Next Article in Journal
Friedelin: Structure, Biosynthesis, Extraction, and Its Potential Health Impact
Next Article in Special Issue
Exploring the Chemical Profile, In Vitro Antioxidant and Anti-Inflammatory Activities of Santolina rosmarinifolia Extracts
Previous Article in Journal
Urea-Functionalized Heterocycles: Structure, Hydrogen Bonding and Applications
Previous Article in Special Issue
Six New Phenolic Glycosides from the Seeds of Moringa oleifera Lam. and Their α-Glucosidase Inhibitory Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Traditional Uses, Pharmacological Activities, and Phytochemical Analysis of Diospyros mespiliformis Hochst. ex. A. DC (Ebenaceae): A Review

by
Thanyani Emelton Ramadwa
* and
Stephen Meddows-Taylor
Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, Florida Campus, University of South Africa, Private Bag X6, Florida 1710, South Africa
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(23), 7759; https://doi.org/10.3390/molecules28237759
Submission received: 18 October 2023 / Revised: 21 November 2023 / Accepted: 22 November 2023 / Published: 24 November 2023

Abstract

:
Diospyros mespiliformis Hochst. ex. A. DC is widely distributed throughout Africa and around the world. It is utilized ethnobotanically to treat fevers, wounds, malaria, diabetes mellitus, and other diseases. This review aims to provide an exhaustive overview of the traditional uses, pharmacology, and phytochemical analysis of D. mespiliformis, with the objective of identifying its therapeutic potential for further research. Scientific resources, including Google Scholar, Science Direct, Web of Science, Pub Med, and Scopus, were used to find pertinent data on D. mespiliformis. Secondary metabolites tentatively identified from this species were primarily terpenoids, naphthoquinones, phenolics, and coumarins. D. mespiliformis has been reported to demonstrate pharmacological activities, including antimicrobial, antiproliferative, antiparasitic, antioxidant, anti-inflammatory, antiviral, anti-hypersensitivity, and antidiabetic properties. The phytochemicals and extracts from D. mespiliformis have been reported to have some pharmacological effects in in vivo studies and were not toxic to the animal models that were utilized. The D. mespiliformis information reported in this review provides researchers with a comprehensive summary of the current research status of this medicinal plant and a guide for further investigation.

1. Introduction

Diospyros mespiliformis Hochsr. ex. A. DC, commonly known as Jackal Berry or African Ebony and musuma in Venda, is a member of the Ebenaceae plant family. Diospyros is a general name that means “divine”, and mespiliformis comes from the Greek words “mesos”, which means “half”, and “pilos”, which means “bullets”. The plant is widely distributed throughout Africa, Asia, and parts of Europe [1]. In Africa, it can be found from as far as east Senegal, and all the way to Eritrea, Ethiopia, and Kenya, as well as in southern Namibia, northern South Africa, and Swaziland [2].
D. mespiliformis is a tall plant, with dense, rounded, and buttressed stems that can reach heights of 15 to 50 m, as shown in Figure 1. It grows in savannahs and woodlands, as well as along riverbanks in areas with regular rainfall, which support natural regeneration. The optimal conditions for the growth of the plant include a mean annual temperature of 16–27 °C, 500–1270 mm of annual rainfall, and an altitude of 350–1250 m [3]. The leaves are simple, alternately arranged, and dark green in color as presented in Figure 2. The plant is dioecious, flowers in April and May, and produces mature fruits in the form of big yellow berries [4]. Its bark is rough in texture and has a dense, evergreen canopy that is black to grey in color. The fruit is a fleshy berry with an enlarged calyx, which is yellow to orange when ripe [5].
Different plant parts have been well documented in ethnobotanical utilization to treat bacterial-, fungal-, viral-, parasitic-, and inflammatory-related ailments, among other things. The leaves are used as a treatment for fevers, as wound dressings, and as an antidote for a variety of poisonous substances. The roots and bark are used to treat diseases such as malaria, syphilis, and leprosy and to stop purging [6]. The bark and roots are also used traditionally for the treatment of diabetes mellitus in Vhembe district, Limpopo province, South Africa. Ground dry bark is mixed in hot water and, in some cases, mixed with Bridelia micrantha (Hochst) Baill bark, as well as Elephantorrhiza elephantina roots. The administered dosage is consumed through drinking a full cup twice daily [7]. D. mespiliformis has been reported to have a variety of pharmacological activities, including antimicrobial, antiproliferative, antiparasitic, antioxidant, anti-inflammatory, antiviral, anti-hypersensitivity, and antidiabetic properties. Preliminary phytochemical screening of D. mespiliformis revealed the presence of carbohydrates, flavonoids, saponins, tannins, phenols, steroids, triterpenoids, anthraquinones, anthocyanins, alkaloids, and cardenolides [8,9,10]. Different bioactive compounds have been tentatively identified in this medicinal plant species. Several studies have been undertaken to isolate bioactive compounds from the leaves, roots, stem, and bark. As a result, several secondary metabolites have been isolated, primarily from chemical constituents, such as terpenoids, naphthoquinones, phenolics, and coumarins [4,11,12,13,14]. No previous study has yet comprehensively reviewed the traditional uses, phytochemistry, pharmacological activities, and in vivo animal investigations of D. mespiliformis. Therefore, the current review aims to comprehensively examine and summarize the previously published articles regarding the traditional medicinal uses, phytochemistry, and pharmacological activities of this plant.

2. Results and Discussion

2.1. Traditional Uses

As shown in Table 1, different parts of D. mespiliformis are traditionally employed in the treatment of a wide range of disorders. Occasionally, various plant parts of the plant are utilized to treat the same conditions. Ringworm infections are treated topically using a decoction of the roots and leaves [15,16,17]. Furthermore, the bark is used in South Africa for ethnoveterinary purposes and for milk production [18]. The bark is boiled and the blend is ingested for stomach problems, such as to stop vomiting or diarrhoea, in Limpopo province, South Africa [19]. A root decoction is used to alleviate febrile symptoms by the Venda people of South Africa [20]. They crush the raw fruit, add a little water, and, thereafter, the infusion is used to treat fungal infections, particularly as a mouthwash or douche, 3× per day [21]. Decoctions of roots or leaves are used as infusions to treat urinary and sexually transmitted infections in Mpumalanga Province, South Africa [22]. In Burkina Faso, a decoction made from the bark and roots is used to alleviate toothache [23]. The roots are crushed and mixed with hot water, and the extraction is ingested to treat abdominal pain in the Midlands Province of Zimbabwe [24]. In Nigeria, a decoction of the root is usually ingested for a week or more to alleviate malaria [25]. In Zimbabwe, a log made of D. mespiliformis and Gardenia spatulifolia is placed at the base of the kraal for animals to leap over each day as a way to manage blackleg illnesses in cattle [26]. The fruits are pound and the juice is mixed with cow milk for drinking to treat dysentery in Benin [27]. A decoction obtained from the leaves or fruits, in association with the leaves of Vitellaria parodoxa, is given orally (0.5 L) and applied topically on the body for the treatment of skin diseases or tonic and zootechnically in veterinary medicine in northern Côte d’Ivoire, particularly for cattle [28]. The bark and roots are boiled in water, and the decoction is ingested to manage pneumonia and syphilis. The leaves are boiled in water and steaming is conducted to manage malaria fever. Pound leaves are rubbed into the skin for the treatment of skin diseases [29].

2.2. Phytochemical Analysis

Chivandi et al. [53] determined the lipid content and profile of oil from D. mespiliformis seeds. The total oil content of the seeds was low at 0.70 ± 0.17%. The lipid component of the seeds contained 39.54% saturated fatty acids and palmitic acid (C16:0), accounting for most of the saturated fatty acids at 30.06 ± 0.61%. The primary monounsaturated fatty acid was palmitoleic acid (C16:1n7), which accounted for 29.37 0.38% of the total monounsaturated fatty acids. Two polyunsaturated fatty acids were identified as linoleic acid (C18:2n6) and linolenic acid (C18:3n3), with levels of 28.71 ± 1.79% and 0.95 ± 0.61%, respectively. Hegazy et al. [54] determined the phytochemical content of the primary metabolites from D. mespiliformis fruits. The highest contents of total protein, hydrolysable carbohydrates, total soluble sugars, and free amino acids were found to be 9.28, 15.88, 9.82, and 2.95%, respectively. In another study by Ebbo et al. [55], the bark had the highest levels of vitamin E (140.91 ± 1.66 mg/dL), but low levels of vitamin A (1275 ± 2.90 mg/dL) and vitamin C (4.8 ± 0.11 mg/dL). The roots have the highest vitamin A concentration (1710 ± 577 mg/dL), as well as the highest levels of vitamin C (23.13 ± 0.43) and vitamin E (129.43 ± 0.42 mg/dL). The leaves had 1366.00 ± 6.88 mg/dL of vitamin A and 25.43 ± 1.18 mg/dL of vitamin C. Adewuyi et al. [56] evaluated the fatty acid composition and lipid profile of D. mespiliformis seed oils from Nigeria. The oils were analyzed for their fatty acid composition, lipid classes, distribution of fatty acids in the lipid fractions, and molecular speciation of the phospholipids, glycolipids, and triacylglycerols. D. mespiliformis was found to have an oil yield of 4.72 ± 0.2%. It was discovered that D. mespiliformis contained 0.84 ± 0.10 g/100 g of C12:0 and 0.82 ± 0.10 g/100 g of C14:0 fatty acids, respectively. Additionally, C18:2 was shown to be the most prevalent fatty acid (34.97 ± 0.40 g/100 g fatty acids). Exactly 60.14 g of unsaturated fatty acids was detected. The outcomes were in line with earlier research on D. mespiliformis fatty acid levels conducted by Chivandi et al. [53]. The neutral lipid content was 93.60 ± 0.20%. C16:1 was exclusively found in the neutral (0.41 ± 0.05 g/100 g fatty acids) and glycolipid (0.36 ± 0.05 g/100 g fatty acids) lipids, but not in the phospholipids.
Petzke et al. [57] conducted research on D. mespiliformis seeds to ascertain their nitrogen and amino acid contents, chemical score, protein-digestibility-corrected amino acid score, accessible lysine, and in vitro digestibility. The seeds contained 5.44% crude nitrogen, 0.87% nitrogen, and 8.99% moisture. Moreover, arginine (501 mg/gn), aspartic acid (507 mg/gn), and glutamic acid (1002 mg/gn) were found to be more abundant in D. mespiliformis seeds than other amino acids. Cysteine and methionine (95%) and tryptophan (75%), respectively, had the highest percentages when it came to the protein-digestibility-corrected amino acid score of important amino acids and in vitro protein digestibility in D. mespiliformis seed samples. The amino acid, fatty acid, and mineral contents of yari, a mixture of lichens that primarily consists of Rimelia reticulate and grows on D. mespiliformis, were examined by Glew et al. [58] in their analysis of plant food in West Africa. When the separate amino acid contents were added together, the estimated protein concentration of D. mespiliformis was 5.31% in yari. The food’s proportions of necessary amino acids did not surpass that of the ideal protein, lysine, in yari (74%). The fatty acid compositions of the plant food are expressed on a dry weight basis. The plant contained less than 1% fatty acid of yari (0.25%). More than 15 mg/g of dry weight calcium was found exclusively in yari, a plant meal used as a condiment. The dry weight of D. mespiliformis exhibited a zinc concentration ranging from 12.1 to 19.0 µg. Achaglinkame et al. [59] determined the nutritional characteristics of wild fruits from D. mespiliformis that are of dietary interest in Ghana. According to the proximate and physicochemical characteristics of the fruits, D. mespiliformis had the maximum moisture content of 6%, but in dry matter, its highest percentage was 93.99%. The fruits’ ash content was 3%, the crude fiber content was 2%, and the pH value was 5.44, meaning that the fruits are quite acidic. The fruits of D. mespiliformis exhibited the highest levels of magnesium (162.98 ± 0.42), potassium (129.4 ± 1.62), and phosphorus (64.78 ± 2.98) in terms of mineral content (mg/100 g dry weight). The fruits of D. mespiliformis had the highest vitamin composition (mg/100 g) of vitamin B3 (310.22 ± 8.15).

2.3. Secondary Metabolites

Maitera et al. [10] evaluated the tannin content accumulated in the unripe fruit, leaves, and bark of D. mespiliformis extracted with acetone, methanol, 70% methanol, and hot- and cold-water extracts. According to the study, unripe fruits had the most tannin content, but the weight of the extracts from 100 g of the powdered material in 70% methanol (15.94 g) and acetone (13.52 g) was much greater. Furthermore, the weight of the leaves after tannin extraction in acetone and 70% methanol extracts was 12.35 g and 11.55 g, respectively, while the weight of the bark after tannin extraction in acetone was 12.33 g. The root bark aqueous extract of D. mespiliformis was quantitatively analyzed by Vandi et al. [8] for the presence of various phytochemicals, including polyphenols (86.58), flavonoids (55.22), tannins (21.71), anthocyanins (10.14), and saponins (21.92) (mEq/100 g of dry).

2.4. Isolated or Tentatively Identified Compounds from D. mespiliformis

According to multiple studies on phytochemistry analyses using various chromatographic and spectroscopic techniques, there are triterpenes such as α-amyrin-baurenol (21), trihydroxy-triterpenoid acid (32), α-amyrin (19), β-sitosterol (29), lupeol (27), betulin (24), and betulinic acid (25) in the stem bark and wood of D. mespiliformis [41,60,61] in addition to naphthoquinones, e.g., diospyrin (1), isodiospyrin (3), diosquinone (2), and plumbagin (12) [62,63], as shown in Table 2. Additionally, Mohamed et al. [14] isolated lupeol (27), betulin (24), betulinic acid (25), and lupenone (26) from the stems and bark of D. mespiliformis. Anas et al. [13] and Adzu et al. [61] have reported the isolation and identification of lupeol (27) from the stem bark of D. mespiliformis. Diosquinone (2) and plumbagin (12) were also isolated from the roots of D. mespiliformis by Lajubutu et al. [64].
Ultra-performance liquid chromatography-electrospray ionization-mass spectrometry was used to tentatively identify several secondary metabolites from the methanol extract of D. mespiliformis, comprising kaempferol (5), myricetin (11), quercetin (13), 4,4′,6,7-tetrahydroxyaurone (17), 8-methoxy-3-methyl-1,2-naphthoquinone (10), and the tetrahydrodiospyrin (40), as shown in Figure 3. Three lupane-type triterpenes (30-hydroxylup-20(29)-en-3β-ol, betulinaldehyde (23), betulinic acid (25)) and betulafolienetriol (22) were also identified alongside δ-tocopherol (18) and the pentagallic acid ester of glucose (39), as shown in Figure 4 [12]. In another study, Dangoggo et al. [4] tentatively identified secondary metabolites from D. mespiliformis leaves using Fourier transform infrared spectroscopy and gas chromatography-mass spectroscopy (GC-MS). Three compounds were identified, namely, 4-hydroxyl-4-methylpentan-2-one (34), octadecanoic acid (35), and 1-octadecyne (37), as presented in Figure 5. David et al. [11] recently conducted GC–MS analysis of dichloromethane fractions from a woody stem methanol extract and highlighted the presence of natural products such as pentadecanoic acid (35), octadecanoic acid methyl ester (36), cis-vaccenic acid (41), β-sitosterol (19), lupeol (27), stigmastan,3,5-diene (31), and a lupeol derivative: 3β-lup-20(30)-en-3-olacetate (28). Following an investigation of D. mespiliformis leaves by Hawas et al. [65], a new acylated flavone isoscutellarein 7-O-(4′′′-O-acetyl)-β-allopyranosyl (1′′′ → 2″)-β-glucopyranoside (4) was isolated and characterized. Furthermore, eight known flavonoid metabolites were identified: luteolin 3′,4′,6,8-tetramethyl ether (9), luteolin 4′-O-β-neohesperidoside (10), luteolin 7-O-β-glucoside (7), luteolin (6), quercetin (13), quercetin 3-O-β-glucoside (14), quercetin 3-O-α-rhamnoside (15), and rutin (16). In addition, their structures were determined via acid hydrolysis of the separated glycosides and via spectroscopic (UV, NMR, and MS) data analyses.
Table 2. Reported isolated or tentatively identified compounds from D. mespiliformis.
Table 2. Reported isolated or tentatively identified compounds from D. mespiliformis.
No.CompoundsPlant PartDetection/Isolation MethodReference
1DiospyrinStem bark or woodIsolated[61,62]
2DiosquinoneStem bark, wood, rootsIsolated[60,61,62]
3IsodiospyrinStem bark or woodIsolated[60,61]
4Isoscutellarein 7-O-(4′′′-O-acetyl)-β-allopyranosyl (1′′′→2″)-β-glucopyranosideLeavesIsolated[66]
5KaempferolStem barkUPLC-ESI-MS[12]
6LuteolinLeavesIsolated[66]
7Luteolin 7-O-β-glucosideLeavesIsolated[66]
8Luteolin 4′-O-β-neohesperidosideLeavesIsolated[66]
9Luteolin 3′,4′,6,8-tetramethyl etherLeavesIsolated[66]
108-methoxy-3-methyl-1,2-naphthoquinoneStem barkUPLC-ESI-MS[12]
11MyricetinStem barkUPLC-ESI-MS[12]
12PlumbaginStem bark, wood, rootsIsolated[60,61,65]
13QuercetinStem bark, leavesIsolated, UPLC-ESI-MS[12,66]
14Quercetin 3-O-β-glucosideLeavesIsolated[66]
15Quercetin 3-O-α-rhamnosideLeavesIsolated[66]
16RutinLeavesIsolated[66]
174,4′,6,7-TetrahydroxyauroneStem barkUPLC-ESI-MS[12]
18δ-TocopherolStem barkUPLC-ESI-MS[12]
19β-AmyrinSeedsGC-MS[54]
20α-AmyrinStem bark or woodIsolated[41,60,61,63]
21α-Amyrin-baurenolStem bark or woodIsolated[41,60,61,63]
22BetulafolienetriolStem barkUPLC-ESI-MS[12]
2330-Hydroxylup-20(29)-en-3β-ol, betulinaldehydeStem barkUPLC-ESI-MS[12]
24BetulinStem bark or woodIsolated, GC-MS[14,41,60,61,63]
25Betulinic acidStem bark or woodIsolated, UPLC-ESI-MS [12,14,41,60,61,63]
26LupenoneStem barkIsolated[14]
27LupeolStem bark or woodIsolated, GC-MS[11,13,14,41,60,61,63,64]
283β-Lup-20(30)-en-3-olacetateWood stemGC-MS[11]
29β-SitosterolStem bark or woodIsolated[11,41,60,61,63]
30γ-SitosterolSeedsGC-MS[54]
31Stigmastan,3,5-dieneWood stemGC-MS[11]
32Trihydroxy-triterpenoid acidStem bark or woodIsolated[41,60,61,63]
33HexadecaneSeedsGC-MS[54]
344-Hydroxyl-4-methylpentan-2-oneLeavesGC-MS[4]
35Octadecanoic acidLeaves, wood stemGC-MS[4,11]
36Octadecanoic acid methyl esterWood stemGC-MS[11]
371-OctadecyneLeavesGC-MS[4]
38OctadieneSeedsGC-MS[54]
39Pentagallic acid ester of glucoseStem barkUPLC-ESI-MS[12]
40TetrahydrodiospyrinStem barkUPLC-ESI-MS[12]
41cis-Vaccenic acidWood stemGC-MS[11]

2.5. Pharmacological Activity

A summary of the pharmacological activities of the different parts and major compounds from D. mespiliformis is described in Table 3.

2.5.1. Antimicrobial Activity

Esimone et al. [5] tested the leaf and root extracts of D. mespiliformis in methanol and water, as well as their combination, for possible antimycobacterial activity against Mycobacterium smegmatis. The methanol leaf and root extracts of D. mespiliformis had minimum inhibitory concentrations (MICs) of 167 µg/mL and 250 µg/mL, respectively. The highest synergistic antimycobacterial activity was shown by the 8:2 ratio of D. mespiliformis and Anthocleista djalonensis against M. smegmatis [5]. Green et al. [50] used tetrazolium microplate tests to determine the MIC of D. mespiliformis hexane leaf extracts against Mycobacterium tuberculosis H37Ra, a clinical strain that was resistant to first-line drugs and one second-line drug. The MIC of the hexane leaf extracts against both M. tuberculosis H37Ra and the clinical isolate was 100 µg/mL.
The antibacterial activity of ethanol extracts of D. mespiliformis was assessed by Dangoggo et al. [4] using the disc diffusion method. The ethanol leaf extracts of D. Mespiliformis inhibited E. coli at concentrations of 90 mg/mL and 120 mg/mL, and P. aeruginosa was inhibited at concentrations of 12 mg/mL and 13 mg/mL. The zone of inhibition for the water extract was 10–13 mm on S. aureus at 30–90 mg/mL and 120 mg/mL, 11–13 mm on P. aeruginosa and 11–14 mm on E. coli at 90–120 mg/mL, and 10–11 mm at 90–120 mg/mL and 120 mg/mL on Shigella spp. D. mespiliformis dichloromethane and methanol crude extracts have been investigated for their antibacterial properties by Mabona et al. [15]. The MIC values of the extracted mixture of dichloromethane and methanol against Propionibacterium acnes ATCC 11827 and Trichophyton mentagrophytes ATCC 9533 were 50 µg/mL and 100 µg/mL, respectively. Shai et al. [66] determined the antibacterial activity of acetone leaf extracts against twenty different bacterial species and D. mespiliformis acetone leaf extracts had an MIC of 80 µg/mL against Bacillus stearothermophilus.
According to a study by Shikwambana and Mahlo [86], the aqueous leaf and bark extracts of D. mespiliformis had excellent antifungal activity against Candida albicans with an MIC of 20 µg/mL and good activity against acetone leaf extracts with an MIC of 80 µg/mL after 48 h. Additionally, D. mespiliformis acetone leaf extracts demonstrated strong antifungal activity against Microsporum canis with an MIC value of 40 µg/mL after 48 h and outstanding antifungal activity against M. canis with MIC values of 20 µg/mL after 24 and 48 h. With regards to Trichophyton rubrum, the aqueous and acetone leaf extracts showed outstanding activity, with MICs of 20 µg/mL after 24 and 48 h. Previous investigations indicated that acetone extracts had an excellent antifungal activity [83]. According to the study conducted by Mamba et al. [33], the MICs of D. mespiliformis 70% ethanol leaf extracts ranged between 3.1 and 6.3 mg/mL against C. albicans ATCC 10231, Gardnerella vaginalis ATCC 14018, Neisseria gonorrhoeae ATCC 19424, and Olivella ureolytica ATCC 43534. Hawas et al. [65] tested the antimicrobial activity of the flavonoids that were isolated from D. mespiliformis leaves against four human pathogenic bacteria. Flavonol O-rhamnoside (15) had moderate activity against S. aureus, with an MIC value of 9.77 μg/mL, while methylated flavone showed strong action against E. coli, with an inhibition zone of 34 mm. The minimal bactericidal concertation (MBC)/MIC ratio was used to assess the antibacterial activity of the isolated flavonoids. Furthermore, the study discovered that flavonoids were bactericidal against S. aureus and that flavonoids were bactericidal against E. coli. Lajubutu et al. [64] tested the antibacterial activity of diosquinone (2) and plumbagin (12) that were isolated from the roots of D. mespiliformis [64]. The diosquinone (2) MICs ranged from 3 to 30 μg/mL for S. aureus NCTC 6571 and S. aureus E3T, while they were 15 to 16 μg/mL for E. coli KL16 and P. aeruginosa NCTC 6750. Furthermore, S. aureus NCTC 6571 responded paradoxically and biphasically to diosquinone (2) in nutritional broth, yet its bacterial activity against E. coli KL16 increased as the concentration rose to the maximum diosquinone (2) concentration measured.

2.5.2. Anti-Inflammatory Activity

Adzu et al. [61] evaluated the in vivo antipyretic, analgesic, and anti-inflammatory effects of D. mespiliformis methanol stem bark extracts in rats and mice. The extract demonstrated significant efficacy (PB/0.05) against all analgesic and anti-inflammatory models applied at 100 mg/kg and had an antipyretic effect at 50 and 100 mg/kg i.p. According to the findings, the extract’s LD50 in mice was 513.809 ± 33.92 mg/kg i.p. In a different investigation, Adzu et al. [80] extracted D. mespiliformis stem bark progressively using hexane, chloroform, and methanol, and then performed preliminary analgesic action on the extract. The most active of the three extracts was the chloroform extract, which was also subjected to column chromatography, resulting in the isolation of lupeol (27). In rats, lupeol (27) reduced the pain stimulus brought on by the analgesic meter and formalin. It has been found that lupeol (27) functions either individually or together with different compounds, and it may have been responsible for the plant’s benefits in the treatment of pain-related disorders.
Mamba et al. [33] evaluated the anti-inflammatory properties of D. mespiliformis root extracts using the 15-lipoxygenase (15-LOX) model of inhibition. The IC50 value for the D. mespiliformis root extract’s anti-inflammatory effects was only 188.1 µg/mL. In a different study, Lawal et al. [72] revealed that D. mespiliformis had a modest activity against xanthine oxidase (XO), with an IC50 value of 142 8 µg/mL, but had an inhibitory effect against 15-LOX at the highest tested dose. At the maximum measured concentration of 100 µg/mL, D. mespiliformis extracts suppressed the formation of nitric oxide (NO) by around 68.1%, which is just under the 70% threshold. The immunomodulatory effects of solvent fractions of D. mespiliformis were investigated by David et al. [11] in mice infected with a Plasmodium berghei (NK 65)-sensitive strain. Compared to the pharmacological control, the levels of IgG, IgM, and tumor necrosis factor alpha (TNF) were considerably greater in the dichloromethane fraction group, although the interleukin 1 beta (IL-1β) and interleukin 6 (IL-6) values did not change proportionally with the dose. The study established that the dichloromethane fraction had immunomodulatory effects on infected mice.
Ebbo et al. [55] highlighted the D. mespiliformis crude bark, leaf, and root methanol extracts’ wound-healing capabilities using in vivo animal models. After 11 days of treatment with the crude methanol extracts of the bark and roots of D. mespiliformis, the rats’ dorso-caudal lesions were healed. Exactly 13 days after the initial wound, the wound was healed in the same amount of time as rats given penicillin and a leaf extract of D. mespiliformis. The lesions on the rats treated with carboxyl methyl cellulose were healed exactly 15 days after it was administered. Swelling and reddening were noticeable in the bark, leaf, and root treatment groups throughout the first five days of the trial. On day 9, the wounds of rats fed with D. mespiliformis bark fractions in ethyl acetate and hexane had fully healed. Animals given butanol and water fractions did not statistically differ (p > 0.05) from groups given ethyl acetate and hexane extracts. On the eleventh day following wounding, complete wound closure was attained in all groups for the D. mespiliformis leaves. On day 9, the water-fraction-treated group had the largest incision length, measuring 3.5 ± 0.29 mm. This was statistically (p < 0.05) greater than the incision lengths for the hexane- and ethyl-acetate-treated groups, which were 1.0 mm on the experiment day. In the same experiment, Ebbo et al. [55] investigated the in vitro anti-inflammatory activity of various D. mespiliformis fractions against the LOX-15 enzyme. The hexane fraction showed the maximum percentage of inhibition at 10 mg/mL and 5 mg/mL, with values of 32.05 ± 2.79 and 31.21 ± 0.84, respectively. The water component was inhibited by 19.67 ± 2.29 percent at a dosage of 10 mg/mL compared to zero at a concentration of 5 mg/mL. The butanol and ethyl acetate fractions at 5 and 10 mg/mL appear to activate the enzyme.

2.5.3. Antiparasitic Activity

Aderbauer et al. [67] examined the in vitro antitrypanosomal activity of dichloromethane leaf extracts from D. mespiliformis against Trypanosoma brucei in a long-term viability assay. The crude extract had a poor MIC of only 500 µg/mL against T. brucei, which is higher than the criterion for pharmacological significance of 100 µg/mL. Nafuka [73] evaluated the in vitro antiplasmodial effectiveness of methanol and aqueous extracts of D. mespiliformis (leaf and root) against Plasmodium falciparum. After P. falciparum 3D7A had been treated with crude methanol leaf extracts from D. mespiliformis for 24 and 48 h, the average percentage of parasitemia decreased across all concentrations; however, this was not statistically significant (p = 0.3 and 0.5, respectively). There was only time-dependent antiplasmodial activity at 5 µg/mL and 24 h efficacy at 10 µg/mL. The average percentage of parasitemia for the aqueous extract did not decrease statistically significantly at 24 h (p = 0.6) or 48 h (p = 0.1). At 24 h as opposed to 48 h, the leaf extract was more effective against P. falciparum 3D7A. At 24 h, the leaf extract was more effective against P. falciparum 3D7A than at 48 h. D. mespiliformis aqueous root extracts had an IC50 of 2.91 µg/mL and leaf extracts had an IC50 of 3.01 µg/mL for aqueous extracts. The D. mespiliformis methanol leaf extracts were the most effective, with an IC50 of 1.51 µg/mL, while the methanol root extracts also had good activity at 2.12 µg/mL [86].
The in vitro antiplasmodial activity of root extracts from D. mespiliformis was evaluated by Bapela et al. [20]. The extractant utilized on the powdered root material was dichloromethane: 50% methanol (1:1). Antiplasmodial activity against the chloroquine-sensitive strain of Plasmodium falciparum (NF54) was investigated. Polar extracts from the roots of D. mespiliformis prevented the growth of plasmodial cells (IC50 = 28.4 µg/mL) and showed significant dichloromethane action (IC50 = 4.40 µg/mL). The in vitro antiprotozoal activity of D. mespiliformis leaf extracts in 70% ethanol was discovered by Traore et al. [69]. The Trypanosoma brucei brucei IC50 values for D. mespiliformis were 25.8 µg/mL for Trypanosoma cruzi, >64 µg/mL for Leishmania infantum, and 24.9 µg/mL for Plasmodium falciparum. The effect of a crude ethanolic extract of D. mespiliformis on the clinicopathological variables of Yankasa sheep experimentally infected with Haemonchus contortus was determined by Luka et al. [25]. Throughout the duration of the trial, L3 H. contortus larvae infection of Yankasa sheep did not result in statistically significant alterations (p > 0.05) in the mean rectal temperature. The extract showed some effectiveness against H. contortus at the tested dosages. Bapela et al. [76] evaluated the inhibitory effects of D. mespiliformis dichloromethane (DCM) and 50% MeOH root extracts against axenically grown amastigote forms of Leishmania donovani (MHOM-ET-67/L82). A considerable antileishmanial effect was shown by the D. mespiliformis DCM root extract, with an IC50 of 7.7 µg/mL, while some antileishmanial effects were observed for 50% MeOH root extracts, with an IC50 of 54 µg/mL.
The in vivo antimalarial effects of bark extracts of D. mespiliformis were examined by Chinwe et al. [81] in adult Swiss albino mice that had been infected with a chloroquine-resistant NK65 lineage of Plasmodium berghei. Bark extracts of D. mespiliformis demonstrated a more substantial antimalaria efficacy, with an inhibition percentage of 53% at a dose of 800 mg/kg. Agbadoronye et al. [77] investigated the antitrypanosomal activities of D. mespiliformis leaf extracts and an alkaloidal fraction in Trypanosoma evansi-infected rats. White blood cells, the packed cell volume, the mean corpuscular hemoglobin, the mean corpuscular hemoglobin concentration, and crude extracts at 400 mg/kg BW and 100 and 200 mg/kg BW substantially (p < 0.05) increased the red blood cells and elevated bilirubin levels while decreasing the crude extract. Furthermore, the extract considerably reduced the total proteins. The effectiveness of D. mespiliformis against chloroquine-sensitive and -resistant strains of malarial parasites in mice was examined by Olanlokun et al. [12]. At 400 mg/kg, D. mespiliformis decreased mean percentage parasitemia values (5 ± 1), increased the packed cell volume (36% ± 1.4), and increased platelets (2 ± 1.4). At the same dose, D. mespiliformis decreased the activities of alkaline phosphatase (56 ± 0.7 U/L), alanine aminotransferases (6.2 ± 0.8 U/L), and alanine aminotransferases (8 ± 3.8 U/L). D. mespiliformis reversed the start of the permeability transition while decreasing ATPase enhancement and lipid peroxidation. Although D. mespiliformis was effectively tolerated at the maximal dose in the infected control group, liver histology in that group showed severe widespread congestion and wide hemorrhagic lesions. In an additional investigation carried out by Olanlokun et al. [78], the antiplasmodial effects of D. mespiliformis root extracts were investigated in Plasmodium berghei-infected mice. According to the results, D. mespiliformis had a high rate of parasite clearance (84.7%) and a lower proportion of parasitemia (0.67%). The fractions and extracts of D. mespiliformis considerably reduced the production of β-hematin due to their cell-free antiplasmodial activity. David et al. [11] carried out a study to evaluate the bioactivity-guided antiplasmodial efficacy of solvent fractions of D. mespiliformis in mice infected with a susceptible strain of Plasmodium berghei (NK 65). The crude methanol extract of the stems of D. mespiliformis was partitioned between n-hexane, dichloromethane, ethyl acetate, and methanol. The dichloromethane fraction had the highest parasite clearance and improved hematological indices relative to the drug control. The heme values increased, while the hemozoin content significantly (p < 0.05) decreased. The highest dose of n-hexane and methanol opened the mitochondrial permeability transition (mPT) pore, while the reversal effects of dichloromethane on the mPT, mitochondrial F1F0 ATPase, and lipid peroxidation were dose-dependent.

2.5.4. Antidiabetic Activity

Mohamed et al. [14] studied the α-glucosidase enzyme inhibition activity of isolated bioactive compounds from D. mespiliformis. Lupeol, botulin, and lupenone had an α-glucosidase inhibitory activity, with an IC50 ranging from 0.002 to 0.46 mM.

2.5.5. Antiviral Activity

D. mespiliformis root extracts were tested for anti-HIV activity against recombinant HIV-1 enzyme by Mamba et al. [33] using a non-radioactive HIV-RT colorimetric assay. With a 17.4% inhibition of the HIV-1 RT, the root extracts of D. mespiliformis demonstrated a poor inhibitory efficacy. Similar to the findings of Hedimbi [68], who demonstrated that D. mespiliformis leaf extracts at 0.1 mg/mL had 78.7% HIV-1 RT activity, it was confirmed that extracts of D. mespiliformis have varying degrees of activity against HIV-1 RT. Chukwuma [70] investigated the antiviral activities of D. mespiliformis aqueous, ethanolic, and methanolic extracts on the avian viruses Newcastle disease virus (NDV), fowl pox virus (FPV), and infectious bursal disease virus (IBDV). Aqueous extracts of D. mespiliformis at 400 mg/mL, 200 mg/mL, and 100 mg/mL, respectively, inhibited the virus (NDV) in percentages of 91%, 86%, and 85%. The percentage inhibition of the ethanolic extracts was 95%, 90.5% and 89%, respectively. At a concentration of 400 mg/mL of the crude extract of D. mespiliformis, the tested FPV showed an extremely high activity. All plant extracts were shown to have 100% egg mortality at the end of the experiment with the infectious bursal disease virus (IBDV).

2.5.6. Anti-Hypersensitivity

Belemtougri et al. [71] tested the efficacy of the crude, aqueous, and ethanolic extracts of D. mespiliformis to inhibit the effects of caffeine on the release of calcium from the sarcoplasmic reticulum of rat skeletal muscle cells. Different D. mespiliformis extracts failed to function in rat skeletal muscle cells when applied alone, demonstrating that they are unable to change the resting calcium levels of skeletal muscle cells. When caffeine (10 mmol/L) was given to myotubes, Ca2+ was released from the SR. The reaction was used as a control, and each cell was given a 10 mmol/L caffeine solution. Then, utilizing caffeine and plant extracts, a second cell was investigated. The crude extract of D. mespiliformis at a concentration of 10 mg/mL reduced the amplitude of Ca2+ release from the SR. This proved that these extracts significantly restrict the release of Ca2+, which is sensitive to caffeine from the SR. The suppression of intracellular calcium release by various extracts was dose-dependent, with crude decoctions being the most effective. The following are some categories for the effects of several D. mespiliformis extracts at 10 mg/mL: The aqueous extract follows the ethanolic extract after the crude decoction. The crude decoction contains 51% D. mespiliformis, with an IC50 of 8.84 mg/mL at the same dose. However, the IC50 of the D. mespiliformis ethanolic extract was 9.23 mg/mL, whereas the IC50 of the other extracts exceeded 10 mg/mL. Calcium release from the sarcoplasmic reticulum was inhibited in ethanolic extracts by 54% and in aqueous extracts by 29% in 10 mg/mL crude D. mespiliformis decoctions.

2.5.7. Antioxidant Activity

Ndhlala et al. [85] investigated the methanol extracts of D. mespiliformis wild fruits and analyzed them for their scavenging effect of the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical, reducing power, and anion radical effect on superoxide anions using a colorimetric method. There was an increase in the radical-scavenging effect, reducing power, and superoxide-anion-radical-scavenging effect as the concentration of the sample increased. D. mespiliformis had a high DPPH-radical-scavenging capacity. According to Sombie et al. [87], the leaf extracts of D. mespiliformis exhibited a TEAC of 1.170 ± 00 mM (ABTS) (p < 0.05), which was a significantly higher antioxidant capacity than Trolox (1 mM). Extracts of D. mespiliformis (1.17 ± 0.00 mM TEAC/g and 70.77 ± 0.4 M ET/g) showed the greatest antioxidant activity. The acetone leaf extracts of D. mespiliformis were also tested for their radical scavenging potential and had an IC50 of 25 ± 2 μg/mL in a DPPH assay [80]. D. mespiliformis fruit extracts were studied for their ability to scavenge DPPH by Hegazy et al. [54]. D. mespiliformis displayed a greater level of DPPH-radical-scavenging activity (87.36%) at a concentration of 1 mg/mL of the radical. A higher hydrogen peroxide scavenging activity than 85% inhibition was demonstrated by the D. mespiliformis extracts at methanol concentrations of 1 mg/mL. The antioxidant capacity of the powdered D. mespiliformis fruit was investigated in relation to the effects of solvent extractions (ethanolic and hydroethanolic extracts) [84]. The IC50 values of DPPH-radical-scavenging activity were found to be 1.037 ± 0.204 mg/mL for ethanolic extracts and 1.111 ± 0.133 mg/mL for hydroethanolic extracts. To evaluate the in vivo antioxidant activity against high-fat diet (HFD)-induced hyperlipidemia in rats and different particle size powder fractions, ethanolic and hydroethanolic extracts of D. mespiliformis fruits were administered orally (600 mg/kg, p.o.) for 30 days with a HFD, and the effect of the extracts on enzymatic antioxidants like superoxide dismutase (SOD), catalase (CAT), and peroxidase was estimated in the blood, heart, liver, and kidneys [78]. In comparison to the control group, various samples of D. mespiliformis fruit powder considerably increased the levels of SOD, catalase, peroxidase, alanine transaminase, and aspartate aminotransferase enzymes.
In vitro antioxidant activities of root bark aqueous extracts of D. mespiliformis (ABTS, DPPH and FRAP) were determined [8]. The ABTS radical’s inhibitory concentration of 50% (IC50) was 220 µg/mL, and the root bark aqueous extract had the ability to scavenge ABTS and DPPH radicals as well as reduce FRAP. Ebbo et al. [55] showed the DPPH-radical-scavenging properties of crude methanol extracts and fractions of D. mespiliformis leaves, bark, and roots. The crude methanol extracts of the leaves, bark, and roots of D. mespiliformis had IC50 values of 6.94 ± 0.49 µg/mL, 7.82 ± 0.76 µg/mL, and 3.47 ± 0.05 µg/mL, respectively. The ethyl acetate fraction showed the lowest IC50 (1.08 ± 0.04 µg/mL) and the greatest antioxidant activity. Antioxidant activity was observed in both the water and butanol fractions, with IC50 values of 4.73 µg/mL and 1.44 µg/mL, respectively. Hawas et al. [65] evaluated the antioxidant activity of D. mespiliformis secondary metabolites using a DPPH radical-scavenging assay. The new acylated flavone (8) and flavonol O-rhamnoside (15) demonstrated the most potent antioxidant activity, with IC50 values of 15.46 μg/mL and 12.32 μg/mL, respectively.

2.5.8. Antiproliferative Activity

Adeniyi et al. [36] assessed the cytotoxicity activity of diosquinone (17) previously isolated from the root bark of D. mespiliformis against ten cancer cell lines (human breast (BC-1), colon (COL-2), human fibrosarcoma (HT-1080), human lung cancer (LU-1), human nasopharyngeal carcinoma (KB), oral epidermoid carcinoma (KB) and KBV1, prostrate (LNCaP), human glioblastoma cells (U373), human neuroblastoma (SKNSH), multiple-drug-resistant or vinblastine-resistant human nasopharyngeal carcinoma (KB-V(V-VLB))). Diosquinone (2) was more active against human glioblastoma with an ED50 of 0.18 μg/mL. It is interesting to note that naphthoquinone epoxide significantly inhibits BC-1, HT-1080, Lu-1, KB, and SKNSH with the same ED50 of 0.2 µg/mL. It is noteworthy that diosquinone has an excellent cytotoxicity capability against vinblastine or multiple-drug-resistant human nasopharyngeal cancer (KB-V(V-VLB)), with an ED50 range of 1–1.7 µg/mL.
Aderbauer et al. [67] investigated the cytotoxicity of D. mespiliformis dichloromethane leaf extracts against fibroblast-like mammalian cells. The findings were summarized as the minimum toxic concentration (MTC), which is the concentration at which fibroblast damage in the form of morphological change or ablation could be seen under a microscope. The leaf extract was not toxic at the tested MTC values of more than 500 µg/mL. D. mespiliformis 70% ethanol leaf extract was tested for in vitro cytotoxicity against MRC-5 fibroblasts and showed a modest toxicity, with an IC50 of >64 µg/mL [69]. Adoum [79] investigated the cytotoxic effects of D. mespiliformis ethanol extracts, which were reported as IC50 values in μg/mL. Extracts prepared from the plant were solvent partitioned and screened for activity in the brine shrimp (Artemia cysts) lethality test (BST). D. mespiliformis showed a very low brine shrimp lethality at LC50 > 1000 μg/mL. By growing rat skeletal myoblast L6 cells in the presence of D. mespiliformis dichloromethane (DCM) and 50% MeOH roots extracts, spanning a concentration range of 0.002 to 100 µg/mL, Bapela et al. [76] evaluated the in vitro inhibition of mammalian cell proliferation. The DCM extract showed a level of toxicity against the test cell line, with an IC50 of 24.3 µg/mL and 60.4 µg/mL in the 50% MeOH extract. The toxic properties of several root bark extracts of D. mespiliformis were examined by Mustapha et al. [9] utilizing brine shrimp cytotoxicity. The lethality test of the D. mespiliformis root bark extracts was evaluated using brine shrimp (Artemia salina) nauplii as the test organism. The n-hexane extract had the highest lethal dose concentration of 8203.52 μg/mL compared to the water extract, which was 100% safe at the investigated concentrations.

2.5.9. In Vivo Studies

The neuropharmacological effects of the aqueous extract of D. mespiliformis stem bark were examined in mice by Adzu et al. [80]. The extracts (100 and 200 mg/kg p.o.) significantly (PB/0.05) increased the duration of pentobarbital-induced sleep and decreased exploratory and spontaneous motor behavior. However, the extract barely protected mice from death brought on by pentylenetetrazole, and only protected against the commencement of stages of seizure activity. Additionally, it had no impact on the motor coordination test. The effects of sub-chronic treatment with crude D. mespiliformis root extracts on a few biochemical markers in mice were examined by Jigam et al. [79]. There was minimal variation in the packed cell volumes or overall body weights of the animals given the extracts. In relation to some organ weights, triacyglycerides (148.25 ± 2.78 mg/dL) and alkaline phosphatase (41.50 ± 1.71 mg/dL) were not statistically significant (p > 0.05). However, there were significant (p > 0.05) differences between the animals treated with the extracts and controls in the heart (0.74%), the lungs (4.43%), glucose (113.92 ± 2.43 mg/dL), the total proteins (4.75 ± 1.25 mg/dL), aspartate transaminase (40.50 ± 1.50 L), and alanine transaminase (43.52 4.50 L). The effects of the acute and chronic toxicity profile of ethanolic root extracts on the clinical, hematological, and biochemical parameters of albino rats were studied by Luka et al. [25]. The intraperitoneal LD50 of the extract was 570 mg/kg. Although there was no statistically significant change in body weight (p > 0.05), there was a substantial increase in hematological parameters such as the packed cell volume, the hemoglobin concentration, red blood cells, white blood cells, and differential leucocyte counts following delivery (p > 0.05). The mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentrations all increased significantly (p > 40.05) in a comparable manner. The acute and subchronic toxicity of the crude methanolic extract of D. mespiliformis and its fraction (hexane, ethyl acetate, and butanol) in Wistar rats was evaluated by Ebbo et al. [88]. Acute oral administration of the methanolic extract (5 g/kg bw) did not result in death, overt behavioral alterations, or any other physiological activities, and the LD50 of the crude methanolic leaf and bark extract was higher than 5 g/kg bw in Wistar rats. In a 28-day repeated dosage oral toxicity experiment, no notable adverse effects were observed in any of the parameters studied.
To evaluate the gastroprotective efficacy of leaf aqueous extracts of D. mespiliformis, Amang et al. [75] employed three experimental models of stomach ulcers in mice: HCl/ethanol, HCl/ethanol with indomethacin pre-treatment, and indomethacin (p.o). By administering the extract, stomach lesions caused by necrotizing drugs were avoided. The extract reduced the risk of developing ulcers after HCl/ethanol induction by 28.36%, 29.19%, and 35.82% at doses of 100, 200, and 400 mg/kg, respectively. Indomethacin pre-treatment reduced the extract’s preventive effectiveness to 19.69% and 28.24% at 200 and 400 mg/kg, respectively. During indomethacin induction, the extract at 200 mg/kg had the highest level of ulcer inhibition (88.13%). For each of the three induction models, a significant increase in mucus secretion between 44.75% and 121.34% was observed. Nwaogu et al. [82] reported the findings of acute administration of D. mespiliformis stem bark extracts in methanol at a dose of 5000 mg/kg body weight. After 48 h of observation, an acute dose of 5000 mg/kg body weight of methanol stem bark extract did not cause any deaths. Therefore, it was concluded that the extract’s median lethal dosage (LD50) was greater than 5000 mg/kg body weight. The extracts had no noticeable negative behavioral effects, such as retching, depression, tremors, weakness, refusing food and water, salivation, discharge from the eyes and ears, skin changes, or hair loss. Vandi et al. [8] detailed the antisecretory mechanism of D. mespiliformis root bark aqueous extract in Wistar rats. Three experimental animal models of excessive stomach acid secretion were used to test the extract: pyloric ligation, pyloric ligation plus histamine, and carbachol pretreatments. The ulcerated surface, the amount of mucus, the pH, the gastric acidity, and the pepsin activity were all measured. Malondialdehyde, superoxide dismutase, catalase, and reduced glutathione have all been identified as in vivo indicators of oxidative stress. Root bark aqueous extracts increased the mucus mass and stomach ulcer inhibition percentages in the three models under study, ranging from 9.50% to 59.52%. This increase was accompanied by a reduction in acidity and pepsin activity. Administration of the root bark aqueous extract of D. mespiliformis resulted in a significant decrease (p < 0.05, p < 0.01) in malondialdehyde levels, correlated with a significant increase (p< 0.05, p < 0.01) in catalase and nitrite levels compared with the negative control.

3. Materials and Methods

From multiple databases, including Science Direct, Google Scholar, Scopus, Web of Science, and Pub Med, every relevant scientific paper on the botanical description, traditional medicinal uses, phytochemical constituents, pharmacological and biological activities, clinical studies, and toxicology of Diospyros mespiliformis was retrieved. During the literature search, search terms including “Diospyros mespiliformis”, “traditional use”, “ethnomedicinal use”, “biological activity”, “toxicity”, “phytochemistry”, and “isolated compound” were combined.

4. Conclusions and Future Perspectives

D. mespiliformis has been used traditionally to treat a wide range of bacterial, fungal, parasitic, and viral diseases mainly in southern and western Africa. This plant has been shown in numerous studies to have strong pharmacological efficacy against a range of bacteria, fungi, viruses, and parasites. This review reports the first comprehensive summary of the traditional uses, phytochemical constituents, pharmacological activities, toxicity, and some in vivo studies of D. mespiliformis. The plant has a wide range of traditional uses, bioactive compounds, and pharmacological activities. According to the literature review, it can serve as a potential source of antimicrobial, antiparasitic, antiviral, anti-inflammatory, hypoglycemic, and antioxidant activities. Some of the different parts of the plant have also been tested for in vivo pharmacological activity and toxicity. The pharmacological effects of several compounds characterized by D. mespiliformis have still not been investigated. Therefore, in vitro and in vivo investigations to determine the potential efficacy and toxicity profile of these isolated compounds could fill in the identified gaps. No molecular research has been performed on this plant. Consequently, further research is needed to comprehend the molecular mechanisms underlying the documented pharmacological effects of the extracts and identified isolated compounds against a range of infectious illnesses. It is crucial to note that most of the data that were reviewed were evaluations of in vitro pharmacological activities. Considering the widespread antibiotic resistance, more work is required to gather more thorough evidence that will validate the in vitro findings in in vivo animal models. Furthermore, extensive pre-clinical and clinical research is also needed to determine the efficacy of this plant to establish it and its constituents as a potential effective alternative for disease prevention.

Author Contributions

Conceptualization, T.E.R. and S.M.-T.; methodology, T.E.R.; writing—original draft preparation, S.M.-T.; writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available upon request from the corresponding authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Atta-U-Rahman, M.; Igbal, C.; Thomson, W.J. Bioassay Techniques for Drug Development, 2nd ed.; Taylor and Francis: Singapore, 2005. [Google Scholar]
  2. Gerard, J.; Louppe, D. Afzelia africana Sm. ex pers. (Internet) Record from PROTA4U; PROTA (Plant Resources of Tropical Africal/Ressources Vegetales de l’Afrique Tropicale): Wageningen, The Netherlands, 2011. [Google Scholar]
  3. Orwa, C.; Mutua, A.; Kindt, R.; Jamnadass, R.; Simons, A. Agroforestree Database: A Tree Reference and Selection Guide; Version 4; World Agroforestry Centre: Nairobi, Kenya, 2009. [Google Scholar]
  4. Dangoggo, S.M.; Hassan, L.G.; Sadiq, I.S.; Manga, S.B. Phytochemical analysis and antibacterial screening of leaves of Diospyros mespiliformis and Ziziphus spina-christi. J. Chem. Eng. 2012, 1, 31–37. [Google Scholar]
  5. Esimone, C.O.; Nworu, C.S.; Onuigbo, E.B.; Omeje, J.U.; Nsirim, K.L.; Ogbu, J.C.; Ngwu, M.I.; Chah, K.F. Anti-mycobacterial activity of root and leaf extracts of Anthocleista djalonensis (Loganiaceae) and Diospyros mespiliformis (Ebenaceae). Int. J. Green Pharm. 2009, 3, 201–205. [Google Scholar]
  6. Adeniyi, B.A.; Odelola, H.A.; Oso, B.A. Antimicrobial potentials of Diospyros mespiliformis (Ebenaceae). Afr. J. Med. Med. Sci. 1996, 25, 221–224. [Google Scholar]
  7. Mudau, T.E.; Olowoyo, J.O.; Amoo, S.O. Ethnobotanical assessment of medicinal plants used traditionally for treating diabetes in Vhembe district, Limpopo Province, South Africa. S. Afr. J. Bot. 2022, 146, 304–324. [Google Scholar] [CrossRef]
  8. Vandi, V.L.; Amang, A.P.; Mezui, C.; Siwe, G.T.; Ndji, G.L.; Mbida, H.; Baponwa, O.; Tan, P.V. Antihistaminergic and anticholinergic properties of the root bark aqueous extract of Diospyros mespiliformis (Ebenaceae) on hypersecretion of gastric acid induced in wistar rats. eCAM 2022, 2022, 5190499. [Google Scholar] [CrossRef] [PubMed]
  9. Mustapha, M.D.; Hassan, L.G.; Umar, K.J.; Abubakar, K.; Yusuf, H.M.A. Phytochemical screening and cytotoxic effects of Diospyros mespiliformis Hochst (Ebenaceae) root bark extract using Brine Shrimp (Artemia salina) Test. AJBAR 2022, 1, 1–7. [Google Scholar] [CrossRef]
  10. Maitera, O.N.; Louis, H.; Oyebanji, O.O.; Anumah, A.O. Investigation of tannin content in Diospyros mespiliformis extract using various extraction solvents. J. Anal. Pharm. Res. 2018, 7, 55–59. [Google Scholar]
  11. David, O.M.; Olanlokun, J.O.; Owoniyi, B.E.; Ayeni, M.; Ebenezer, O.; Koorbanally, N.A. Studies on the mitochondrial, immunological and inflammatory effects of solvent fractions of Diospyros mespiliformis Hochst in Plasmodium berghei-infected mice. Sci. Rep. 2021, 11, 6941. [Google Scholar] [CrossRef]
  12. Olanlokun, J.O.; Bodede, O.; Prinsloo, G.; Olorunsogo, O.O. Comparative antimalarial, toxicity and mito-protective effects of Diospyros mespiliformis Hochst. ex A. DC. and Mondia whitei (Hook. f.) Skeels on Plasmodium berghei infection in mice. J. Ethnopharmacol. 2021, 268, 113585. [Google Scholar] [CrossRef]
  13. Anas, A.; Ahmed, A.; Umar, S.; Jajere, U.M.; Mshelia, E.H.; Natasha, O. Inhibitory effect of isolated lupeol from stem bark of Diospyros mespiliformis Horsch (Ebenaceae) against some microbial pathogens. Bayero J. Pure Appl. Sci. 2017, 10, 293–299. [Google Scholar] [CrossRef]
  14. Mohamed, I.E.; El Bushra, E.; Choudhary, M.I.; Khan, S.N. Bioactive natural products from two Sudanese medicinal plants Diospyros mespiliformis and Croton zambesicus. Rec. Nat. Prod. 2009, 3, 198. [Google Scholar]
  15. Mabona, U.; Viljoen, A.; Shikanga, E.; Marston, A.; Van Vuuren, S. Antimicrobial activity of southern African medicinal plants with dermatological relevance: From an ethnopharmacological screening approach, to combination studies and the isolation of a bioactive compound. J. Ethnopharmacol. 2013, 148, 45–55. [Google Scholar] [CrossRef]
  16. Koenen, E.V. Medicinal, Poisonous and Edible Plants in Namibia; Klaus Hess Verlag: Göttingen, Germany, 1996. [Google Scholar]
  17. Watt, J.M.; Breyer-Brandwijk, M.G. The Medicinal and Poisonous Plants of Southern and Eastern Africa, 2nd ed.; Livingstone: Edinburg, UK; London, UK, 1962. [Google Scholar]
  18. Luseba, D.; Van der Merwe, D. Ethnoveterinary medicine practices among Tsonga speaking people of South Africa. Onderstepoort J. Vet. Res. 2006, 73, 115–122. [Google Scholar] [CrossRef] [PubMed]
  19. Mahwasane, S.T.; Middleton, L.; Boaduo, N. An ethnobotanical survey of indigenous knowledge on medicinal plants used by the traditional healers of the Lwamondo area, Limpopo province, South Africa. S. Afr. J. Bot. 2013, 88, 69–75. [Google Scholar] [CrossRef]
  20. Bapela, M.J.; Meyer, J.M.; Kaiser, M. In vitro antiplasmodial screening of ethnopharmacologically selected South African plant species used for the treatment of malaria. J. Ethnopharmacol. 2014, 156, 370–373. [Google Scholar] [CrossRef] [PubMed]
  21. Masevhe, N.A.; McGaw, L.J.; Eloff, J.N. The traditional use of plants to manage candidiasis and related infections in Venda, South Africa. J. Ethnopharmacol. 2015, 168, 364–372. [Google Scholar] [CrossRef]
  22. Tshikalange, T.E.; Mophuting, B.C.; Mahore, J.; Winterboer, S.; Lall, N. An ethnobotanical study of medicinal plants used in villages under Jongilanga tribal council, Mpumalanga, South Africa. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 83–89. [Google Scholar] [CrossRef] [PubMed]
  23. Tapsoba, H.; Deschamps, J.P. Use of medicinal plants for the treatment of oral diseases in Burkina Faso. J. Ethnopharmacol. 2006, 104, 68–78. [Google Scholar] [CrossRef] [PubMed]
  24. Maroyi, A. An ethnobotanical survey of medicinal plants used by the people in Nhema communal area, Zimbabwe. J. Ethnopharmacol. 2011, 136, 347–354. [Google Scholar] [CrossRef]
  25. Luka, J.; Badau, S.J.; Mbaya, A.W.; Gadzama, J.J.; Kumshe, H.A. Acute toxicity study and effect of prolonged administration (28 days) of crude ethanolic root extract of Diospyros mespiliformis Hochst (Ebenaceae) on clinical, haematological and biochemical parameters of albino rats. J. Ethnopharmacol. 2014, 153, 268–273. [Google Scholar] [CrossRef]
  26. Moyo, B.; Ndlovu, S.L.; Moyo, S.; Masika, P.J.; Muchenje, V.; Ndhlovu, D.N.; Maphosa, V. Alternative remedies and approaches used by resources-challenged farmers in the management of cattle black-leg disease in Umzingwane district, Matabeleland South, Zimbabwe. Int. J. Livest. Prod. 2014, 6, 97–102. [Google Scholar]
  27. Dossou-Yovo, H.O.; Vodouhe, F.G.; Sinsin, B. Assessment of the medicinal uses of plant species found on termitaria in the Pendjari biosphere reserve in Benin. J. Med. Plant Res. 2014, 8, 368–377. [Google Scholar]
  28. Koné, W.M.; Atindehou, K.K. Ethnobotanical inventory of medicinal plants used in traditional veterinary medicine in Northern Côte d’Ivoire (West Africa). S. Afr. J. Bot. 2008, 74, 76–84. [Google Scholar] [CrossRef]
  29. Chinsembu, K.C.; Hijarunguru, A.; Mbangu, A. Ethnomedicinal plants used by traditional healers in the management of HIV/AIDS opportunistic diseases in Rundu, Kavango East Region, Namibia. S. Afr. J. Bot. 2015, 100, 33–42. [Google Scholar] [CrossRef]
  30. Sulaiman, A.N.; Arzai, A.H.; Taura, D.W. Ethnobotanical survey: A comprehensive review of medicinal plants used in treatment of gastrointestinal diseases in Kano state, Nigeria. Phytomed. Plus. 2022, 2, 100180. [Google Scholar] [CrossRef]
  31. Setshego, M.V.; Aremu, A.O.; Mooki, O.; Otang-Mbeng, W. Natural resources used as folk cosmeceuticals among rural communities in Vhembe district municipality, Limpopo province, South Africa. BMC Complement. Med. 2020, 20, 81. [Google Scholar] [CrossRef]
  32. Chinsembu, K.C. Ethnobotanical study of medicinal flora utilised by traditional healers in the management of sexually transmitted infections in Sesheke District, Western Province, Zambia. Rev. Bras. Farmacogn. 2016, 26, 268–274. [Google Scholar] [CrossRef]
  33. Mamba, P.; Adebayo, S.A.; Tshikalange, T.E. Anti-microbial, Anti-inflammatory and HIV-1 reverse transcriptase activity of selected South African plants used to treat sexually transmitted diseases. Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 1870–1876. [Google Scholar]
  34. Nadembega, P.; Boussim, J.I.; Nikiema, J.B.; Poli, F.; Antognoni, F. Medicinal plants in Baskoure, Kourittenga province, Burkina Faso: An ethnobotanical study. J. Ethnopharmacol. 2011, 133, 378–395. [Google Scholar] [CrossRef]
  35. Van Wyk, B.; Van Wyk, P.; Van Wyk, B.E. Photo Guide to Trees of Southern Africa; Briza: Pretoria, South Africa, 2008. [Google Scholar]
  36. Adeniyi, B.A.; Robert, M.F.; Chai, H.; Fong, H.H.S. In vitro cytotoxicity activity of diosquinone, a naphthoquinone epoxide. Phytother. Res. 2003, 17, 282–284. [Google Scholar] [CrossRef] [PubMed]
  37. Atawodi, S.E.; Bulus, T.; Ibrahim, S.; Ameh, D.A.; Nok, A.J.; Mamman, M.; Galadima, M. In vitro trypanocidal effect of methanolic extract of some Nigerian savannah plants. Afr. J. Biotechnol. 2003, 2, 317–321. [Google Scholar]
  38. Von Koenen, E. Medicinal, Poisonous and Edible Plants of Namibia; Klaus Hess Publishers: Windhoek, Namibia; Gottingen, Germany, 2001. [Google Scholar]
  39. Etkin, N.L. Antimalarial plants used by Hausa in Northern Nigeria. Trop. Dr. 1997, 27, 12–16. [Google Scholar] [CrossRef] [PubMed]
  40. Mabogo, D.E.N. The Ethnobotany of the Vhavenda. Master’s Thesis, University of Pretoria, Pretoria, South Africa, 1990. [Google Scholar]
  41. Khan, M.R.; Nkunya, M.H.H.; Wevers, H. Triterpenoids from leaves of Diospyros species. Planta Med. 1980, 38, 380–381. [Google Scholar] [CrossRef]
  42. Arnold, H.; Gulumian, M. Pharmacopoeia of traditional medicine in Venda. J. Ethnopharmacol. 1984, 12, 35–74. [Google Scholar] [CrossRef] [PubMed]
  43. Kerharo, J. Historic and ethnopharmacognostic review on the belief and traditional practices in the treatment of sleeping sickness in West Africa. Bull. Soc. Med. Afr. Black Lang. Fr. 1974, 19, 400. [Google Scholar]
  44. Kantati, Y.T.; Kodjo, K.M.; Dogbeavou, K.S.; Vaudry, D.; Leprince, J.; Gbeassor, M. Ethnopharmacological survey of plant species used in folk medicine against central nervous system disorders in Togo. J. Ethnopharmacol. 2016, 181, 214–220. [Google Scholar] [CrossRef] [PubMed]
  45. Ziblim, I.A.; Timothy, K.A.; Deo-Anyi, E.J. Exploitation and use of medicinal plants, Northern Region, Ghana. J. Med. Plant Res. 2013, 7, 1984–1993. [Google Scholar]
  46. Klotoé, J.R.; Dougnon, T.V.; Koudouvo, K.; Atègbo, J.M.; Loko, F.; Akoègninou, A.; Aklikokou, K.; Dramane, K.; Gbeassor, M. Ethnopharmacological survey on antihemorrhagic medicinal plants in South of Benin. Eur. J. Med. Plants 2012, 3, 40–51. [Google Scholar] [CrossRef]
  47. Nfi, A.N.; Mbanya, J.N.; Ndi, C.; Kameni, A.; Vabi, M.; Pingpoh, D.; Yonkeu, S.; Moussa, C. Ethnoveterinary medicine in the Northern Provinces of Cameroon. Vet. Res. Commun. 2001, 25, 71–76. [Google Scholar] [CrossRef]
  48. Semenya, S.S.; Maroyi, A. Ethnobotanical survey of plants used by Bapedi traditional healers to treat tuberculosis and its opportunistic infections in the Limpopo Province, South Africa. S. Afr. J. Bot. 2019, 122, 401–421. [Google Scholar] [CrossRef]
  49. Cheikhyoussef, A.; Shapi, M.; Matengu, K.; Mu Ashekele, H. Ethnobotanical study of indigenous knowledge on medicinal plant use by traditional healers in Oshikoto region, Namibia. J. Ethnobiol. Ethnomedicine 2011, 7, 10. [Google Scholar] [CrossRef]
  50. Green, E.; Samie, A.; Obi, C.L.; Bessong, P.O.; Ndip, R.N. Inhibitory properties of selected South African plants against Mycobacterium tuberculosis. J. Ethnopharmacol. 2010, 130, 151–157. [Google Scholar] [CrossRef]
  51. Kokwaro, J.O. Medicinal Plants of East Africa; University of Nairobi Press: Nairobi, Kenya, 2009. [Google Scholar]
  52. Stafford, G.I.; Pedersen, M.E.; van Staden, J.; Jager, A.K. Review on plants with CNS-effects used in traditional South African medicine against mental diseases. J. Ethnopharmacol. 2008, 119, 513–537. [Google Scholar] [CrossRef] [PubMed]
  53. Chivandi, E.; Erlwanger, K.H.; Davidson, B.C. Lipid content and fatty acid profile of the fruit seeds of Diospyros mespiliformis. Int. J. Integr. Biol. 2009, 5, 121–124. [Google Scholar]
  54. Hegazy, A.K.; Mohamed, A.A.; Ali, S.I.; Alghamdi, N.M.; Abdel-Rahman, A.M.; Al-Sobeai, S. Chemical ingredients and antioxidant activities of underutilized wild fruits. Heliyon 2019, 5, e01874. [Google Scholar] [CrossRef] [PubMed]
  55. Ebbo, A.A.; Sani, D.; Suleiman, M.M.; Ahmad, A.; Hassan, A.Z. Assessment of antioxidant and wound healing activity of the crude methanolic extract of Diospyros mespiliformis Hochst ex a. Dc (Ebenaceae) and its fractions in Wistar rats. S. Afr. J. Bot. 2022, 150, 305–312. [Google Scholar] [CrossRef]
  56. Adewuyi, A.; Oderinde, R.A. Fatty acid composition and lipid profile of Diospyros mespiliformis, Albizia lebbeck, and Caesalpinia pulcherrima seed oils from Nigeria. Int. J. Food Sci. 2014, 2014, 283614. [Google Scholar] [CrossRef]
  57. Petzke, K.J.; Ezeagu, I.E.; Proll, J.; Akinsoyinu, A.O.; Metges, C.C. Amino acid composition, available lysine content and in vitro protein digestibility of selected tropical crop seeds. Foods Hum. Nutr. 1997, 50, 151–162. [Google Scholar] [CrossRef]
  58. Glew, R.S.; Vanderjagt, D.J.; Chuang, L.T.; Huang, Y.S.; Millson, M.; Glew, R.H. Nutrient content of four edible wild plants from West Africa. Foods Hum. Nutr. 2005, 60, 187–193. [Google Scholar] [CrossRef]
  59. Achaglinkame, M.A.; Aderibigbe, R.O.; Hensel, O.; Sturm, B.; Korese, J.K. Nutritional characteristics of four underutilized edible wild fruits of dietary interest in Ghana. Foods 2019, 8, 104. [Google Scholar] [CrossRef]
  60. Zhong, S.M.; Waterman, P.G.; Jeffreys, J.A.D. Naphthoquinones and triterpenes from African Diospyros species. Phytochemistry 1984, 23, 1067–1072. [Google Scholar] [CrossRef]
  61. Adzu, B.; Amos, S.; Dzarma, S.; Muazzam, I.; Gamaniel, K.S. Pharmacological evidence favouring the folkloric use of Diospyros mespiliformis Hochst in the relief of pain and fever. J. Ethnopharmacol. 2002, 82, 191–195. [Google Scholar] [CrossRef]
  62. Adeniyi, B.A.; Fong, H.H.S.; Pezzuto, J.M.; Luyengi, L.; Odelola, H.A. Antibacterial activity of diospyrin, isodiospyrin and bisisodiospyrin from the root of Diospyros piscatoria (Gurke)(Ebenaceae). Phytother. Res. 2000, 14, 112–117. [Google Scholar] [CrossRef]
  63. Fallas, A.L.; Thomson, R.H. Ebenaceae extractives. Part III. Binaphthaquinones from Diospyros species. J. Chem. Society C Org. 1968, 2279–2282. [Google Scholar] [CrossRef]
  64. Lajubutu, B.A.; Pinney, R.J.; Roberts, M.F.; Odelola, H.A.; Oso, B.A. Antibacterial activity of diosquinone and plumbagin from the root of Diospyros mespiliformis (Hostch)(Ebenaceae). Phytother. Res. 1995, 9, 346–350. [Google Scholar] [CrossRef]
  65. Hawas, U.W.; El-Ansari, M.A.; El-Hagrassi, A.M. A new acylated flavone glycoside, in vitro antioxidant and antimicrobial activities from Saudi Diospyros mespiliformis Hochst. ex A. DC (Ebenaceae) leaves. ZNC 2022, 77, 387–393. [Google Scholar] [CrossRef] [PubMed]
  66. Shai, L.J.; Chauke, M.A.; Magano, S.R.; Mogale, A.M.; Eloff, J.N. Antibacterial activity of sixteen plant species from Phalaborwa, Limpopo Province, South Africa. J. Med. Plants Res. 2013, 7, 899–906. [Google Scholar]
  67. Aderbauer, B.; Clausen, P.H.; Kershaw, O.; Melzig, M. In vitro and in vivo trypanocidal effect of lipophilic extracts of medicinal plants from Mali and Burkina Faso. J. Ethnopharmacol. 2008, 119, 225–231. [Google Scholar] [CrossRef]
  68. Hedimbi, M. Evaluation of Selected Namibian Ethno-Medicinal Plants for Anti-HIV Properties. Ph.D. Thesis, University of Namibia, Windhoek, Namibia, 2015. [Google Scholar]
  69. Traore, M.S.; Diane, S.; Diallo, M.S.T.; Balde, E.S.; Balde, M.A.; Camara, A.; Diallo, A.; Keita, A.; Cos, P.; Maes, L.; et al. In vitro antiprotozoal and cytotoxic activity of ethnopharmacologically selected Guinean plants. Planta Med. 2014, 80, 1340–1344. [Google Scholar] [CrossRef]
  70. Chukwuma, O.J.T. Antiviral activities of the aqueous, ethanolic and methanolic extracts of Diospyros mespiliformis leaf on some pathogenic Avian viruses. IDOSR J. Exp. Sci. 2017, 2, 35–49. [Google Scholar]
  71. Belemtougri, R.G.; Constantin, B.; Cognard, C.; Raymond, G.; Sawadogo, L. Effects of two medicinal plants Psidium guajava L. (Myrtaceae) and Diospyros mespiliformis L. (Ebenaceae) leaf extracts on rat skeletal muscle cells in primary culture. J. Zhejiang Univ. Sci. B 2006, 7, 56–63. [Google Scholar] [CrossRef]
  72. Lawal, F.; Bapela, M.J.; Adebayo, S.A.; Nkadimeng, S.M.; Yusuf, A.A.; Malterud, K.E.; McGaw, L.J.; Tshikalange, T.E. Anti-inflammatory potential of South African medicinal plants used for the treatment of sexually transmitted infections. S. Afr. J. Bot. 2019, 125, 62–71. [Google Scholar] [CrossRef]
  73. Nafuka, S.N. In Vitro Antiplasmodial Activity and Phytochemicals Screening of Ethnomedicinal Plants Used to Treat Malaria Associated Symptoms. Ph.D. Thesis, University of Namibia, Windhoek, Namibia, 2014. [Google Scholar]
  74. Adoum, O.A. Screening of medicinal plants native to Kano and Jigawa states of northern Nigeria, using Artemia cysts (brine shrimp test). Am. J. Pharmacol. Sci. 2016, 4, 7–10. [Google Scholar]
  75. Amang, A.P.; Bouvourne, P.; Mezui, C.; Siwe, G.T.; Kuissu, M.T.; Vernyuytan, P. Gastro-protective activity of the leaves aqueous extract of Diospyros mespiliformis on gastric ulcers in Swiss mice. Int. J. Pharmacogn 2020, 7, 44–51. [Google Scholar]
  76. Bapela, M.J.; Kaiser, M.; Meyer, J.J.M. Antileishmanial activity of selected South African plant species. S. Afr. J. Bot. 2017, 108, 342–345. [Google Scholar] [CrossRef]
  77. Agbadoronye, P.; Abolarinwa, S.; Lawal, B.; Odeyemi, S.; Irhue, A.; Achagwa, S.; Ngamdu, S. Alkaloidal fraction of Diospyros mespiliformis protect against Trypanosoma evansi-mediated haematological and hepatic impairment in infected rats. J. Appl. Nat. Sci. 2021, 1, 66–78. [Google Scholar]
  78. Olanlokun, J.O.; Adetutu, J.A.; Olorunsogo, O.O. ln vitro inhibition of beta-hematin formation and in vivo effects of Diospyros mespiliformis and Mondia whitei methanol extracts on chloroquine-susceptible Plasmodium berghei-induced malaria in mice. Interv. Med. Appl. Sci. 2021, 11, 197–206. [Google Scholar] [CrossRef] [PubMed]
  79. Jigam, A.A.; Abdulrazaq, U.T.; Suleiman, R.S.; Kali, P.S. Effects of sub-chronic administration of Diospyros mespiliformis Hochst (Ebenaceae) root extracts on some biochemical parameters in mice. J. Appl. Pharm. Sci. 2012, 2, 60–64. [Google Scholar] [CrossRef]
  80. Adzu, B.; Chindo, B.A.; Tarfa, F.D.; Salawu, O.A.; Igoli, O.J. Isolation and analgesic property of lupeol from Diospyros mespiliformis stem bark. J. Med. Plant Res. 2015, 9, 813–819. [Google Scholar]
  81. Chinwe, U.J.; Adedotun, A.A.; Mudi, S.Y.; Musa, H.; Oluwagbemiga, A.O.; Babatunde, A. In-vivo therapeutic efficacy and phytochemical investigation of three commonly used plants for malaria treatment by the Hausa community in Kano, Nigeria. J. Pharmacogn. Phytochem. 2021, 10, 111–117. [Google Scholar]
  82. Nwaogu, J.; Fakai, I.M.; Yahaya, I. Hepatoprotective effect of methanol stem bark extract of Diospyros mespiliformis against carbon tetrachloride induced liver damage. Asian J. Biochem. 2022, 12, 18–25. [Google Scholar] [CrossRef]
  83. Mallavadhani, U.V.; Panda, A.K.; Rao, Y.R. Pharmacology and chemotaxonomy of Diospyros. Phytochemistry 2007, 49, 901–951. [Google Scholar] [CrossRef] [PubMed]
  84. Chantal, M.M.B.; Josiane, N.M.T.; Baudelaire, N.E.; Nicolas, N.Y. Antioxidant properties of granulometric classes and solvent extraction of Diospyros mespiliformis Hochst. ex A. fruits powder. Int. J. Mod. Biol. Res. 2021, 9, 1–16. [Google Scholar]
  85. Ndhlala, A.R.; Chitindingu, K.; Mupure, C.; Murenje, T.; Ndhlala, F.; Benhura, M.A.; Muchuweti, M. Antioxidant properties of methanolic extracts from Diospyros mespiliformis (jackal berry), Flacourtia indica (Batoka plum), Uapaca kirkiana (wild loquat) and Ziziphus mauritiana (yellow berry) fruits. Int. J. Food Sci. Technol. 2008, 43, 284–288. [Google Scholar] [CrossRef]
  86. Shikwambana, N.; Mahlo, S.M. A survey of antifungal activity of selected south African plant species used for the treatment of skin infections. Nat. Prod. Commun. 2020, 15, 1934578X20923181. [Google Scholar] [CrossRef]
  87. Sombie, E.N.; Tibiri, A.; N’do, J.Y.P.; Traore, T.K.; Ouedraogo, N.; Hilou, A.; Guissou, P.I.; Nacoulma, O.G. Ethnobotanical study and antioxidant activity of anti-hepatitis plants extracts of the COMOE province, Burkina Faso. Int. J. Biol. Chem. Sci. 2018, 12, 1308–1319. [Google Scholar] [CrossRef]
  88. Ebbo, A.A.; Sani, D.; Suleiman, M.M.; Ahmad, A.; Hassan, A.Z. Acute and sub-chronic toxicity evaluation of the crude methanolic extract of Diospyros mespiliformis hochst ex a. Dc (ebenaceae) and its fractions. Toxicol. Rep. 2020, 7, 1138–1144. [Google Scholar] [CrossRef]
Figure 2. D. mespiliformis leaves and fruits; https://suntrees.co.za/diospyros-mespiliformis-jackalberry-jakkalsbessie-motlouma/ (accessed on 31 July 2023).
Figure 2. D. mespiliformis leaves and fruits; https://suntrees.co.za/diospyros-mespiliformis-jackalberry-jakkalsbessie-motlouma/ (accessed on 31 July 2023).
Molecules 28 07759 g002
Figure 3. Major phenolic compounds in D. mespiliformis.
Figure 3. Major phenolic compounds in D. mespiliformis.
Molecules 28 07759 g003aMolecules 28 07759 g003bMolecules 28 07759 g003c
Figure 4. Triterpenoids isolated or detected from D. mespiliformis.
Figure 4. Triterpenoids isolated or detected from D. mespiliformis.
Molecules 28 07759 g004aMolecules 28 07759 g004b
Figure 5. Structures of long-chain fatty acids and other classes of phytochemicals from D. mespiliformis.
Figure 5. Structures of long-chain fatty acids and other classes of phytochemicals from D. mespiliformis.
Molecules 28 07759 g005aMolecules 28 07759 g005b
Table 1. Traditional uses of the different parts of D. mespiliformis.
Table 1. Traditional uses of the different parts of D. mespiliformis.
Part UsedTraditional UsesCountryReferences
LeavesRingworm, urinary, and sexually transmitted infections, sleeping sickness, malaria, headaches, anthelmintic, wounds, dysentery, fever, leprosy, scars, skin rashes, bruises, styptic to staunch bleeding, diarrhoea, tonic, febrifuge, stomach aches, and coughsSouth Africa, Ivory Coast, Nigeria, Zambia, Burkina Faso, Namibia[15,16,17,28,30,31,32,33,34,35,36,37,38,39,40,41,42,43]
StemBlackleg disease in cattle, diabetes mellitus, stroke, traumatic brain injury, and malariaSouth Africa, Zimbabwe, Burkina Faso, Togo[7,26,34,44]
BarkOral diseases, stomach problems, diarrhoea, coughs, leprosy, STIs and urinary tract infections, dysentery, fever, vomiting, pneumonia, syphilis, and hemorrhages. Ethnoveterinary: Helminthiasis, milk production in animals, mental illness, headaches, epilepsy, and convulsionsSouth Africa, Burkina Faso, Nigeria, Tanzania, Senegal, Ivory Coast, Ghana, Benin, Cameroon[18,19,23,39,40,41,42,43,45,46,47]
Roots Ringworm, urinary, and sexually transmitted infections, abdominal pains, stomach aches, tuberculosis, male sexual dysfunction, scars, skin rashes, bruises, wounds, ringworm, dysentery, fever, coughs, epilepsy, pneumonia, syphilis, mental illness, headaches, epilepsy, convulsions, and worm expellantSouth Africa, Namibia, Zimbabwe, Nigeria, Ghana, Kenya[15,16,17,24,33,35,36,38,40,45,48,49,50,51,52]
FruitsDysentery, fungal infections, diarrhoea, tonic, febrifuge, skin diseases, menstrual pain, and ringwormsSouth Africa, Benin, Burkina Faso[21,27,28,31,34,40]
TwigsTeeth cleaningSouth Africa[31]
Table 3. Pharmacological activities of different parts of plants and major compounds of D. mespiliformis.
Table 3. Pharmacological activities of different parts of plants and major compounds of D. mespiliformis.
Plant Part/CompoundsSolvents UsedPharmacological ActivityBioassay ModelResultsReferences
LeavesAcetone Antioxidant DPPHIC50 = 25 ± 2 μg/mL[55]
Antibacterial MIC (B. stearothermophilus)80 µg/mL[65]
Antifungal MIC (C. albicans and M. canis and T. rubrum)80 µg/mL for C. albicans, 20 µg/mL for M. canis and 20 µg/mL for T. rubrum [66]
DCMAntiproliferativeIn vitro cytotoxicityMTC > 500 µg/mL on fibroblast-like mammalian cells[67]
DCM: MeOHAntibacterialMIC (P. acnes ATCC 11827 and T. mentagrophytes)50 µg/mL for P. acnes and 100 µg/mL for T. mentagrophytes.[15]
Antiparasitic Long-term viability assay
(T. brucei)
MIC = 500 µg/mL[67]
70% Ethanol Antimicrobial MIC (C. albicans ATCC 10231, G. vaginalis ATCC 14018, N. gonorrhoeae ATCC 19424 and O. ureolytica ATCC 43534)3.1–6.3 mg/mL[33]
Antiviral HIV-1 RT colorimetric ELISA kit (Roche)78.7% at 0.1 mg/mL had [68]
Antiparasitic In vitro antiplasmodial activity IC50 = 25.8 µg/mL for Trypanosoma cruzi, IC50 = >64 µg/mL for Leishmania infantum [69]
AntiproliferativeIn vitro cytotoxicityIC50 = >64 µg/mL for MRC-5 fibroblasts[69]
Ethanol Antiviral In vitro allantoic sac routes of developing chick embryos95.0%, 90.5%, and 89.0% at 400 mg/mL, 200 mg/mL, and 100 mg/mL respectively, for Newcastle disease virus[70]
Anti-hypersensitivityIntracellular free calcium measurementsReduced amplitude of Ca2+ release from SR at 10 mg/mL. IC50 = 9.23 mg/mL and 54% inhibited calcium release. [71]
MeOHAntioxidant DPPHIC50 = 6.94 ± 0.49 µg/mL[72]
AntimycobacterialMIC (M. smegmatis)167 µg/mL [5]
Antiparasitic In vitro antiplasmodial bioassay IC50 = 1.51 µg/mL for P. falciparum 3D7A[73]
Antiviral In vitro allantoic sac routes of developing chick embryos100.0%, 92.8%, and 90.5% at 400 mg/mL for Newcastle disease virus[70]
Toxicity Acute and subchronic toxicity in rats LD50 of >5g/kg. No notable adverse effects seen on parameters studied[72]
WaterAntioxidant ABTS, FRAP1.17 ± 0.00 TEAC in mM (ABTS). 70.77 ± 0.4 M ET/g[74]
Antifungal MIC (C. albicans, and T. rubrum)20 µg/mL for C. albicans, 40 µg/mL for T. rubrum. [66]
Antiparasitic In vitro antiplasmodial bioassay (P. falciparum 3D7A)IC50 = 3.01 µg/mL[73]
Antiviral In vitro allantoic sac routes of developing chick embryos91.0%, 86.0%, and 85.0% at 400 mg/mL, 200 mg/mL, and 100 mg/mL, respectively, for Newcastle disease virus[70]
Anti-hypersensitivityIntracellular free calcium measurementsIC50 = 8.84 mg/mL at 10 mg/mL. 29% inhibited calcium release. [71]
Toxicity Gastroprotective efficacy: stomach ulcer 200 mg/kg had the highest level of ulcer inhibition (88.13%)[75]
Leaf fractionsButanol Antioxidant DPPHIC50 = 1.44 ± 0.01 µg/mL[55]
HexaneAntioxidant DPPHIC50 = 28.03 ± 2.57 µg/mL[55]
Ethyl acetate Antioxidant DPPHIC50 = 1.08 ± 0.04 µg/mL[55]
Water Antioxidant DPPHIC50 = 4.73 ± 0.23 µg/mL[55]
RootsDCM: 50% MeOHAntiparasitic In vitro hypoxanthine incorporation assay (P. falciparum NF54)IC50 = 4.40/28.4 µg/mL[20]
Antileishmanial, resazurin assay (L. donovani MHOM-ET-67/L82)IC50 = 7.7 µg/mL for DCM and IC50 = 54 µg/mL for 50% MeOH.[76]
AntiproliferativeIn vitro inhibition of mammalian cell proliferationIC50 = 24.3 µg/mL for DCM and 60.4 µg/mL for MeOH[77]
EthanolAntiparasiticAcute toxicity and prolonged administration in ratsIntraperitoneal LD50 of 570 mg/kg[25]
70% Ethanol Anti-inflammatory15-LOXIC50 = 188.1 µg/mL[33]
Antiviral HIV-1 RT colorimetric ELISA kit (Roche)17.4% inhibition[33]
MeOHAntioxidant DPPHIC50 = 3.47 ± 0.05 µg/mL[55]
Antiparasitic In vitro antiplasmodial bioassay (P. falciparum 3D7A)IC50 = 2.12 µg/mL[73]
In vivo antiplasmodial activity in mice (Plasmodium berghei)High rate of parasite clearance (84.7%) and lower parasitemia (0.67%) [78]
Toxicity Subchronic in vivo studies Safe dose of 400mg/Kg bw and LD50 of 620mg/kg bw of mice.[79]
WaterAntibacterialDisc diffusion (S. aureus, P. aeruginosa, E. coli and Shigella spp). 10–13 mm on S. aureus, 11–13 mm on P. aeruginosa, 11–14 mm on E. coli and 10–11 mm on Shigella spp. [4]
Antiparasitic In vitro antiplasmodial bioassay (P. falciparum 3D7A)IC50 = 2.91 µg/mL[73]
Root barkAcetoneAnti-inflammatoryXO, NOIC50 = 142 8 µg/mL (XO) and IC50 = 79.8 ± 2.7 µg/mL (NO)[80]
Hexane Antiproliferative Brine shrimp (Artemia salina) cytotoxicity8203.52 μg/mL lethal dose [9]
WaterAntiproliferativeBrine shrimp (Artemia salina) cytotoxicity100% safe at 10–1000 μg/mL[9]
Antioxidant ABTS, DPPH and FRAPIC50 = 220 µg/mL for ABTS, 494 µg/mL DPPH and 543 µg/mL[8]
Antisecretory mechanismPyloric ligation, pyloric ligation plus histamine, and carbachol pretreatmentsIncreased mucus mass and stomach ulcer inhibition ranging from 9.50% to 59.52%[8]
Bark 95% Ethanol Antiparasitic In vivo antitrypanosomal activity of Trypanosoma evansi-infected rats Increased red blood cells and elevated bilirubin. Reduced total proteins[77]
HexaneAntimycobacterial MIC (M. tuberculosis H37Ra)100 µg/mL[50]
MeOH Antioxidant DPPHIC50 = 7.82 ± 0.76 µg/mL[55]
Antiparasitic In vivo antiplasmodial activity in mice (Plasmodium berghei NK65)53% at 800 mg/kg dosage[81]
Bark fractionsEthyl acetate and hexaneAnti-inflammatory Wound healing Fully healed [55]
Stem Ethanol Antiproliferative Brine shrimp (Artemia cysts) lethality test (BST)LC50 >100 μg/mL[79]
Stem bark MeOHAntipyreticIn vivo studiesLD50 = 513.80 ± 33.92 mg/kg i.p. in mice.[63]
Antiparasitic In vivo antiplasmodial activity against P. berghei ANKA in miceParasitemia (5 ± 1), increased packed cell volume (36% ± 1.4), increased platelets (2 ± 1.4 105 mm3), decreased alkaline phosphatase (56 ± 0.7 U/L), alanine aminotransferases (6.2 ± 0.8 U/L), and alanine aminotransferases (8 ± 3.8 U/L).[12]
Toxicity Acute toxicity and hepatoprotective effects LD50 > 5000 mg/kg bw. Possess hepatoprotective property by inhibiting lipid peroxidation.[82]
Water NeuropharmacologicalIn vivo studies in mice Increased pentobarbital-induced sleep, decreased exploratory and spontaneous motor behavior[83]
Stem fractionsDCMAntiparasitic In vivo antiplasmodial activity against P. berghei NK 65 in miceHigh parasite clearance[11]
FruitsEthanol Antioxidant DPPH. In vivo antioxidants in rats. IC50 = 1.037 ± 0.204 mg/mL. Increased the levels of the enzymes SOD, catalase, peroxidase, alanine transaminase, and aspartate aminotransferase[84]
Hydroethanolic Antioxidant DPPHIC50 = 1.111 ± 0.135 mg/mL[84]
MeOHAntioxidant DPPH radical scavenging, reducing power effects, and superoxide-anion-radical scavenging.Increase radical-scavenging effect, reducing power and superoxide-anion-radical-scavenging. [85]
Antioxidant DPPH, H2O2 scavenging 87.36% at 1 mg/mL for DPPH. >85% at 1 mg/m for H2O2.[58]
Isoscutellarein 7-O-(4′′′-O-acetyl)-β-allopyranosyl (1′′′ → 2″)-β-glucopyranoside (8) Antioxidant DPPHIC50 = 15.46 μg/mL[65]
Luteolin 3′,4′,6,8-tetramethyl ether (9) AntimycobacterialMIC (M. smegmatis)250 µg/mL[5]
Antibacterial Disc diffusion (E. coli)34 mm[65]
AntibacterialMIC (S. aureus) 9.77 μg/mL[65]
AntioxidantDPPH IC50 = 15.46 μg/mL[65]
Quercetin 3-O-α-rhamnoside (15) Antibacterial MIC (S. aureus NCTC 6571, S. aureus E3T, E. coli KL16 and P. aeruginosa NCTC 6750)3 to 30 μg/mL for S. aureus, 15 for E. coli and 16 μg/mL for P. aeruginosa[64]
AntioxidantDPPHIC50 = 12.32 μg/mL[65]
Diosquinone (2) Antiproliferative In vitro cytotoxicityED50 = 0.18 μg/mL for U373 cells, ED50 = 0.2 µg/mL for BC-1, HT-1080, Lu-1, KB, and SKNSH cells, ED50 = 1–1.7 µg/mL for KB-V(V-VLB) cells [36]
Plumbagin (12) Antidiabetic α- Glucosidase enzyme inhibition assayIC50 = 0.002 ± 0.004 mM[14]
Lupeol (27) Antidiabeticα- Glucosidase enzyme inhibition assayIC50 = 0.46 ± 0.002 mM[14]
Betulin (24) Antidiabeticα- Glucosidase enzyme inhibition assayIC50 = 0.0624 ± 0.002 mM[14]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ramadwa, T.E.; Meddows-Taylor, S. Traditional Uses, Pharmacological Activities, and Phytochemical Analysis of Diospyros mespiliformis Hochst. ex. A. DC (Ebenaceae): A Review. Molecules 2023, 28, 7759. https://doi.org/10.3390/molecules28237759

AMA Style

Ramadwa TE, Meddows-Taylor S. Traditional Uses, Pharmacological Activities, and Phytochemical Analysis of Diospyros mespiliformis Hochst. ex. A. DC (Ebenaceae): A Review. Molecules. 2023; 28(23):7759. https://doi.org/10.3390/molecules28237759

Chicago/Turabian Style

Ramadwa, Thanyani Emelton, and Stephen Meddows-Taylor. 2023. "Traditional Uses, Pharmacological Activities, and Phytochemical Analysis of Diospyros mespiliformis Hochst. ex. A. DC (Ebenaceae): A Review" Molecules 28, no. 23: 7759. https://doi.org/10.3390/molecules28237759

Article Metrics

Back to TopTop