnoi, A. Yadav, B. Patni, A.P. Mishra, A.R. Nautiyal, Antimicrobial resistance and the alternative resources with special emphasis on plant-based antimicrobials—a review, Plants 6 (2) (2017) 16. M.F. Mahomoodally, Traditional medicines in Africa: an appraisal of ten potent African medicinal plants, Evid. Based Complement. Altern. Med. eCAM 2013 (2013), 617459, https://doi.org/10.1155/2013/617459. K.S. Walter, H.J. Gillett (Eds.), 1997 IUCN Red List of Threatened Plants, IUCN, 1998. H.M. Elamin, Trees and Shrubs of the Sudan, Ithaea Press, Exeter, 1990. H.J. Von Maydell, Trees and Shrubs of the Sahel, Their Characteristics and Uses, GTZ, Germany, 1986. ISBN: 3880853185. E. Taha, A. Mariod, S. Abouelhawa, M. El-Geddawy, M. Sorour, B. Matthäus, Antioxidant activity of extracts from six different Sudanese plant materials, Eur. J. Lipid Sci. Tech. 112 (11) (2010) 1263–1269. E.Y.A. Salih, 2019. Ethnobotany, phytochemistry and antimicrobial activity of Combretum, Terminalia and Anogeissus species (Combretaceae) growing naturally in Sudan. Tropical Forestry Reports-URN:ISSN:0786–8170, 2019. Doctoral thesis submitted to University of Helsink, Finland, 193 pages. http://urn.fi/URN:ISBN:978-951-51-5421-7. Dayamba SD. , 2010. Fire, Plant-derived Smoke and Grazing Effects on Regeneration, Productivity and Diversity of the Sudanian Savanna- woodland Ecosystem. Doctoral Thesis. Acta Universitatis agriculturae Sueciae, p. 73. ISSN 1652–6880: ISBN 978–91-576–7510-1. Print house: SLU Service/Repro, Alnarp 2010. https://pub.epsilon.slu.se/2365/1/Dayamba_S_D_101011.pdf. G.B. El Ghazali, M.S. El Tohami, A.B. El Egami, W.S. Abdalla, M.G. Mohammed, Medicinal plants of the Sudan. Part IV. Medicinal Plants of Northern Kordofan, Omdurman Islamic University Press, Khartoum, Sudan, 1997. H. Ali, G.M. König, S.A. Khalid, A.D. Wright, R. Kaminsky, Evaluation of selected Sudanese medicinal plants for their in vitro activity against hemoflagellates, selected bacteria, HIV-1-RT and tyrosine kinase inhibitory, and for cytotoxicity, J. Ethnopharmacol. 83 (3) (2002) 219–228. A.A. Elegami, E.I. El-Nima, M.E. Tohami, A.K. Muddathir, Antimicrobial activity of some species of the family Combretaceae, Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 16 (6) (2002) 555–561, https://doi. org/10.1002/ptr.995. A.Z. Al Magboul, A.K. Bashir, A.M. Salih, A. Farouk, S.A. Khalid, Antimicrobial activity of certain Sudanese plants used in flokloric medicine: screening for antibacterial activity (V), Fitoterapia 59 (1) (1988) 57–62. I.M.S. Eldeen, J. Van Staden, Antimycobacterial activity of some trees used in South African traditional medicine, S. Afr. J. Bot. 73 (2) (2008) 248–251. A.A. Mariod, N. Fadle, A.A. Hasan, Antimicrobial screening of wood extracts of Combretum hartmannianum, Acacia seyal and Terminalia brownii, Eur. J. Mol. Biol. Biochem 1 (2) (2014) 77–80, e - ISSN - 2348–2206: Print ISSN - 2348–2192. Idris, M. , 2014. Chemical Characterization and Biological Activity of Flavonoids in Some Medicinal Plants. Doctoral thesis submitted to Sudan University of Science and Technology, Graduate College. Khartoum, Sudan. 89 pages. I.M.S. Eldeen, J. Van Staden, In vitro pharmacological investigation of extracts from some trees used in Sudanese traditional medicine, S. Afr. J. Bot. 73 (3) (2007) 435–440. B.O. Burham, 2016. Antimicrobial Activity of Petroleum Ether, Ethyl acetatate and Methanolic Fraction of Five Sudanese Medicinal Plant. Int. J. Sci. Res. 5(4). Paper ID: NOV162454. A.M. Muddathir, T. Mitsunaga, Evaluation of anti-acne activity of selected Sudanese medicinal plants, J. Wood Sci. 59 (1) (2013) 73–79. A.M. Morgan, A.E. Mohamed, C. Saophea, S.U. Park, Y.H. Kim, Pentacyclic triterpenes from the stem bark of Combretum hartmannianum Schweinf, Biochem Syst. Ecol. 77 (2018) 48–50. H.A. Ali, A.A. Hamza, A.E. Abass, O.E. Ahmed, LC/PDA/ESI-MS/MS polyphenols profiling in the in vitro active leaves extracts of Combratum hartmannianum against human pathogens with special emphasis to Madurella mycetomatis, J. Chem. Biol. Phys. Sci. 7 (3) (2017) 757, https://doi.org/10.24214/jcbps. B.7.3.75774. E.A.M. Mohieldin, A.M. Muddathir, T. Mitsunaga, Inhibitory activities of selected Sudanese medicinal plants on Porphyromonas gingivalis and matrix metalloproteinase-9 and isolation of bioactive compounds from Combretum hartmannianum (Schweinf) bark, Evid. Based Complement. Altern. Med 17 (1) (2017) 1–11, https://doi.org/10.1186/s12906-017-1735-y. C. Orwa , A. Mutua , R. Kindt, R. Jamnadass, S. Anthony , 2009. Agrofores tree Database: a tree reference and selection guide version 4.0. http://apps. worldagroforestry.org/treedb2/AFTPDFS/Combretum_aculeatum.PDF. K.M. Hamad, M.M. Sabry, S.H. Elgayed, A.R. El Shabrawy, A.M. El-Fishawy, G.A. A. Jaleel, Anti-inflammatory and phytochemical evaluation of Combretum aculeatum Vent growing in Sudan, J. Ethnopharmacol. 242 (2019), 112052. E.A. Diop, J. Jacquat, N. Drouin, E.F. Queiroz, J.L. Wolfender, T. Diop, J. Schappler, S. Rudaz, Quantitative CE analysis of punicalagin in Combretum [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] [58] [59] [60] [61] [62] [63] [64] [65] [66] [67] [68] [69] 16 aculeatum extracts traditionally used in Senegal for the treatment of tuberculosis, Electrophoresis 40 (21) (2019) 2820–2827. A. Dawé, M. Mbiantcha, F. Yakai, A. Jabeen, M.S. Ali, M. Lateef, B.T. Ngadjui, Flavonoids and triterpenes from Combretum fragrans with anti-inflammatory, antioxidant and antidiabetic potential, Z. Naturforsch. C J. Biosci. 73 (5–6) (2018) 211–219. H.H. Gibreel, A Taxonomic Study on Trees and Shrubs of El Nour Natural Forest Reserve, University of Khartoum, Blue Nile, Sudan, 2008, p. 185 (M. Sc. thesis), https://core.ac.uk/download/pdf/71672105.pdf. D.M.W. Anderson, J.F. Howlett, C.G.A. McNab, Amino acid composition of gum exudates from some african Combretum, Terminalia and Anogeissus species, Phytochemistry 26 (3) (1987) 837–839. S.M. Maregesi, O.D. Ngassapa, L. Pieters, A.J. Vlietinck, Ethnopharmacological Survey of the Bunda district, Tanzania: plants Used to Treat Infectious Diseases, in: J Ethnopharmacol, 113, 2007, pp. 457–470. H.M. , Burkill, 1995. The useful plants of west tropical Africa, Vols. 1–3. The useful plants of west tropical Africa, Vols. 3, Families J-L. ed. 2. Royal Botanic Gardens, Kew. Richmond TW9 3AB United Kingdom. P. Marquardt, R. Seide, C. Vissiennon, A. Schubert, C. Birkemeyer, V. Ahyi, K. Fester, Phytochemical characterization and in vitro anti-inflammatory, antioxidant and antimicrobial activity of Combretum collinum Fresen leaves extracts from Benin, Molecules 25 (2) (2020) 288. G.G. Alowanou, A.P. Olounlade, E.V.B. Azando, V.F.G.N. Dedehou, F.D. Daga, M. Hounzangbe-adote, A review of Bridelia ferruginea, Combretum glutinosum and Mitragina inermis plants used in zoo therapeutic remedies in West Africa: historical origins, current uses and implications for conservation, J. Appl. Biosci. 87 (2015) 8003–8014. Combretum glutinosum Perrot. ex DC., in: H. Vautier, M. Sanon, M. Sacandé, L. Schmidt (Eds.), Seed Leaflet, 2007. A. Jossang, J.L. Pousset, B. Bodo, Combreglutinin, a hydrolyzable tannin from Combretum glutinosum, J. Nat. Prod. 57 (6) (1994) 732–737. A. Dawe, S. Pierre, Habtemariam Phytochemical Constituents of Combretum Loefl, (Combretaceae) Pharm. Crop 438 (2013) 59. T. Yoshida, Y. Amakura, M. Yoshimura, Structural features and biological properties of ellagitannins in some plant families of the order Myrtales, Int. J. Mol. Sci. 11 (1) (2010) 79–106. G.E.B. El Ghazali, M.S. El Tohami, A.A.B. El Egami, Medicinal plants of the Sudan. Part III. Medicinal plants of the White Nile province. National Center for Research, Khartoum, Sudan, Omdurman Islamic University Press, Khartoum, Sudan, 1994. H.H. EL-Kamali, Ethnopharmacology of medicinal plants used in North Kordofan (Western Sudan), Ethnobot. Leafl. 13 (2009) 89–97. N.F. Amako, J.C. Nnaji, GC/MS analysis and antimicrobial activity of Pentacyclic Triterpenoids isolated from Combretum glutinosum Perr. Ex. DC. stem bark, J. Chem. Soc. Niger. 41 (2016) 2. D.M. Anderson, P.C. Bell, Analytical and structural features of the gum exudate from Combretum hartmannianum schweinf, Carbohydr. Res. 49 (1976) 341–349. E.Y.A. Salih, M.T. Kanninen, R. Julkunen-Tiitto, M.H. Sipi, M.O. Luukkanen, H.J. Vuorela, P.J. Fyhrqvist, 2018a. Ellagitannins in antimycobacterial extracts of Combretum hartmannianum, a savannah woodland tree. Conference paper: 66th Congress and Annual Meeting of the Society for Medicinal Plant and Natural Product Research (GA), 24.8–30.8.2018, Shanghai, China. Planta Medica -GA 2018a. 〈https://coms.events/GA2018/data/abstracts/en/abstract_0112.html〉. A.H. Albagouri, A.A. Elegami, W.S. Koko, E.E. Osman, M.M. Dahab, In vitro anticercarial activities of some Sudanese medicinal plants of the family Combretaceae, J. For. Prod. Ind. 3 (2) (2014) 93–99. M.M. Iwu, Handbook of African Medicinal Plants, CRC Press, Boca Raton, Florida, USA, 1993. H. Neuwinger, 1996. African Ethnobotany Poisons and Drugs Chemistry (Germany). Chapman and Hall Gm bH, D-69469 Weinheim (Bundes Republik Deutschland), ISBN 3–8261-0077–8. J.C. Weber, C.S. Montes, T. Abasse, C.R. Sanquetta, D.A. Silva, S. Mayer, G. I. Muniz, R.A. Garcia, Variation in growth, wood density and carbon concentration in five tree and shrub species in Niger, N. For. 49 (1) (2018) 35–51. Combretum nigricans Lepr. ex Guill. & Perr, in: M. Sacandé, M. Sanon, L. Schmidt (Eds.), Seed Leaflet, 2007. D.M.W. Anderson, J.R.A. Millar, W. Weiping, The gum exudate from Combretum nigricans gum, the major source of West African ‘gum combretum’, Food Addit. Contam. 8 (4) (1991) 423–436. K. Asres, F. Bucar, S. Edelsbrunner, T. Kartnig, G. Höger, W. Thiel, Investigations on antimycobacterial activity of some Ethiopian medicinal plants, Phytother. Res. 15 (4) (2001) 323–326. E.Y.A. Salih, M. Kanninen, M. Sipi, O. Luukkanen, R. Hiltunen, H. Vuorela, R. Julkunen-Tiitto, P. Fyhrquist, Tannins, flavonoids and stilbenes in extracts of African savanna woodland trees Terminalia brownii , Terminalia laxiflora and Anogeissus leiocarpus showing promising antibacterial potential, S. Afr. J. Bot. 108 (2017) 370–386. E.Y. Salih, P. Fyhrquist, A. Abdalla, A.Y. Abdelgadir, M. Kanninen, M. Sipi, O. Luukkanen, M.K. Fahmi, M.H. Elamin, H.A. Ali, LC-MS/MS tandem mass spectrometry for analysis of phenolic compounds and pentacyclic triterpenes in antifungal extracts of Terminalia brownii (Fresen), Antibiotics 6 (4) (2017) 37. E.Y. Salih, R. Julkunen-Tiitto, A.M. Lampi, M. Kanninen, O. Luukkanen, M. Sipi, M. Lehtonen, H. Vuorela, P. Fyhrquist, Terminalia laxiflora and Terminalia brownii contain a broad spectrum of antimycobacterial compounds including ellagitannins, ellagic acid derivatives, triterpenes, fatty acids and fatty alcohols, J. Ethnopharmacol. 227 (2018) 82–96. E.Y.A. Salih et al. Biomedicine & Pharmacotherapy 144 (2021) 112264 [96] N. Lall, J.J.M. Meyer, In vitro inhibition of drug-resistant and drug-sensitive strains of Mycobacterium tuberculosis by ethnobotanically selected South African plants, J. Ethnopharmacol. 66 (3) (1999) 347–354. [97] P. Fyhrquist, E.Y. Salih, S. Helenius, I. Laakso, R. Julkunen-Tiitto, HPLC-DAD and UHPLC/QTOF-MS analysis of polyphenols in extracts of the African species Combretum padoides, C. zeyheri and C. psidioides related to their antimycobacterial activity, Antibiotics 9 (8) (2020) 459. [98] P. Masoko, K.M. Nxumalo, Validation of antimycobacterial plants used by traditional healers in three districts of the Limpopo province (South Africa), Evid. Based Complement Altern. Med 2013 (2013), 586247, https://doi.org/10.1155/ 2013/586247. [99] R. Magwenzi, C. Nyakunu, S. Mukanganyama, The Effect of selected Combretum species from Zimbabwe on the growth and drug efflux systems of Mycobacterium aurum and Mycobacterium smegmatis, J. Microb. Biochem. Technol. 3 (2014) 003. [100] T. Nyambuya, R. Mautsa, S. Mukanganyama, Alkaloid extracts from Combretum zeyheri inhibit the growth of Mycobacterium smegmatis, BMC Complement Alter. Med. 17 (1) (2017) 1–11, https://doi.org/10.1186/s12906-017-1636-0. [101] E.F. Queiroz, L. Marcourt, S. Kicka, S. Rudaz, T. Diop, T. Soldati, J.L. Wolfender, Antimycobacterial activity in a single-cell infection assay of ellagitannins from Combretum aculeatum and their bioavailable metabolites, J. Ethnopharmacol. 238 (2019), 111832. [102] A. Mariod, B. Matthäus, I.H. Hussein, Antioxidant activities of extracts from Combretum hartmannianum and Guiera senegalensis on the oxidative stability of sunflower oil, Emir. J. Food Agric. 18 (2006) 20–28, https://doi.org/10.9755/ ejfa.v12i1.5136. [103] L.E.A. Hassan, M.B.K. Ahamed, A.S.A. Majid, H.M. Baharetha, N.S. Muslim, Z. D. Nassar, A.M.A. Majid, Correlation of antiangiogenic, antioxidant and cytotoxic activities of some Sudanese medicinal plants with phenolic and flavonoid contents, BMC Complement Alter. Med. 14 (1) (2014) 1–14, https://doi.org/ 10.1186/1472-6882-14-406. [104] N. Wu, Y. Zu, Y. Fu, Y. Kong, J. Zhao, X. Li, J. Li, M. Wink, T. Efferth, Antioxidant activities and xanthine oxidase inhibitory effects of extracts and main polyphenolic compounds obtained from Geranium sibiricum L, J. Agric. Food Chem. 58 (8) (2010) 4737–4743. [105] N.P. Seeram, L.S. Adams, S.M. Henning, Y. Niu, Y. Zhang, M.G. Nair, D. Heber, In vitro antiproliferative, apoptotic and antioxidant activities of punicalagin, ellagic acid and a total pomegranate tannin extract are enhanced in combination with other polyphenols as found in pomegranate juice, J. Nutr. Biochem. 16 (6) (2005) 360–367. [106] A.M. Muddathir, K. Yamauchi, T. Mitsunaga, Anti-acne activity of tannin-related compounds isolated from Terminalia laxiflora, J. Wood Sci. 59 (5) (2013) 426–431. [107] L.C.J. Araujo, V.C. da Silva, E.L. Dall’Oglio, J. Teixeira de Sousa, Flavonoids from Combretum lanceolatum Pohl, Biochem. Syst. Ecol. 49 (2013) 37–38. [108] M. Chairat, J.B. Bremner, S. Samosorn, W. Sajomsang, W. Chongkraijak, A. Saisara, Effects of additives on the dyeing of cotton yarn with the aqueous extract of Combretum latifolium Blume stems, Color Technol. 131 (4) (2015) 310–315. [109] U.F. Ntite, Determination of antimicrobial potentials of ethanol extract of Combretum dolichopentalum leaves by total dehydrogenase activity assay, Int. J. Pharm. Phytochem. Ethnomed. (2017) 27. [110] M.L. Falcone Ferreyra, S. Rius, P. Casati, Flavonoids: biosynthesis, biological functions, and biotechnological applications, Front. Plant Sci. 3 (2012) 222. [111] S. Kumar, A.K. Pandey, Chemistry and biological activities of flavonoids: an overview, Sci. World J. 2013 (2013) 1–16. [112] D. Lechner, S. Gibbons, F. Bucar, Modulation of isoniazid susceptibility by flavonoids in Mycobacterium, Phytochem. Lett. 1 (2) (2008) 71–75. [113] I. Górniak, R. Bartoszewski, J. Króliczewski, Comprehensive review of antimicrobial activities of plant flavonoids, Phytochem Rev. 18 (1) (2019) 241–272. [114] S. Mickymaray, F.A. Alfaiz, A. Paramasivam, Efficacy and mechanisms of flavonoids against the emerging opportunistic nontuberculous mycobacteria, Antibiotics 9 (8) (2020) 450. [115] G. Gutiérrez-Venegas, J.A. Gómez-Mora, M.A. Meraz-Rodríguez, M.A. FloresSánchez, L.F. Ortiz-Miranda, Effect of flavonoids on antimicrobial activity of microorganisms present in dental plaque, Heliyon 5 (12) (2019) 03013. [116] F.J. Osonga, A. Akgul, R.M. Miller, G.B. Eshun, I. Yazgan, A. Akgul, O.A. Sadik, Antimicrobial activity of a new class of phosphorylated and modified flavonoids, ACS Omega 4 (7) (2019) 12865–12871. [117] A. Adamczak, M. Ożarowski, T.M. Karpiński, Antibacterial activity of some flavonoids and organic acids widely distributed in plants, JCM 9 (1) (2020) 109. [118] Y. Zheng, X. Jiang, F. Gao, J. Song, J. Sun, L. Wang, X. Sun, Z. Lu, H. Zhang, Identification of plant-derived natural products as potential inhibitors of the Mycobacterium tuberculosis proteasome, BMC Complement Altern. Med. 14 (1) (2014) 1–7, https://doi.org/10.1186/1472-6882-14-400. [119] A. Pawar, P. Jha, M. Chopra, U. Chaudhry, D. Saluja, Screening of natural compounds that targets glutamate racemase of Mycobacterium tuberculosis reveals the anti-tubercular potential of flavonoids, Sci. Rep. 10 (1) (2020) 1–12. [120] A. Suksamrarn, A. Chotipong, T. Suavansri, S. Boongird, P. Timsuksai, S. Vimuttipong, A. Chuaynugul, Antimycobacterial activity and cytotoxicity of flavonoids from the flowers of Chromolaena odorata, Arch. Pharm. Res. 27 (5) (2004) 507–511. [121] A. Seyoum, K. Asres, F.K. El-Fiky, Structure–radical scavenging activity relationships of flavonoids, Phytochemistry 67 (18) (2006) 2058–2070. [70] E.Y. Salih, R. Julkunen-Tiitto, O. Luukkanen, M. Sipi, M.K. Fahmi, P.J. Fyhrquist, Potential anti-tuberculosis activity of the extracts and their active components of Anogeissus leiocarpa (Dc.) guill. and perr. with special emphasis on polyphenols, Antibiotics 9 (7) (2020) 364. [71] J. Wang, Y.D. Yue, F. Tang, J. Sun, TLC screening for antioxidant activity of extracts from fifteen bamboo species and identification of antioxidant flavone glycosides from leaves of Bambusa. textilis McClure, Molecules 17 (10) (2012) 12297–12311. [72] N.K. Sethiya, M.M. Raja, S.H. Mishra, Antioxidant markers based TLC-DPPH differentiation on four commercialized botanical sources of Shankhpushpi (A Medhya Rasayana): a preliminary assessment, J. Adv. Pharm. Technol. Res. 4 (1) (2013) 25–30. [73] P. Fyhrquist, I. Laakso, S.G. Marco, R. Julkunen-Tiitto, R. Hiltunen, Antimycobacterial activity of ellagitannin and ellagic acid derivate rich crude extracts and fractions of five selected species of Terminalia used for treatment of infectious diseases in African traditional medicine, S. Afr. J. Bot. 90 (2014) 1–16. [74] J. Conrad, B. Vogler, S. Reeb, I. Klaiber, S. Papajewski, G. Roos, E. Vasquez, M. C. Setzer, W. Kraus, Isoterchebulin and 4, 6-O-isoterchebuloyl-d-glucose, novel hydrolyzable tannins from Terminalia macroptera, J. Nat. Prod. 64 (3) (2001) 294–299. [75] B. Pfundstein, S.K. El Desouky, W.E. Hull, R. Haubner, G. Erben, R.W. Owen, Polyphenolic compounds in the fruits of Egyptian medicinal plants (Terminalia bellerica, Terminalia chebula and Terminalia horrida): characterization, quantitation and determination of antioxidant capacities, Phytochemistry 71 (10) (2010) 1132–1148. [76] K. Taulavuori, R. Julkunen-Tiitto, V. Hyöky, E. Taulavuori, Blue mood for superfood, J. Nat. Prod. Commun. 8 (2013) 1–2. [77] R. Julkunen-Tiitto, S. Sorsa, Testing the effects of drying methods on willow flavonoids, tannins and salicylates, J. Chem. Ecol. 27 (2001) 779–789. [78] P. Singariya, P. Kumar, K.K. Mourya, An activity guided isolation and evaluation of various solvent extracts of the leaves of Anjan grass, Hygeia J. D. Med 4 (2) (2012) 49–56. [79] Clinical and Laboratory Standards Institute , 2012. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Ninth Edition. 950 West Valley Road, Suite 2500, Wayne, Pennsylvania 19087, USA. [80] J.N. Eloff, A proposal on expressing the antibacterial activity of plant extracts- a small first step in applying scientific knowledge to rural primary health care in South Africa, S. Afr. J. Sci. 96 (2000) 116–118. [81] İ. Gülçin, Z. Huyut, M. Elmastaş, H.Y. Aboul-Enein, Radical scavenging and antioxidant activity of tannic acid, Arab. J. Chem. 3 (1) (2010) 43–53. [82] K.R. Markham, T.J. Mabry, Ultraviolet-visible and proton magnetic resonance spectroscopy of flavonoids. In The Flavonoids, Springer, Boston, MA, 1975, pp. 45–77. [83] Z. Jurasekova, J.V. Garcia-Ramos, C. Domingo, S. Sanchez-Cortes, Surfaceenhanced Raman scattering of flavonoids, J. Raman Spectrosc. 37 (11) (2006) 1239–1241. [84] R. Julkunen-Tiitto, N. Nenadis, S. Neugart, M. Robson, G. Agati, J. Vepsäläinen, G. Zipoli, L. Nybakken, B. Winkler, M.A. Jansen, Assessing the response of plant flavonoids to UV radiation: an overview of appropriate techniques, Phytochem. Rev. 14 (2) (2015) 273–297. [85] Q. Dong, Y. Jiang, Q. Wang, M. Liu, N. Wang, H. He, H. Zhou, T. Zhang, X. Luo, 2017, June. Analysis on the polyphenols, flavonoids and antioxidant activities of broccoli. In 2017 Global Conference on Mechanics and Civil Engineering (GCMCE 2017). Atlantis Press. 〈https://www.atlantis-press.com/proceedings/gcmce-17/ 25882885〉. [86] J. Tošović, S. Marković, Reproduction and interpretation of the UV–vis spectra of some flavonoids, Chem. Zvesti 71 (3) (2017) 543–552. [87] T.Y. Wang, Q. Li, K.S. Bi, Bioactive flavonoids in medicinal plants: structure, activity and biological fate, Asian J. Pharm. Sci. 13 (1) (2018) 12–23. [88] R.T. Ramos, I.C. Bezerra, M.R. Ferreira, L.A.L. Soares, Spectrophotometric quantification of flavonoids in herbal material, crude extract, and fractions from leaves of Eugenia uniflora Linn, Pharmacogn. Res. 9 (3) (2017) 253–260. [89] X. Wang, G. Ding, B. Liu, Q. Wang, Flavonoids and antioxidant activity of rare and endangered fern: Isoetes sinensis, Plos One 15 (5) (2020), 0232185. [90] B.T. Schaneberg, J.R. Mikell, E. Bedir, I.A. Khan, V. Nachname, An improved HPLC method for quantitative determination of six triterpenes in Centella asiatica extracts and commercial products, Die Pharm. Int. J. Pharm. Sci. 58 (6) (2003) 381–384. [91] B.K. Ponou, L. Barboni, R.B. Teponno, M. Mbiantcha, T.B. Nguelefack, H.J. Park, K.T. Lee, L.A. Tapondjou, Polyhydroxyoleanane-type triterpenoids from Combretum molle and their anti-inflammatory activity, Phytochem Lett. 1 (4) (2008) 183–187. [92] W.M. Oluyemi, B.B. Samuel, H. Kaehlig, M. Zehl, S. Parapini, S. D’Alessandro, D. Taramelli, L. Krenn, Antiplasmodial activity of triterpenes isolated from the methanolic leaf extract of Combretum racemosum P. Beauv, J. Ethnopharmacol. 247 (2020), 112203. [93] A.P. Kundu, S.B. Mahato, Triterpenoids and their glycosides from Terminalia chebula, Phytochemistry 32 (4) (1993) 999–1002. [94] F.R. Garcez, W.S. Garcez, A.L. Santana, M.M. Alves, M.D.F.C. Matos, A.D. M. Scaliante, Bioactive flavonoids and triterpenes from Terminalia fagifolia (Combretaceae), J. Braz. Chem. Soc. 17 (7) (2006) 1223–1228. [95] Ghazali, G.E.B. El, Medicinal plants of Sudan, Part IV. Medicinal Plants of the White Nile Province. Khartoum, University Press, Khartoum, Sudan, 1998. 17 Biomedicine & Pharmacotherapy 144 (2021) 112264 E.Y.A. Salih et al. [122] K. Asres, F. Bucar, E. Knauder, V. Yardley, H. Kendrick, S.L. Croft, In vitro antiprotozoal activity of extract and compounds from the stem bark of Combretum molle, Phytother. Res. 15 (7) (2001) 613–617. [123] M.N. Shuaibu, K. Pandey, P.A. Wuyep, T. Yanagi, K. Hirayama, A. Ichinose, T. Tanaka, I. Kouno, Castalagin from Anogeissus leiocarpus mediates the killing of Leishmania in vitro, Parasitol. Res. 103 (6) (2008) 1333–1338. [124] J. Moilanen, Ellagitannins in Finnish Plant Species–Characterization, Distribution and Oxidative Activity, Doctoral dissertation submitted to University of Turku, Finland, 2015, p. 112. https://www.utupub.fi/bitstream/handle/10024/105203/ AnnalesAI518Moilanen.pdf?sequence=2&isAllowed=y. [125] O. Silva, E.T. Gomes, J.L. Wolfender, A. Marston, K. Hostettmann, Application of high performance liquid chromatography coupled with ultraviolet spectroscopy and electrospray mass spectrometry to the characterisation of ellagitannins from Terminalia macroptera roots, Pharm. Res 17 (11) (2000) 1396–1401. [126] T.C. Lin, G. Nonaka, I. Nishioka, F.C. Ho, Tannins and related compounds. CII: structures of terchebulin, an ellagitannin having a novel tetraphenylcarboxylic acid (terchebulic acid) moiety, and biogenetically related tannins from Terminalia chebula Retz, Chem. Pharm. Bull. 38 (1990) 3004–3008. [127] S. Wu, L. Tian, Diverse phytochemicals and bioactivities in the ancient fruit and modern functional food pomegranate (Punica granatum), Molecules 22 (10) (2017) 1606. [128] A. Mehta, , 2012. Principle of Reversed-Phase Chromatography HPLC/UPLC (with Animation)’’. PharmaXchange.info. https://pharmaxchange.info/2012/11/thinlayer-chromatography-tlc-principle-with-animation/. [129] L. Feng, Y. Yin, Y. Fang, X. Yang, Quantitative determination of punicalagin and related substances in different parts of pomegranate, Food Anal. Method. 10 (11) (2017) 3600–3606. [130] G. Millo, A. Juntavee, A. Ratanathongkam, N. Nualkaew, J. Peerapattana, S. Chatchiwiwattana, Antibacterial inhibitory effects of Punica granatum gel on cariogenic bacteria: an in vitro study, Int. J. Clin. Pediatr. Dent. 10 (2) (2017) 152–157. [131] V. Sorrenti, C.L. Randazzo, C. Caggia, G. Ballistreri, F.V. Romeo, S. Fabroni, N. Timpanaro, M. Raffaele, L. Vanella, Beneficial effects of pomegranate peel extract and probiotics on pre-adipocyte differentiation, Front Microbiol. 10 (2019) 660. [132] O. Silva, S. Viegas, C. de Mello-Sampayo, M.J.P. Costa, R. Serrano, J. Cabrita, E. T. Gomes, Anti-Helicobacter pylori activity of Terminalia macroptera root, Fitoterapia 83 (5) (2012) 872–876. [133] E. Puljula, G. Walton, M.J. Woodward, M. Karonen, Antimicrobial activities of ellagitannins against Clostridiales perfringens, Escherichia coli, Lactobacillus plantarum and Staphylococcus aureus, Molecules 25 (16) (2020) 3714. [134] S.H. Mun, O.H. Kang, R. Kong, T. Zhou, S.A. Kim, D.W. Shin, D.Y. Kwon, Punicalagin suppresses methicillin resistance of Staphylococcus aureus to oxacillin, J. Pharm. Sci. 137 (4) (2018) 317–323. [135] C. Santiago, E.L. Pang, K.H. Lim, H.S. Loh, K.N. Ting, Inhibition of penicillinbinding protein 2a (PBP2a) in methicillin resistant Staphylococcus aureus (MRSA) by combination of ampicillin and a bioactive fraction from Duabanga grandiflora, BMC Complement Altern. Med 15 (1) (2015) 1–7, https://doi.org/10.1186/ s12906-015-0699-z. [136] P.X. Guo, S. Erickson, D. Anderson, A small viral RNA is required for in vitro packaging of bacteriophage phi 29 DNA, Science 236 (4802) (1987) 690–694. [137] S. Kinoshita, Y. Inoue, S. Nakama, T. Ichiba, Y. Aniya, Antioxidant and hepatoprotective actions of medicinal herb, Terminalia catappa L. from Okinawa Island and its tannin corilagin, Phytomedicine 14 (11) (2007) 755–762. [138] Z. Chang, Q. Zhang, W. Liang, K. Zhou, P. Jian, G. She, L. Zhang, A comprehensive review of the structure elucidation of tannins from terminalia linn, Evid. Based Complement. Altern. Med. eCAM 2019 (2019), 8623909, https://doi.org/ 10.1155/2019/8623909. [139] T.C. Lin, F.L. Hsu, J.T. Cheng, Antihypertensive activity of corilagin and chebulinic acid, tannins from Lumnitzera racemosa, J. Nat. Prod. 56 (4) (1993) 629–632. [140] N. Li, M. Luo, Y.J. Fu, Y.G. Zu, W. Wang, L. Zhang, L.P. Yao, C.J. Zhao, Y. Sun, Effect of corilagin on membrane permeability of Escherichia coli , Staphylococcus aureus and Candida albicans, Phytother. Res. 27 (10) (2013) 1517–1523. [141] M. Shimizu, S. Shiota, T. Mizushima, H. Ito, T. Hatano, T. Yoshida, T. Tsuchiya, Marked potentiation of activity of β-lactams against methicillin-resistant Staphylococcus aureus by corilagin, Antimicrob. Agents Chemother. 45 (11) (2001) 3198–3201. [142] A. Gupta, A.K. Singh, R. Kumar, R. Ganguly, H.K. Rana, P.K. Pandey, G. Sethi, A. Bishayee, A.K. Pandey, Corilagin in cancer: a critical evaluation of anticancer activities and molecular mechanisms, Molecules 24 (18) (2019) 3399. [143] C.P. Anokwuru, S. Tankeu, S. van Vuuren, A. Viljoen, I.D. Ramaite, O. Taglialatela-Scafati, S. Combrinck, Unravelling the antibacterial activity of Terminalia sericea root bark through a metabolomic approach, Molecules 25 (16) (2020) 3683. [144] S. Quideau, T. Varadinova, D. Karagiozova, M. Jourdes, P. Pardon, C. Baudry, P. Genova, T. Diakov, R. Petrova, Main structural and stereochemical aspects of the antiherpetic activity of nonahydroxyterphenoyl-containing C- glycosidic ellagitannins, Chem. Biodivers. 1 (2004) 247–258. [145] S. Quideau, M. Jourdes, D. Lefeuvre, D. Montaudon, C. Saucier, Y. Glories, P. Pardon, P. Pourquier, The chemistry of wine polyphenolic C-glycosidic ellagitannins targeting human topoisomerase II, Chem. Eur. J. 11 (2005) 6503–6513. [146] G.-I. Nonaka, K. Ishimaru, M. Watanabe, I. Nishioka, T. Yamauchi, A.S.C. Wan, Tannins and related compounds. LI. Elucidation of the stereochemistry of the [147] [148] [149] [150] [151] [152] [153] [154] [155] [156] [157] [158] [159] [160] [161] [162] [163] [164] [165] [166] [167] [168] [169] [170] [171] [172] [173] 18 triphenoyl moiety in castalagin and vescalagin, and isolation of 1-O-galloyl castalagin from Eugenia grandis, Chem. Pharm. Bull. 35 (1987) 217–220. A.R. Araú jo, A.C. Araújo, R.L. Reis, R.A. Pires, Vescalagin and castalagin present bactericidal activity toward methicillin-resistant bacteria, ACS Biomater. Sci. Eng. 7 (3) (2021) 1022–1030. M.K. Reddy, S.K. Gupta, M.R. Jacob, S.I. Khan, D. Ferreira, Antioxidant, antimalarial and antimicrobial activities of tannin-rich fractions, ellagitannins and phenolic acids from Punica granatum L, Planta Med. 53 (05) (2007) 461–467. S. Štumpf, G. Hostnik, M. Primožič, M. Leitgeb, J.P. Salminen, U. Bren, The effect of growth medium strength on minimum inhibitory concentrations of tannins and tannin extracts against E. coli, Molecules 25 (12) (2020) 2947. S. Shiota, M. Shimizu, J.I. Sugiyama, Y. Morita, T. Mizushima, T. Tsuchiya, Mechanisms of action of corilagin and tellimagrandin I that remarkably potentiate the activity of β-lactams against methicillin-resistant Staphylococcus aureus, Microbiol. Immunol. 48 (1) (2004) 67–73. M. Toda, J. Kawabata, T. Kasai, α-Glucosidase inhibitors from clove (Syzgium aromaticum ), Biosci. Biotechnol. Biochem. 64 (2) (2000) 294–298. T. Tanaka, G.I. NONAKA, I. NISHIOKA, Tannins and Related Compounds. XLII.: isolation and Characterization of Four New Hydrolyzable Tannins, Terflavins A and B, Tergallagin and Tercatain from the Leaves of Terminalia catappa L, Chem. Pharm. Bull. 34 (3) (1986) 1039–1049. F.J. Mininel, C.S. Leonardo Junior, L.G. Espanha, F.A. Resende, E.A. Varanda, C. Q.F. Leite, W. Vilegas, L.C. Dos Santos, Characterization and quantification of compounds in the hydroalcoholic extract of the leaves from Terminalia catappa Linn. (Combretaceae) and their mutagenic activity, Evid. Based Complement Altern. Med 2014 (2014), 676902, https://doi.org/10.1155/2014/676902. V. Kuete, T.K. Tabopda, B. Ngameni, F. Nana, T.E. Tshikalange, B.T. Ngadjui, Antimycobacterial, antibacterial and antifungal activities of Terminalia superba (Combretaceae), S. Afr. J. Bot. 76 (1) (2010) 125–131. M. Dembek, S. Bocian, Pure water as a mobile phase in liquid chromatography techniques, TrAC Trends Anal. Chem. 123 (2020), 115793. D.K.B. Runyoro, S.K. Srivastava, M.P. Darokar, N.D. Olipa, C.J. Cosam, I.N. M. Mecky, Anticandidiasis agents from a Tanzanian plant, Combretum zeyheri, Med. Chem. Res. 22 (2013) 1258–1262. D.P.A. Gossan, A.A. Magid, P.A. Yao-Kouassi, J. Josse, S.C. Gangloff, H. Morjani, L. Voutquenne-Nazabadioko, Antibacterial and cytotoxic triterpenoids from the roots of Combretum racemosum, Fitoterapia 110 (2016) 89–95. L.A. Collins, S.G. Franzblau, Microplate alamar blue assay versus BACTEC 460 system for high-throughput screening of compounds against Mycobacterium tuberculosis and Mycobacterium avium, Antimicrob Agents Chemother. 41 (5) (1997) 1004–1009. A. Jossang, M. Seuleiman, E. Maidou, B. Bodo, Pentacyclic triterpenes from Combretum nigricans, Phytochemistry 41 (2) (1996) 591–594. S. Roy, D. Gorai, R. Acharya, R. Roy, Combretum (combretaceae): Biological activity and phytochemistry, Am. J. Pharm. 4 (2014) 11. N., Magrone, JR , 2017. Novel Combination of Naturally Occurring Compounds to Assist With Suspected Mycobacterial Infections Related to Autoimmune Conditions. U.S. Patent Application 15/354,975. M.A. Jyoti, T. Zerin, T.H. Kim, T.S. Hwang, W.S. Jang, K.W. Nam, H.Y. Song, In vitro effect of ursolic acid on the inhibition of Mycobacterium tuberculosis and its cell wall mycolic acid, Pulm. Pharm. Ther. 33 (2015) 17–24. D.R. Katerere, A.I. Gray, R.J. Nash, R.D. Waigh, Antimicrobial activity of pentacyclic triterpenes isolated from African Combretaceae, Phytochemistry 63 (1) (2003) 81–88. V.D. Gangan, P. Pradhan, A.T. Sipahimalani, A. Banerji, Norditerpene furan glycosides from Tinospora cordifolia, Phytochemistry 39 (5) (1995) 1139–1142. S.K. Dwivedi, A. Enespa, Tinospora cordifolia with reference to biological and microbial properties, Int. J. Curr. Microbiol Appl. Sci. 5 (6) (2016) 446–465. R. Perumal Samy, P. Gopalakrishnakone, Therapeutic potential of plants as antimicrobials for drug discovery, Evid. Based Complement. Altern. Med. 7 (3) (2010) 283–294, https://doi.org/10.1093/ecam/nen036. K. Spelman, Traditional and clinical use of Tinospora cordifolia , Guduchi, Aust. J. Med. Herbal. 13 (2) (2001). P. Nidhi, P. Swati, R. Krishnamurthy, Indian Tinospora species: natural immunomodulators and therapeutic agents, Int. J. Pharm. Biol. Chem. Sci. 2 (2013) 1–9. M.M. Pandey, S. Rastogi, A.K.S. Rawat, Indian traditional ayurvedic system of medicine and nutritional supplementation, Evid. -Based Complement. Altern. Med. 2013 (2013) 1–12, https://doi.org/10.1155/2013/376327. P. Pradhan, V.D. Gangan, A.T. Sipahimalani, A. Banerji, Stereochemical assignment of tertiary hydroxyl group in diterpene furan glycosides by pyridine induced Shifts, 13C-and 2D NMR spectroscopy, Spectrosc. Lett. 30 (7) (1997) 1467–1474, https://doi.org/10.1080/00387019708006737. K. Chow, D. Ng, R. Stokes, P. Johnson, Protein tyrosine phosphorylation in Mycobacterium tuberculosis, FEMS Microbiol. Lett. 124 (2) (1994) 203–207. K. Grāve, M.D. Bennett, M. Högbom, Structure of Mycobacterium tuberculosis phosphatidylinositol phosphate synthase reveals mechanism of substrate binding and metal catalysis, Commun. Boil. 2 (1) (2019) 1–11. A. Niesteruk, H.R. Jonker, C. Richter, V. Linhard, S. Sreeramulu, H. Schwalbe, The domain architecture of PtkA, the first tyrosine kinase from Mycobacterium tuberculosis , differs from the conventional kinase architecture, J. Biol. Chem. 293 (30) (2018) 11823–11836, https://scifinder.cas.org. (accessed 12 March 2021). https://www.tballiance.org/why-new-tb-drugs/global-pandemic. (accessed 24 August 2020).