Next Article in Journal
Antioxidative Defense, Suppressed Nitric Oxide Accumulation, and Synthesis of Protective Proteins in Roots and Leaves Contribute to the Desiccation Tolerance of the Resurrection Plant Haberlea rhodopensis
Next Article in Special Issue
Exploring Carob (Ceratonia siliqua L.): A Comprehensive Assessment of Its Characteristics, Ethnomedicinal Uses, Phytochemical Aspects, and Pharmacological Activities
Previous Article in Journal
Response of Flavor Substances in Tomato Fruit to Light Spectrum and Daily Light Integral
Previous Article in Special Issue
A Systematic Review on the Therapeutic Effects of Ayahuasca
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Genus Diospyros: A Review of Novel Insights into the Biological Activity and Species of Mozambican Flora

Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
Plants 2023, 12(15), 2833; https://doi.org/10.3390/plants12152833
Submission received: 26 June 2023 / Revised: 21 July 2023 / Accepted: 29 July 2023 / Published: 31 July 2023
(This article belongs to the Special Issue Medicinal Plants and Their Marker Compounds)

Abstract

:
Species of the Diospyros L. genus (Ebenaceae family) have been largely used in traditional medicine for the treatment of several diseases, especially infectious ones. To date, active major compounds such as naphthoquinones, triterpenoids, and tannins have been isolated and pharmacologically validated from Diospyros species. The present study summarizes the information available in the literature on the species described in the Flora of Mozambique. To do so, scientific databases (e.g., PubMed, Scopus, Web of Science, and Google Scholar) were searched using various keywords and Boolean connectors to gather and summarize the information. Of the 31 native and naturalized species in the Flora of Mozambique, 17 are used in different regions of Africa and were described for their traditional uses. They were reported to treat more than 20 diseases, mostly infectious, in the gastrointestinal and oral cavity compartments. This work provides an overview of the therapeutical potential of Diospyros species and explores novel insights on the antimicrobial potential of extracts and/or isolated compounds of these Mozambican species.

1. Introduction

The genus Diospyros L. (Ebenaceae family) contains species that have been recognized and used in traditional medicine (extended ethnomedical use) and have potential new health benefits supported by in vitro biological, in vivo pharmacological, and clinical tests [1,2,3,4]. Furthermore, within certain cultures or communities, various traditional systems have used all plant parts of this botanical genus (leaf, fruit, bark, twig, hardwood, and root) as herbal medicines [1,4].
Beyond their pharmacological value, Diospyros spp. have distinct and complementary important qualities, namely valuable wood, and edible fruits, which provide significant economic benefits and are recognized and utilized in various industrial and commercial sectors [1,4].
Generally, Diospyros spp. are tree shrubs or subshrubs with entire alternate leaves, solitary flowers, and fleshy fruits (berries) with usually two or more seeds. The characteristics of the leaves and flowers of these species are often used to identify fossil casts [5,6,7].
Diospyros species are predominantly distributed between the tropics, and the most notable diversity of this botanical genus occurs in Africa [5,6,8]. As confirmed in The Plant List [9], the WFO Plant List currently contains 1575 species related to the genus Diospyros, of which 734 have accepted scientific names [10]. Regarding the Mozambican flora, the genus is represented by 31 species (Table 1), corresponding to 18 accepted scientific name species, seven accepted subspecies (subsp.), three species that are considered synonyms, and three species that are not yet in the WFO plant list as of 12 February 2022 [10,11,12].
Concerning primary health care (PHC), herbal medicines are used by 80% of the African population, and more than 70% of the population of Mozambique uses such medicines for treating all diseases [13,14,15]. For instance, several Diospyros species with antimicrobial potential have been reported [4,16,17,18]. Worldwide, the magnitude of infectious diseases (ID), encompassing antimicrobial resistance (AMR), represents a major health problem (approximately 700,000 people die every year) [19,20]. Infectious diseases have a high impact in Africa, particularly in Mozambique [21].
Most of the native and naturalized Diospyros species of Mozambique’s flora are generally recognized as traditionally used in different regions of Africa to treat different diseases, with a particular focus on infections affecting the gastrointestinal tract and oral cavity. This work will present a comprehensive overview of the therapeutic potential of Mozambican Diospyros species based on chemical, biological, and toxicological experimental data, particularly addressing its antimicrobial properties and including comparative elements concerning the biological activity of other Diospyros species.

2. Results

2.1. Ethnomedical Use of Diospyros Species of Mozambican Flora

Table 2 shows the results of the collected ethnomedical data from seventeen Mozambican species, namely D. abyssinica, D. anitae, D. ferrea, D. kabuyeana, D. loureiriana subsp. loureiriana, D. lycioides subsp. sericea, D. mafiensis, D. mespiliformis, D. rotundifolia, D. mafiensis, D. mespiliformis, D. quiloensis, D. rotundifolia, D. squarrosa, D. usambarensis, D. verrucosa, D. villosa, D. villosa var. parvifolia, D. whyteana, and D. zombensis. In addition, information is given on the part of the plant used as medicine, the manufacturing process of the traditional formulation, the main traditional therapeutic use, and the country from which the information originates.
The results show that 54.8% of the total Diospyros species from Mozambique are referred to for their traditional use (Table 2). Among these, D. rotundifolia (Figure 1), traditionally used to treat diarrhea [22], is a prevailing species of dense undergrowth in the coastal area of the Marracuene District [23].
Furthermore, among the Diospyros species present in the Mozambican flora, D. villosa (Figure 2) is a species with a well-established traditional use of both leaf [24] and root [25]; the latter mainly used as a toothbrush for hygiene purposes [26].
Diospyros species have been reported to be used to treat the signals and symptoms of over 20 diseases. Two of these species (D. abyssinica and D. mespiliformis) have been mentioned most frequently and are used in two to five different countries in Africa (Table 2).
Based on the diverse description in the literature for the human use of the different parts of Diospyros, the results are grouped into infectious diseases (antibacterial, antifungal, anthelminthic, and antiviral); gastrointestinal (diarrhea, dysentery, emetic, flatulence, and other gastrointestinal disorders), oral cavity (oral hygiene, healing of oral wounds, and toothaches); urogenital (anti-hemorrhagic, dysmenorrhea, and infertility); skin diseases (dermatitis, fresh wounds, bedsores, and rashes); musculoskeletal (body pain, bruises, painful fractures, and rheumatism); and others conditions (diabetes, internal injuries, antidotes, hemostatic agents, and snake bites).
Among all the different Diospyros plant parts used in traditional medicine (Figure 3), the root is the most-used part (82%, Figure 3a) and is most used to treat infectious diseases. In the treatment of gastrointestinal disorders, it corresponds to 59%, for oral cavity infections, 41%, and for skin diseases, 18%, as well as for the management of other conditions, comprising 12% (Figure 3b).
The leaf is the second-most used part of the Diospyros species, but it is used in a similar percentage (18%) to the root to treat skin conditions and more commonly (24%) for musculoskeletal bruises, painful fractures, body aches, and rheumatism (Figure 3a,b).
Table 2. Reported ethnomedical use of Mozambican Diospyros species.
Table 2. Reported ethnomedical use of Mozambican Diospyros species.
SpeciesPart UsedPreparation MethodTraditional UseCountryRef.
D. abyssinica
leafdecoction malariaMali[4,27]
wound healing
fruit (dry)decoctionastringent and cholagogue
gastrointestinal disorders
barkunspecifiedastringent and antipyretic
root decoctionantihelminthic
abdominal pain, dysentery, and
diarrhea
leafjuicesnake bite Mali, Guinea
Zimbabwe
[28]
barkastringent
barkdecoctioninternal injuriesKenya[29]
laxative
rash
malaria and ringworm
leafsqueeze
and apply
ringworm Uganda[30]
seedwound healing
leafjuicetropical ulcer (skin and soft tissue polymicrobial infection, feet, or lower legs localized)[31]
tuberdecoction upset stomach[32]
D. anitae
rootunspecifieddental hygiene Mozambique[33]
healing of oral wounds
D. ferrea
fruitunspecifieddiarrhea and sore throatsIndia[34]
internal bleeding
renal lithiasis
rootunspecifiedanti-hemorrhagic [35,36]
infertility
bark oral hygiene
skin diseases
D. kabuyeana
rootunspecifiedantiviralTanzania[37]
D. loureiroana subsp. loureiroana
rootchewing stickoral hygiene South Africa
East Africa
[4]
D. lycioides subsp. sericea
root barkdecoctionbloody feces South Africa
South Central Zimbabwe
[38,39,40]
dysentery
headache
rootchewing stickinfertilityNamibia
Zambia
[41]
D. mafiensis
rootunspecifieddiarrheaMozambique
Tanzania
[42]
leprosy
skin diseases (including fungal infections)
D. mespiliformis
leafdecoctionanalgesic and antipyreticCentral
Southern
Eastern
Western
Africa
[27,43,44,45,46,47]
antihelminthic
dermatomycosis
fungal infections
induction of childbirth
hemostatic agent
malaria, pneumonia, and trypanosomiasis
sexually transmitted diseases
leaf and barkdecoctiondiarrhea and dysentery
leprosy
oral infections
whooping cough
leafdecoction bruises, bedsores, rash, and wounds
ringworm
rootchewing stickoral hygiene
leaf, bark
and root
decoctiontoothacheBurkina Faso[48]
leafdecoctionantipyreticGhana[27]
dermatitis
diarrhea and dysentery
malaria
fruitdecoctionheadache
pneumonia
rheumatism
stem barkdecoctionmalaria and pneumonia
rootdecoctioninfection with fever
leafdecoctionantipyreticNigeria[49,50,51]
antidote for a variety of poisonous substances
diarrhea and dysentery
haemostatic agent
oral infections
wound healing
rootdecoctionmalaria and oral candida infection
(used as mouthwash, management of HIV/AIDS opportunistic diseases)
Zambia[44,52]
rootinfusionabdominal pain, body and heart painSouth Central Zimbabwe[53]
seed unspecifiedantibacterialGuinea[4]
D. quiloensis
stem barkdecoctionmalariaZambia[44]
sexually transmitted diseases
D. rotundifolia
rootnot reportdiarrheaSouth African[22]
D. squarrosa
rootnot reportsexually transmitted diseasesTanzania[37]
D. usambarensis
root barkunspecifiedschistosomiasisMalawi[54]
rootchewing stick oral hygiene Tanzania[55,56]
decoctionfungal infections and overt symptoms of type 2 diabetes (i.e., polyuria, polydipsia, excessive thirst, and sweating)
D. verrucosa
rootunspecifiedleprosyTanzania[4,57]
D. villosa
leafunspecifiedgastrointestinal disordersSouth African [24]
painful fractures
rootdecoctiongastrointestinal disordersMozambique[4,25,58]
laxative
musculoskeletal system
toothbrush oral hygiene
decoctionwounds (skin/subcutaneous tissue)
D. villosa var. parvifolia
leafinfusionemeticSouth Africa[59]
rootantihelminthic
emetic and flatulence
gastrointestinal disorders
D. whyteana
rootunspecifiedantibacterial South Africa[60]
dysmenorrhea
rash
D. zombensis
root barkunspecifiedschistosomiasisMalawi[4,61]
The majority of documented medicinal uses of Diospyros species are attributed to their effectiveness in treating microbial infections, encompassing bacterial, fungal, and parasitic infections. These include conditions such as diarrhea, dysentery, and various skin and oral cavity infections.

2.2. Chemical Composition of Mozambican Diospyros Species

The main classes of chemical constituents identified in Diospyros species from the Mozambican flora are listed in Table 3.
The presence of phenolic acid derivatives, like flavonoids and naphthoquinones (NQs), particularly 1,4-naphthoquinones (1,4-NQs), and terpenoids, mainly triterpenoids (especially lupan, ursane, oleanane derivatives) [3,4,17,62,63] and tetraterpenoids (carotenoids), have been reported [4]. Other chemical constituents in these Diospyros species include hydrocarbons, lipids, amino acids, and sugars [1,4,5,62].
Table 3. Chemical compounds identified in Mozambican Diospyros species.
Table 3. Chemical compounds identified in Mozambican Diospyros species.
SpeciesPart UsedChemical ClassCompoundsExtractRef.
D. abyssinica
root barknaphthoquinoneplumbagin (2-methyl-5-hydroxy-1,4-naphthoquinone)P. ether, CF, DCM, H2O,
MeOH,
EtOH 80%
[28]
stem barknaphthoquinonediospyrin, isodiospyrin[64,65]
leaftriterpenoidbetulinic acid, betulin and lupeolMeOH
D. consolatae
n.rtriterpenoidbetulinic acid, betulin and lupeoln.r[4]
n.rnaphthoquinonediosindigo An.r[4]
D. dichrophylla
seednaphthoquinoneisodiospyrinHex[66]
D. ferrea
leaftriterpenoidpregnenolone and androstan-6-oneMeOH[67]
n.rβ-sitosteroln.r[4]
leafmonoterpenoidcitronellolMeOH[67]
leafditerpenoidphytolMeOH[67]
thunbergolEtOAc
leaftriterpenoidbetulin, α-amyrin,
friedelan-3-one and olen-12-ene
EtOAc[67]
fruittriterpenoidfriedelin, epifriedelinol, lupeol, lupenone, and betulin n-Hex[68]
fruittriterpenoidβ-sitosterol and stigmasteroln-Hex[68]
root
fruit
naphthoquinone7-methyljuglone, isodiospyrin, diosindigo A
and 8-hydroxyisodiospyrin
CF, n-Hex[68,69]
rootphenolgallic acidEtOH[70]
leaftriterpenoidfriedelin, friedelin-3-ol, taraxerol and taraxeroneEtOH[4]
n.rtriterpenoidursolic acidn.r[4]
D. inhacaensis
stemnaphthoquinone7-methyljuglone and diospyrinn.r[71]
D. kirkii
n.rtriterpenoidbauerenol, betulin and lupeoln.r[4]
n.r β-sitosteroln.r[4]
n.rnaphthoquinonediosindigo An.r[4]
D. lycioides
branchenaphthalene Diospyroside A, B, C and DMeOH[72]
naphthoquinone7-methyljuglone and jugloneMeOH[41]
triterpenoidlupeol and ursolic acidn.r[53]
fruitnaphthoquinoneisodiospyrin and bisisodiospyrinn.r[71]
root, stemnaphthoquinone7-methyljuglone and isodiospyrinCF[71]
n.rnaphthoquinonemamegakinone, methylnaphthazarin
and 8-hydroxyisodiospyrin
n.r[4]
D. mafiensis
root barknaphthoquinonediosquinone, diosindigo A, 7-methyljuglone,
3-hydroxiquinone, and 6,8-bisdiosquinone
CF, DCM,
MeOH
[42,73,74]
stem barknaphthoquinone7-methyljuglone and diosindigo A [73]
leaftriterpenoidα-amyrin, lupeol and betulinic acidCF, MeOH[75]
barknaphthoquinonediosquinone, isodiospyrin, and plumbaginEe[4,52]
stem barktriterpenoidlupeol, betulin, betulinic acid, α-amyrin, and bauerenolCF[4,76]
D. mespiliformis
stem bark,
leaf, bark
triterpenoidbetulinic acid, betulin, lupeol, bauerenol, and α-amyrinCF,
MeOH
[4,76]
leafflavonoid7-O-(4″′-O-acetyl)-allopyranosyl(1″′ → 2″)-
β-glucopyranoside, along with eight flavonoid metabolites—luteolin 3′,4′,6,8-tetramethyl ether, luteolin 4′-O-β-neohesperidoside, luteolin 7-O-β-glucoside, luteolin, quercetin, quercetin 3-O-β-glucoside, quercetin 3-O-α-rhamnoside, and rutin
n.r[77]
rootnaphthoquinonediosquinone, and plumbaginP. ether[78]
root, barknaphthoquinonediospyrinMeOH[79]
fruitnaphthoquinoneplumbaginMeOH[79]
D. natalensis
root, stemnaphthoquinone7-methyljuglone, and diospyrin n.r[4]
n.rtriterpenoidbetulinic acid, α-amyrin, and lupeoln.r[4]
n.rfatty acidheptacosanoic acidn.r[4]
D. quiloensis
n.rnaphthalene4,5,6,8-tetramethoxy naphthaldhyde,
5-hydroxy-4,6,8-trimethoxy naphthaldehyde,
4,5,6-trimethoxynaphthalehyde,
4,5-dimethoxynaphthaldehyde, and
5-hydroxy-4-methoxy-2-naphthaldehyde
MeOH[4]
D. rotundifolia
n.rtriterpenoidbetulin and lupeoln.r[4]
root naphthoquinone7-methyljuglone, neodiospyrin and rotundiquinone n.r[71]
stemnaphthoquinone7-methyljuglone and diospyrin n.r[71]
D. squarrosa
n.rnaphthoquinone7-methyljuglone n.r[4]
D. usambarensis
root naphthoquinone7-methyljuglone, isodiospyrin, diosindigo A and B, bis-isodiospyrin and mamegakinone MeOH[54,80]
stem barknaphthoquinone7-methyljuglone and diosindigo AMeOH[54]
D. verrucosa
root barknaphthoquinonediosindigo A, 7-methyljuglone, diosquinone and isodiospyrinn.r[57]
root barktriterpenoidbetulinic acid and betulin
stem barknaphthoquinonediosindigo A, 7-methyljuglone, diosquinone and isodiospyrinn.r[57]
stem barktriterpenoidbetulinic acid and betulinn.r[57]
D. whyteana
n.rnaphthoquinone7-methyljuglone n.r[4]
D. zombensis
barktriterpenoidoleanolic acidMeOH[4]
root barknaphthoquinone7-methyljuglone, diosquinone, isodiospyrin and mamegakinonaP. ether, MeOH[4,61]
Extract: Ace—acetone; CF—chloroform; DCM—dichloromethane; Ee—ether; EtOAc—ethyl acetate; EtOH—ethanol; H2O—water; Hex—hexane; MeOH—methanol; P. ether—petroleum ether; n.r—not reported.
Among the NQs (Figure 4), 80% are 1,4-NQs, either as monomers such as plumbagin (1) and 7-methyljuglone (2) or as dimers such as diospyrin (3) and isodiospyrin (4), while trimers and tetramers are less represented in this genus [4,81].
In the Mozambican Diospyros species, plumbagin (1) and 7-methyljuglone (2) are the most prominent 1,4-NQs identified [3,4]. The presence of 7-methyljuglone has been reported in diethyl ether, dichloromethane, chloroform, methanol, and hydroethanol extracts of the root, stem, and bark of most species [1,5] and in the ether extract of D. lycioides branches [41].
Plumbagin has been identified on the root bark of D. abyssinica [28], and isodiospyrin (4), a dimeric 7-methyljuglone derivative [3], has been reported in a hexane extract of D. dichrophylla seeds [66] and in the diethyl ether extract of bark and phylum of almost all Mozambican Diospyros species [4].
D. mespiliformis has been one of the best-studied Mozambican Diospyros species, having NQs identified in different plant parts [4,79] and triterpenoids in leaf, bark, and stem bark [4,76,82].
Triterpenoids (lupane, ursane, oleanane, taraxerane, and friedelane) are present in more than 90% of Diospyros species. Lupane-type compounds (Figure 5), such as betulinic acid (1, Figure 5), betulin (2, Figure 5), and lupeol (3, Figure 5), are the most active substances present in Diospyros African species [4,64,83,84]. These compounds were detected in different types of extracts (petroleum ether, dichloromethane, chloroform, methanol, hydroethanol, and aqueous extracts) and their fractions [1,5,28,41]. Several biological activities have been demonstrated for them, mainly for betulinic acid and its derivatives [83,85,86,87,88].
Condensed tannins (proanthocyanidins and oligopolymeric complex tannins), and particularly hydrolysable tannins (gallotannins, ellagitannins), and have also been identified in Mozambican Diospyros species such as D. villosa [4,25,58] and D. mespiliformis [82,89].
In addition, from the methanolic extract derived from D. lycioides twigs, three naphthalene glycosides were identified [72], and carotenoids were identified in the fruit of this species [90]. The presence of galactiol and vitamin E in the D. ferrea leaf was also reported [67].
So far, the biologically active marker secondary metabolites isolated and studied from several species of the genus Diospyros have mainly been naphthoquinones, triterpenoids, and tannins. Compounds belonging to these chemical classes have been isolated from the twigs, bark, roots, leaves, stems, and fruits of Mozambican species of this genus. Examples include plumbagin, 7-methyljuglone, diospyrin, and isodiospyrin, which have been isolated from the root of several Diospyros species.

2.3. In Vitro and In Vivo Biological Activity of Mozambican Diospyros Species and Marker Compounds

In Table 4, Table 5 and Table 6, the different in vitro and in vivo biological activities and toxicological tests performed on Mozambican Diospyros species, and their isolated marker secondary metabolites are summarized. A total of thirteen species (41.9%), namely D. abyssinica, D. bussei, D. ferrea, D. kabuyeana, D. lycioides, D. loureiriana, D. mafiensis, D. mespiliformis, D. natalensis, D. squarrosa, D. usambarensis, D. verrucosa, and D. villosa, were evaluated for biological activities other than antibacterial activities (Table 4).

2.3.1. Anti-Inflammatory and Analgesic Activity

Aqueous extract of D. abyssinica root bark has shown stronger anti-inflammatory activity (enzyme 15-lipoxygenase (LOX) inhibition) than quercetin [27].
In vivo assays have shown that the hexane fraction of D. mespiliformis leaves has anti-inflammatory properties (inhibits stronger the LOX), and that the methanolic extracts of different plant parts showed wound healing effects. On the other hand, the butanol and ethyl acetate fractions activate LOX activity. These results show that D. mespiliformis extract can have pro-inflammatory and anti-inflammatory effects [51].
Lupeol isolated from D. mespiliformis stem bark has shown analgesic activity in both pain inhibition (neurological-first phase) and origin (inflammatory-second phase) in biphasic tests (in vivo) [76].

2.3.2. Antihyperglycemic Activity

Another finding has revealed that the oral administration of a methanolic extract obtained from the leaves of D. ferrea (400 mg/kg) for a duration of 21 days in diabetic rats showed significant antihyperglycemic activity [91]. The root of this species is rich in phenolic acids, especially gallic acid, and is therefore traditionally used as a potent antioxidant [70].

2.3.3. Antifungal Activity

Several studies have reported the potential antifungal activity of the root and root bark of most Diospyros species [42,54,92]. However, the antifungal activity of a leaf extract of D. mespiliformis has also been confirmed [47,93].
Various Diospyros medicinal plants are also effective against Candida spp. [1]. The methanolic extract of the D. abyssinica root is active against this microorganism [94]; however, in another study, it was only moderately active against the same microorganism [95]. Another medicinal plant, D. mespiliformis, is more active against C. neoformans than against C. albicans. A leaf extract showed anti-C. albicans activity, while a bark extract showed in vitro activity against C. neoformans-isolated strains from South African AIDS patients [96].
D. mespiliformis, traditionally used to treat ringworm, shows remarkable antimicrobial activity against Trichophyton mentagrophytes and Microsporum canis. This result supports the traditional use of this species against dermatophytosis [47]. Aqueous and ethanolic extracts of the leaf and bark of D. mespiliformis showed significant antifungal activity against Aspergillus niger, Aspergillus flavus, and Microsporum gypseum [97].

2.3.4. Antiparasitic Activity

Diospyros species have antiparasitic activity, especially against both chloroquine-sensitive (3D7) and chloroquine-resistant (FcB1) strains of Plasmodium falciparum [31,94].
The decoction of the stem of D. mespiliformis was tested against Plasmodium berghei-infected mice and demonstrated potent activity, including the inhibition of beta-hematin in an in vitro study [98].
In vitro studies from methanolic extracts of D. abyssinica leaves have provided confirmation of its antiparasitic activity against Leishmania donovani [65,94], Trypanosoma cruzi, Trypanosoma brucei [99], Culex, and Anopheles larvae [94].
The isolated compound 7-methyljuglone obtained from the methanolic extract of D. usambarensis root bark has significant schistosomicidal activity [54,92].

2.3.5. Antioxidant Activity

The scavenging activity of crude extract and fractions of four Diospyros species, namely D. abyssinica, D. lycioides, D. mespiliformis, and D. villosa, present in the Mozambican Flora was evaluated spectrophotometrically using the DPPH (1,1-diphenyl-2-picrylhydrazyl) radical assay.
An estimation of the concentration of antioxidant vitamins (i.e., A, C, and E) from crude methanolic extracts obtained from the leaf, bark, and root of D. mespiliformis was also determined using the DPPH [51].
Table 4. In vitro and in vivo non-antibacterial tests of biological activity in Mozambican species of Diospyros and marker compounds.
Table 4. In vitro and in vivo non-antibacterial tests of biological activity in Mozambican species of Diospyros and marker compounds.
Biological Activity/
Species
PUExtract/
Compound
Results Microorganism/
Assay
Control Ref.
Analgesic
D. mespiliformisSB CF/lupeol
25 mg/kg, p.o
Pi1 2.2 ± 0.2/ asa =1.0 ± 0.3
Pi2 1.98 ± 0.1/ asa =1.15 ± 0.1
Biphasic, Wistar rats acetylsalicylic acid (asa), 100 mg/kg, p.o.[76]
D. ferreaLCF
MeOH
100–300 mg/Kg
significant activity
Tail flick method, adult Wistar albino ratsibuprofen[100]
D. ferreaRCF
MeOH
100–200 mg/Kg
significant activity
Tail flick method, adult Wistar albino ratsibuprofen[101]
Anti-inflammatory
D. abyssinicaRbH2O (1)
MeOH (2)
1—IC50 = 16 ± 1 μg/mL
2—IC50 = 86 ± 7 μg/mL
LOX, using soybean lipoxygenase type 1-Bquercetin, IC50 value
11.5 ± 0.6 μg/mL
[27]
D. ferreaLCF
MeOH
100–300 mg/Kg = 26.2–28.2%
100–300 mg/Kg = 29.6–37.6%
PIPE, adult male Wistar ratsibuprofen
41.1%
[100]
D. ferreaRCF
MeOH
100–200 mg/Kg = 37% PIPE, adult Wistar albino ratsibuprofen[101]
D. mespiliformisSbDCM Fraction
maximally
at 400 mg/kg
Modulation of serum
concentrations of Tumour
Necrosis Factor alpha and
Interleukin 1 beta and 6
Cytokine inhibition, Plasmodium berghei-infected miceartemether-lumefantrine[98]
LHex Fraction
5 μg/mL(1)
10 μg/mL (2)
1—IC50 = 31.21 ± 0.84 μg/mL
2—IC50 = 32.05 ± 2.79 μg/mL
LOX, Wistar ratsquercetin, IC50 value
1–46.02 ± 5.46 μg/mL
2–32.05 ± 2.79 μg/mL
[51]
Antihyperglycemic
D. ferreaLMeOH
21 days
400 mg/kg, i.p, significant
antihyperglycemic activity
Streptozotocin induced diabetic Wistar ratsglibenclamide,
0.5 mg/Kg, p.o.
[91]
Antifungal
D. abyssinicaRMeOHActives in test controlled by conidial suspension BA, C. albicans
C. cucumerinum
methylthiazolyltetrazolium chloride (MTT)[94]
D. ferreaW1-isodiospyrin
2-plumbagin
1—active against three fungi
2—active against eight fungi
HMBCPhomopsis sp.
reference spectrum for both H1 and C13
[102]
D. mafiensisRb3-hydroxy-
diosquinone
MIC50 = 14.9 µg/mL
MIC50 = 39.1 µg/mL
CCA, A. flavus, A. parasiticusA. parasiticus B62[42]
Rb3-hydroxy-
diosquinone
Reduced total aflatoxin,
1.145 to 32 ng/plac
ELISA, A. parasiticus,
A. flavus
A. parasiticus B62[42]
RbdiosquinoneMIC50 >100 µg/mLCCA, A. flavus, A. parasiticusA. parasiticus B62[42]
RbdiosquinoneReduced total aflatoxin
1.145 to 45 ng/plac
ELISA, A. flavus, A. parasiticusA. parasiticus B62[42]
RbP. ether, DCM (E) Fraction (F)E = 5 mg/disc IZ: 7–20 mm
F = 0.2 mg/disc IZ: 19–20 mm
DD, C. albicansmiconazole
20 µg/disc
IZ: 29 mm
[103]
D. mespiliformisRb
L
AceMIC = 0.16 μg/mLBD, C. albicans, M. canisamphotericin B
MIC = 0.02 μg/mL
[93]
LDCM:MeOHMIC = 0.10–0.50 mg/mLBD, M. canis, T. mentagrophytes tetrazolium violet[47]
LH2OMIC = 0.08 μg/mLBD, M. canisamphotericin B
MIC = 0.02 μg/mL
[93]
BAceIZ: 7 mm (1)
IZ: 12 mm (2)
ADD, 1-C. albicans,
2-C. neoformans
nystatin[96]
D. usambarensisRb7-methyljugloneMIC = 0.025 μg/mLBA, C. cucumerinum
miconazole
MIC = 0.001 μg/mL
[92]
RbisodiospyrinMIC = 10 μg/mLBA, C. cucumerinum
miconazole
MIC = 0.001 μg/mL
[54]
D. villosaREtOH 70%
Fraction
MIC = 312.5 μg/mL
MIC = 62.5–312.5 μg/mL
BD, C. albicansnot reported[104]
Antiparasitic
D. abyssinicaLEtOAcIC50 = 51.3 ± 8.8 μg/mLBD, P. falciparum (FcB1)chloroquine[31]
BEtOAcIC50 = 1.5 μg/mLL. donovanipentamidine[65]
IC50 = 5.6 μg/mLP. falciparumchloroquine
Bdiospyrin
isodiospyrin
IC50 = 0.5 μML. donovanipentamidine
IC50 = 7 μM
[94]
Bdiospyrin
isodiospyrin
IC50 = 1.5 μMP. falciparum (FcB1)chloroquine
IC50 = 0.1 μM
[94]
RDCM
MeOH
MIC = 500 mg/LCulex, Anopheles larvaenot identified [94]
D. busseiRMeOHIC50 = 65.7 ± 2.7 μg/mLT. brucei (Lister 427)pentamidine
IC50 = 0.000509 μM
[99]
D. kabuyeanaLMeOHIC50 = 3.32 μg/mLT. brucei (Lister 427)pentamidine
IC50 = 0.000509 μM
[99]
D. loureirianaRb
Sb
L
MeOHIC50 = 1.68 ± 0.77 μg/mL
IC50 = 11.53 ± 1.99 μg/mL
IC50 = 19.10 ± 4.41 μg/mL
P. falciparum (3D7)chloroquine
IC50 = 0. 0045 μM
[105]
[105]
[105]
D. mespiliformisSEtoAC (1)
DCM (2)
MeOHfraction (3)
1—IC50 = 3.18 μg/mL
2—IC50 = 0.78 μg/mL
3—IC50 = 0.55 μg/mL
Plasmodium berghei-infected miceartesunate and
chloroquine
diphosphate
[98]
D. natalensisSbMeOHIC50 = 2.85 μg/mLT. brucei (Lister 427)pentamidine
IC50 = 0.000509 μM
[99]
D. squarrosaRbMeOHIC50 = 5.38 μg/mLT. brucei (Lister 427)pentamidine
IC50 = 0.000509 μM
[99]
D. verrucosaSbMeOHIC50 = 1.28 μg/mLT. brucei (Lister 427)pentamidine
IC50 = 0.000509 μM
[99]
RMeOHIC50 = 2.23 μg/mLT. brucei (Lister 427)
LMeOHIC50 = 2.99 μg/mLT. brucei (Lister 427)
D. usambarensisRb7-methyljugloneEfficiency schistosomiasis
MIC = 5 ppm
Biomphalaria glabratanot identified[54]
Antioxidant
D. abyssinicaRbEtOH (1)
MeOH (2)
H2O (3)
1-EC50 = 16.0 ± 2 μg/mL
2-EC50 = 16.6 ± 0.4 μg/mL
3-EC50 = 21 and 29 ± 2 μg/mL
DPPH quercetin
EC50 value
3.4 ± 0.3 μg/mL
[27]
D. lycioidesLAce𝑅𝑓 = 0.54; 0.60; 0.83; 0.89 DPPH on TLC platesphenolic
compounds
[38]
D. mespiliformisFMeOH87.36% at 1 mg/mLDPPH vitamin E[106]
RMeOH IC50 = 3.47 ± 0.05 μg/mLDPPH ascorbic acid
2.36 ± 0.30 μg/mL
trolox
3.43 ± 0.78 μg/mL
[51]
FMeOHIC50 = 6.94 ± 0.49 μg/mLDPPH [51]
BMeOHIC50 = 7.82 ± 0.76 μg/mLDPPH [51]
LEtOAc Fraction IC50 = 1.08 ± 0.04 μg/mlDPPH ascorbic acid
5.08 ± 0.12 μg/mL
[51]
D. villosaSbMeOHIC50 = 9.53 μg/mLDPPH ascorbic acid
10.3 μg/mL
[107]
LCF (1)
Hex (2)
1-IC50 = 10.7 μg/mL
2-IC50 = 11.8 μg/mL
DPPH ascorbic acid
10.3 μg/mL
[107]
Part used (PU): L—leaf; B—bark; F—fruit; R—root; Rb—root bark; Sb—stem bark. Extract: Ace—acetone; ADD—agar disc diffusion; CF—chloroform; DCM—dichloromethane; EtOAc—ethyl acetate; EtOH—ethanol; H2O—water; Hex—hexane; MeOH—methanol; P. ether—petroleum ether. Test: BA—TLC bioautography; BD—broth dilution; CCA—cell culture in agar; DD: disco diffusion method; DPPH—2,2-diphenyl-1-picrylhydrazyl; ELISA—enzyme-linked immunosorbent assay; HMBC—heteronuclear multiple-bond correlation method; PIPE—percent inhibition of paw edema. Abbreviations: LOX-15-lipoxygenase; Pi1—pain inhibition (neurological-first phase); Pi2—pain inhibition (inflammatory-second phase); EC50—half maximal effective concentration; IC50—half maximal inhibitory concentration; MIC—minimum inhibitory concentration.

2.3.6. Cytotoxicity, Genotoxicity, and Toxicity of Mozambican Diospyros Species

The results of in vitro cytotoxicity tests using normal and tumorous human cells and Artemia salina, as well as in vitro genotoxicity and in vivo acute and sub-chronic toxicity assessment of Diospyros species, are summarized in Table 5.
Table 5. In vitro cytotoxicity and genotoxicity studies as well as in vivo toxicity studies in Mozambican Diospyros species.
Table 5. In vitro cytotoxicity and genotoxicity studies as well as in vivo toxicity studies in Mozambican Diospyros species.
SpeciesParts UsedExtractToxicity AssayResultsRef.
D. abyssinica
leafEtOAcCytotoxicity against
MRC-5 human diploid embryonic cells, Taxotere® as standard
IC50 = 6.0 ± 5.0 μg/mL[31]
leafEtOAc
Cytotoxicity against KB human tumor cell lines (squamous cell carcinoma
of the mouth), Taxotere® as standard
>85% cell inhibition
IC50 = 1.0 ± 2.0 μg/mL
[31]
barkEtOAcCytotoxicity against human KB cell (1)
and Rhabditis pseudoelongata (2)
(1) LD50 = 10 μg/mL
(2) LD50 = 1 μg/mL
[65]
D. dichhropylla
seedDCM:MeOH (1)
isodiospyrin (2)
Cytotoxicity using Brine shrimp test
(Artemia salina)
1-(LC50 = 29 μg/mL)
2-(LC50 = 0.13 μg/mL)
[66]
D. ferrea
leafMeOHIn vivo—acute oral toxicity using
male Wistar albino rats
LD50 = 2000 mg/kg[91]
fruitisodiospyrin (1)
8′-hydroxyisodiospyrin (2)
Cytotoxicity strong against
Hep-3B, KB, COLO-205, and HeLa
cancer cells
1(ED50 = 0.17, 1.72, 0.16 and 0.21 μg/mL)
2(ED50 = 1.31, 1.75, 1.96 and 1.79 μg/mL)
[68]
D. lycioides
leafAceCytotoxicity against BUD-8 cell (human
fibroblast cells) in real-time xCELLigence system and 7.4 μg/mL curcumin (control)
IC50 = 500 and 1000 μg/mL[38]
leafAceCytotoxicity against HeLa cells
mobility assayed using the wound healing assay and 7.4 μg/mL curcumin (control)
Nontoxic to the normal
cell at 300 μg/mL
[38]
D. loureiriana
root barkMeOHCytotoxicity against
human embryonic kidney cells (HEK293),
estimated growth inhibition at 400 μg/ml
IC50 = 100.34 ± 9.85 μg/mL[105]
stem barkIC50 = 57.26 ± 0.53 μg/mL[105]
D. mafiensis
root barkP. ether (1)
DCM (2)
EtOH (3)
fraction P. ether (4)
fraction DCM (5)
Cytotoxicity using
brine shrimp larvae test
(Artemia salina)
Standard cyclophosphamide
LC50 value of 17.78 µg/mL
1-LC50 = 25.12 µg/mL
2-LC50 = 69.18 µg/mL
3-LC50 = 120.23 µg/mL
4-LC50 ≤ 8–45.71 µg/mL
5-LC50 = 5.08 µg/mL
[103]
D. mespiliformis
stem bark root barkEtOH
In vivo—acute oral toxicity
using Wistar rats of both sexes
LD50 = 570 mg/kg
Acute toxicity is moderate
[49]
leafMeOHin vivo—acute oral
administration using rats
LD50 ≥ 5 g/kg[108,109]
stem bark
leafEtOAc fractionIn vivo—sub-chronic toxicity using ratsLD50 = 750 g/kg[108]
stem barkLD50 = 500 g/kg
root diosquinoneCytotoxicity against human
glioblastoma cell lines (1) and hormone-dependent human prostate cancer (2)
1-ED50 = 0.18 μg/mL
2-ED50 = 4.50 μg/mL
[84]
D. whyteana
twigsDCMGenotoxicity against
mutagens mitomycin C (MMC) using the Ames test (Salmonella typhimurium TA98)
protective effect
non-genotoxic at
500–2500 μg/mL
[60]
leafDCM
HydroMeOH 90%
Genotoxicity using the Ames test (Salmonella typhimurium TA98)shift mutations of
lowest dose is 0.50 μg/mL higher doses are toxic
[110]
D. villosa
rootHydroEtOH 70%In vivo—acute toxicity using micepossible renal
dysfunction development
[58]
D. zombensis
root bark7-methyljuglone (1) isodiospyrin (2) Cytotoxicity against
human colon carcinoma cells
1-LD50 of 7.0 × 10−2 μg/mL
2-LD50 of 3.8 × 10−2 μg/mL
[61]
Extracts: Ace—acetone; DCM—dichloromethane; EtOAc—ethyl acetate; EtOH—ethanol; H2O—water; Hex—hexane; HydroEtOH—ethanol; HydroMeOH—methanol; MeOH—methanol; P. ether—petroleum ether. Concentration: ED50—median effective dose; IC50—half maximal inhibitory concentration; LC50—lethal concentration 50%, LD50—lethal dose 50%.
Most commonly, studies were found to be related to the in vitro assessment of cytotoxicity. For example, the extract of D. lycioides showed cytotoxicity to HeLa cells but was non-toxic to normal cells [38]. The compound diosquinone has been shown to be toxic against most cancer cell lines (human glioblastoma) and hormone-dependent human prostate cancer [84]. In contrast, 7-methyljuglone and isodiospyrin compounds are active against human colon carcinoma cells [61].
The organic extract of the inner seed of D. dichrophylla (Figure 6) is reported as highly cytotoxic (LC50 = 29 μg/mL), particularly the isodiospyrin isolated from it (LC50 = 0.13 μg/mL) [66].
Preclinical safety assessments of Diospyros species are of paramount importance; however, few studies related to Mozambican Diospyros species have been conducted to date. Cantrell et al. (2003) reported that D. dichrophylla is a potent phytotoxicant due to the presence of isodiospyrin (from the inner seed) at a lethal dose of 0.13 g/mL [66]. In another study, a hydroethanolic root extract of D. villosa showed possible development of renal dysfunction using an acute toxicity test in mice [111].

2.3.7. Antibacterial Activity

In vitro antibacterial activity data collected from eleven Diospyros species (representing 35.5% of the total) are summarized in Table 6. Of the 11 species examined, 47 extracts (including AgNPs) showed antimicrobial activity against multiple bacterial strains. The methanolic extract was the most tested. In some of the studies mentioned, biodirected fractionation was also performed, and the antibacterial activity of the obtained fractions and isolated compounds was determined. The results obtained are also shown in Table 6.
Table 6. In vitro antibacterial activity of Mozambican Diospyros and marker compounds.
Table 6. In vitro antibacterial activity of Mozambican Diospyros and marker compounds.
SpeciesParts UsedTestExtract/
Compound
MIC (μg/mL)MicroorganismControl (MIC)
μg/mL
Ref.
D. abyssinica
barkBDEtOAc12S. aureus ATCC 6538DMSO [65]
D. bussei
leaf
stem bark
BDMeOH125E. coli ATCC 8740ciprofloxacin 0.63[99]
leafBDMeOH8000S. aureus ATCC 25923ciprofloxacin 2.5
B. cereus ATCC 11775ciprofloxacin 0.08
root barkBDMeOH500E. coli ATCC 8740ciprofloxacin 0.63
D. kabuyeana
leafBDMeOH8000S. aureus ATCC 25923ciprofloxacin 2.5[99]
leafBDMeOH4000B. cereus ATCC 11775ciprofloxacin 0.08
stem barkBDMeOH1000
leaf
stem bark
BDMeOH125E. coli ATCC 8740ciprofloxacin 0.63
D. lycioides
brancheBDMeOH1250S. sanguis, P. gingivalis,
S. mutans, P. intermedia
alkaloid sanguinarine[41]
brancheBDDiospyroside A39S. sanguis, P. intermediaalkaloid sanguinarine[41]
78–1250P. gingivalis, S. mutans
brancheBDDiospyroside B39–78S. sanguis, P. gingivalisalkaloid sanguinarine[41]
156–625P. intermedia, S. mutans
brancheBDDiospyroside C39–156P. intermedia, S. mutansalkaloid sanguinarine[41]
312–625P. gingivalis, S, sanguis
branche BDDiospyroside D156–312S. mutans, P. intermedia, P. gingivalis, S. sanguisalkaloid sanguinarine[41]
brancheBDjuglone19–78P. intermedia, S. mutans,alkaloid sanguinarine[41]
39S. sanguis, P. gingivalis
brancheBD7-methyljuglone39–156P. gingivalis, S. mutansalkaloid sanguinarine[41]
78S. sanguis, P. intermedia
leafBAEtOAc0.10–0.16 *P. aeruginosa
ATCC 27853
p-iodonitrotetrazolium chloride[38]
Ace0.12–0.17 *
leafBAEtOAc0.16–0.36 *S. aureus
ATCC 29213
p-iodonitrotetrazolium chloride[38]
Ace0.20–0.45 *
MeOH0.16–0.27 *
leafBAEtOAc0.05–0.45 *E. faecalis
ATCC 29212
p-iodonitrotetrazolium chloride[38]
Ace0.05–0.45 *
MeOH0.05–0.45 *
D. mafiensis
root bark DCMS. aureus
B. anthracis
IZ: 12 mm
S. typhi, S. boydii,
E. coli, K. pneumoniae
S. aureus, V. cholerae
Proteus sp., B. anthracis
gentamycin ampicillin
(20 µg/disc)
[103]
root bark P. ether-FractionIZ: 10–15 mmS. typhi, S. boydii,
E. coli, K. pneumoniae
S. aureus, V. cholerae
Proteus sp., B. anthracis
gentamycin ampicillin
(20 µg/disc)
[103]
D. mespiliformis
leafADDMeOH167S. aureusisoniazid 5.0[50]
rootADDMeOH250S. aureusisoniazid 5.0[50]
leafBDEtOH12,500–25,000 Salmonella spp.,
Shigella spp.,
Campylobacter spp.
ciprofloxacin, cefixime, and gentamicin[95]
leafBDHex (F1)
nBOH (F2)
EtOAc (F3)
H2O (F4)
(1)78.125–312.5
(2)156.25
(3)78.125–156.25
(4)625–2500
1-P. aeroginosa
2-S. aureus
3-E. coli
4-S. typhimurium
gentamicin 19.53
gentamicin 19.53
gentamicin 19.53
gentamicin 19.53
[97]
leaf 625
root625 (1)
>2500 (2 to 4)
leafADH2O
HydroMeOH 10%
250–500
125–500
H. influenzae (6 ci)ampicillin 0.12–15.6[112]
H2O
HydroMeOH 10%
125–250
62.5–125
S. aureus (5 ci)ampicillin 0.06–0.12
H2O
HydroMeOH 10%
250–250
125–125
S. pneumoniae (3 ci)ampicillin 0.015–0.12
H2O
HydroMeOH 10%
250–250
125–125
S. pyogenes (8 ci)ampicillin 0.015–0.06
H2O
HydroMeOH 10%
250–500
125–250
M. catarrhalis (5 ci)ampicillin 0.12–1.9
leafBDflavonol
O-rhamnoside
9770S. aureusnot identified[77]
rootADdiosquinone3–30S. aureus NCT 6571
S. aureus E3T
ampicillin 5[78]
diosquinone15–16E. coli KL16
P. aeruginosa NCT 6750
gentamicin 2
leaf DD methylated
flavone
IZ: 34 mm E. colinot identified [78]
leaf AWD EtOH-Fraction IZ: 20 mm
IZ: 18 mm
IZ: 16 mm
S. aureus, Shigella spp.
P. aeruginosa
septrin 15 mm
spetrin 16 mm
spetrin 15 mm
[113]
D. natalensis
leafBDMeOH250S. aureus ATCC 25923ciprofloxacin 0.08[99]
leafMeOH1000B. cereus ATCC 11775ciprofloxacin 2.5
leafMeOH500E. coli ATCC 8740ciprofloxacin 0.63
root barkMeOH1000E. coli ATCC 8740ciprofloxacin 0.63
stem barkMeOH250E. coli ATCC 8740ciprofloxacin 0.63
D. rotundifolia
not
reported
Ace230–1770S. aureus, E. faecalis,
E. coli and P. aeruginosa
not reported[22]
D. squarrosa
leafBDMeOH4000B. cereus ATCC 11775ciprofloxacin 2.5[99]
MeOH250E. coli ATCC 8740ciprofloxacin 0.63
root barkBDMeOH1000S. aureus ATCC 25923ciprofloxacin 0.08
MeOH4000B. cereus ATCC 11775ciprofloxacin 2.5
stem barkBDMeOH500E. coli ATCC 8740ciprofloxacin 0.63
D. verrucosa
leafBDMeOH1000S. aureus ATCC 25923ciprofloxacin 0.08[99]
MeOH2000B. cereus ATCC 11775ciprofloxacin 2.5
MeOH500E. coli ATCC 8740ciprofloxacin 0.63
root bark
stem bark
BDMeOH<6.25E. coli ATCC 8740ciprofloxacin 0.63[82]
D. villosa
rootBDHydroEtOH 70%
Ee
Fractions
62.5–312.5
15.6–62.5
31.2–62.5
E. faecalis ATCC 435628not reported[104]
E. coli ATCC 25922not reported
M. luteus ATCC 10240not reported
S. aureus ATCC 25923not reported
leafDDAgNPsIZ: 15 mmE. coli ATCC 25922ciprofloxacin 37 mm
gentamicin 20 mm
[107]
AgNPs 80 °CIZ: 18 mmS. aureus ATCC 700698ciprofloxacin 6 mm
gentamicin 11 mm
AgNPsIZ: 16 mmS. epidermidis
ATCC 12228
AgNPsIZ: 16 mmciprofloxacin 28 mm
gentamicin 20
Ace0.05–0.45 *
MeOH0.05–0.45 *
Test: BD—broth dilution; DD—disc diffusion; ADD—agar disc diffusion; AWD—agar well diffusion. Extract: Ace—acetone; DCM—dichloromethane; Ee—ether; EtOAc—ethyl acetate; EtOH—ethanol; H2O—water; Hex—hexane; HydroMeOH—methanol; nBOH—n-butanol; P. ether—petroleum ether. Strains: B. anthracisBacillus anthracis; B. cereusBacillus cereus; E. faecalisEnterococcus faecalis; H. influenzaeHaemophilus influenzae; K. pneumoniaeKlebsiella pneumoniae; M. catarrhalisMoraxella catarrhalis; M. luteusMicrococcus luteus; P. gingivalisPorphyromonas gingivalis; P. intermediaPrevotella intermedia; S. typhiSalmonella typhi; S. typhimuriumSalmonella typhimurium; S. boydiiShigella boydii; S. epidermisStaphylococcus epidermidis; S. sanguisStreptococcus sanguinis; S. mutansStreptococcus mutans; S. pyogenesStreptococcus pyogenes; V. choleraeVibrio cholerae. Abbreviations: ATCC—American type culture collection, BA—TLC bioautography, ci—clinical isolate; IZ—zone of inhibition; MIC—minimum inhibitory concentration; AgNPs—silver nanoparticles; * Rf—retardation factor.
According to the WHO, oral diseases are the most common non-communicable diseases, affecting people throughout life and causing pain, discomfort, disfigurement, and even death [114]. The Global Burden of Disease Study reports that oral diseases are among the leading causes of health problems, estimating that half of the world’s population is affected by these diseases [114,115]. The same study provided a comprehensive assessment, and among the results evaluated, permanent tooth decay was the most common cause, representing a major public health problem in many countries [116]. Therefore, preventing and controlling the spread of this health problem is a global challenge, requiring greater efforts and potentially innovative approaches to achieve it. The branches of several Diospyros (particularly D. lycioides, D mespiliformis, and D. villosa) are used as toothbrushes for oral care [41,44,52,104,117], and their plant extracts have been shown to be effective against common oral pathogens, including Streptococcus mutans, S. sanguis, periodontal pathogens (Porphyromonas gingivalis and Prevotella intermedia), Lactobacillus spp., and several strains of Candida spp. [41,44,52,104,117]. In fact, over the past few decades, the scientific community has become increasingly interested in understanding the versatility of medicinal plants from traditional herbal medicine and their guaranteed availability to improve clinical approaches to infectious diseases with the intention of reducing antimicrobial resistance [4].

2.4. Secondary Metabolites of Mozambican Diospyros Species as Potential Antimicrobial Agents

2.4.1. Naphtoquinones

Antibacterial Activity

Plumbagin (1, Figure 4) is recognized as an effective antibacterial agent against both Gram-positive and Gram-negative strains of bacteria. This compound has also shown significant inhibitory activity (MIC < 12.5 μg/mL) against the resistant strain of Mycobacterium tuberculosis H37Rv [3,78,118]. Plumbagin isolated from the bark extract of D. maritima and showed activity against S. aureus and Aeromonas hydrophila (MIC = 0.625 and 5 µg/mL, respectively) [119]. In addition, it has also been obtained from the root of D. mespiliformis and has been described as one of the active marker compounds as well as an effective antibacterial agent against Gram-positive and Gram-negative bacterial strains [50,77,112].
Another important compound isolated from D. hebecarpa, 7-methyljuglone (2, Figure 4), also present in the root of Euclea natalensis (Ebenaceae), is potentially active against Mycobacterium tuberculosis (H37Rv) [18].
Isodiospyrin (4, Figure 4), a dimeric 7-methyljuglone-derivative, has been reported to be more active than diospyrin (3, Figure 4) against various Gram-positive strains, including Streptococcus pyogenes, S. pneumoniae, Corynebacterium diphtheriae, Bacillus subtilis, Listeria monocytogenes, Mycobacterium chelonae, and Micrococcus luteus. Isodiospyrin demonstrates MIC values ranging from (0.78 to 50 μg/mL), while diospyrin shows MIC values ranging from (1.56 to 100 μg/mL) [17].
Extensive research has unveiled the mechanism of action of diospyrin and 7-methyljuglone against M. tuberculosis, highlighting their crucial role as non-competitive ATPase inhibitors in key enzymatic reactions [120]. Additionally, emerging evidence has demonstrated the anti-tuberculosis potential of other compounds, such as crassiflorone and plumbagin from D. crassiflora, as well as diospyrone and plumbagin from D. canaliculata, both derived from the stem bark [121].
In a study conducted by Kuete et al. (2010), it was demonstrated that isobavacalcone and diospirone, derived from D. canaliculata, show promise as potential drugs against multidrug-resistant Gram-negative strains. These compounds exhibited enhanced activity when used in combination with efflux pump inhibitors, resulting in MIC values decreased to <10 μg/mL [122,123].

Antifungal and Antiviral Activities

The NQs have been well established, particularly against several species of Candida, infectious fungi of the mucosa, deep tissues, and the most common fungal diseases in HIV/AIDS patients [124]. Plumbagin inhibits the growth of C. albicans, C. tropicalis, and other fungi. In addition, fractions derived from plumbagin of Diospyros extracts are active against C. albicans [1]. In comparison with ketoconazole, a standard antifungal compound, plumbagin is considered a promising antifungal agent and has been used against C. albicans, C. glabrata, C. krusei, C. tropicalis, Cryptococcus neoformans, Aspergillus niger, A. flavus, Alternaria sp., Cladosporium sp., Geotrichum candidum, Fusarium sp., Helminthosporum sp., M. gypseum, and Penicillium sp. [125,126,127]. This compound, isolated from the stem bark of D. bipindensis, also exhibits significant activity against C. albicans [128,129,130].
Isolated from the root of D. virginiana, 7-methyljuglone and isodiospyrin have significant antifungal activity against Phomopsis obscurans (leaf blight), with 97.0% and 81.4% growth inhibition at 30 μM, respectively. These compounds also demonstrate activity against the pathogen Phomopsis viticola, with growth inhibition rates of 53.4% and 57.7%, respectively [131].
The antiaflatoxigenic activity of D. mafiensis root, another Mozambican medicinal plant, has been linked to the presence of diosquione and 3-hydroxydioquinone, making this herbal drug also an important natural antifungal for preventing fungal growth and aflatoxin accumulation in food [42]. In addition, this species has also been found to have analgesic, antidiabetic, anti-inflammatory, and antioxidant effects, likely correlated with the presence of these kind of constituents.

Antiparasitic Activity

NQs are highly active against pathogens in neglected tropical diseases, including malaria, leishmaniasis, and trypanosomiasis (sleeping sickness). Studies examining Plasmodium sp. have shown that isodiospyrin-derived isodiospyrol A exhibits antimalarial activity (IC50 = 2.7 μg/mL) [132]. Anti-plasmodial activity has also been reported in the ethanolic extract of leafs of D. monbuttensis (IC50 = 3.2 nM) [133]. Studies on malaria have proposed a redox cycling mechanism (described for the novel antimalarial–antiparasitic drug atovaquone) to support the in vitro activity of diospyrin and its analogues isolated from D. montana against L. donovani [134].
Plumbagin and its derivative was shown to be active against Leishmania spp., while diospyrin was active against Leishmania donovani [87]. Semisynthetic crassiflorone derivatives display trypanocidal activity against T. brucei and T. cruzi [135]. Antiplasmodial activities with IC50 values of 16.5 to 29.4 g/mL against chloroquine-sensitive (3D7) and chloroquine-resistant (K1) strains of P. falciparum were observed for the juglone-based 1,4-NQs present in D. sylvatica [136].
Concerning the assessment of anthelmintic activity, it was demonstrated in vitro that D. oocarpa, D. nigrisence, D. candolleana, and D. montana are active on adult earthworms of Pheritima posthuma [137]. Similarly, NQ derivatives, including diospyrin from D. oocarpa, D. nigrisence, and D. candolleana, are antiprotozoal in addition to possessing anthelmintic constituents [138].

2.4.2. Triterpenoids

Antibacterial and Antifungal Activities

Betulinic acid isolated from the root of D. lotus presents a broad spectrum against several Gram-positive and Gram-negative bacteria [85,139,140,141]. Betulin isolated from D. rubra is an active agent against Streptococcus pyogenes, with a MIC of 85 µg/mL, and Corynebacterium diphtheriae, with a MIC range of 64 to 256 µg/mL [88].
Methanolic extract obtained from D. peregrina bark and seed containing triterpenoids has been studied for its antidiarrheal properties [142]. Similarly, the methanolic extract of D. peregrina fruit showed high activity against E. coli (12.6 mm zone of inhibition) and against fungi C. albicans (10.7 mm zone of inhibition) and Penicillium spp. (7.33 mm) [143].
Betulin present in the hexane fraction isolated from the bark of D. paniculata is very efficient against S. dysenteriae, which is responsible for diarrhea (MIC = 30 μg/mL) [144]. However, a study of a reductive green synthesis of nano-sized Ag particles using methanolic root extracts of D. paniculata showed that the maximum activity was displayed against Gram-positive bacteria compared to Gram-negative bacteria. The maximum activity was observed against Penicillium notatum, A. flavus, and Saccharomyces cerevisiae, with moderate activity towards C. albicans and A. niger [145].
In another study of ursane-type triterpenoids obtained from the leaf of D. dendo Welw. Ex Hiern [EtOH−EtOAc (50:50) extract], antimicrobial activity (62% at 10 μg/mL) against Pseudomonas aeruginosa was observed. This Gram-negative bacterium is considered one of the three main causes of human opportunistic infections and has recently been a useful model for the study of biofilm formation, implying antimicrobial resistance to antibiotics [146].

Antiviral Activity

Structure–activity relationships between betulinic acid and its synthetic derivatives inhibiting HIV-1 replication, HIV-1 entry, and HIV-protease or reverse transcriptase (RT) have been verified [147,148]. Betulinic acid was identified as a highly promising antiviral (anti-dengue) present in high proportions in most extracts of distinct species of Diospyros, particularly from the bark of D. glans [83]. Aridanin, isolated from methanol extracts obtained from the leaf, stem, and root of D. conocarpa, presents anti-HIV-1IN activity [149].
In a recent study, the antiviral activity of D. anisandra was demonstrated against the influenza virus AH1N1pdm09. The n-hexane fruit extract exhibited HA inhibitory (HAI) activity, and a fraction of it inhibited the hemagglutination from 12.5 up to 100 μg/mL, which was attributed to the synergistic effect of the different compounds present [150]. Previously, possible antiviral activity against influenza A and B viruses has been attributed to a redox effect of isolated zeylanone epoxide [151].

Antiparasitic Activity

Using in vitro antimalarial assays, betulinic acid 3-caffeate isolated from the dried leaf, twig, and branch of D. quaesita was shown to be moderately active against both chloroquine-sensitive and chloroquine-resistant P. falciparum clones [86]. Lupeol and lupenone, isolated from the dichloromethane and ethyl acetate extracts of D. rubra stem, have shown moderate antimalarial activity against P. falciparum [88]. On the other hand, hydroethanolic extracts from the trunk of D. gracilescens and the hexane fraction showed higher activity against promastigote and amastigote forms of L. donovani (IC50 = 5.84 μg/mL and IC50 = 0.79 μg/mL, respectively) [87]. Aridanin isolated from methanol extracts of the leaf, stem, and root of D. conocarpa can be sources of new antitrypanosomal active principles [149].

2.4.3. Tannins

Tannins isolated from Mozambican Diospyros species represent an important class of secondary metabolites with remarkable antimicrobial potential against fungi, bacteria, and yeast [152]. Their mechanism of action involves the disruption of microbial enzymes and cell membranes, although their activities are diverse [153]. In addition, recent research has suggested the ability of tannins to generate hydrogen peroxide, which contributes to their important antibacterial properties [154].

Antibacterial and Antifungal Activities

D. melanoxylon bark is another medicinal plant considered to be active against Gram-positive and Gram-negative bacteria, which is traditionally used for diarrhea, urinary, and skin troubles and has confirmed claims against E. coli, S. aureus, S. epidermidis, Shigella flexneri, Bacillus licheniformis, Bacillus brevis, Vibrio cholerae, P. aeruginosa, Streptococcus aureus, Candida kruesi, and Bacillus subtilis [155]. Furthermore, it shows promise in the treatment of candidiasis caused by different Candida species (C. viz, C. albicans, C. krusei, C. parapsilosis, and C. tropicalis), with MIC values ranging from 0.375 to 6.0 mg/mL [156]. Extracts derived from the bark of D. melanoxylon are rich in tannins and possess significant potential as antimicrobial agents. In a recent study using strains isolated from humans, it was effective against both Gram-positive and Gram-negative bacteria, suggesting the presence of a broad spectrum of antibiotic compounds or simply general metabolic toxins in the plant methanolic extract [157,158]. In another study conducted in India, acetone ethyl acetate and methanol extracts of D. melanoxylon showed a MIC < 30 μg/mL against Aeromonas hydrophila, Enterobacter aerogenes, E. coli, and Klebsiella pneumoniae [159].
Methanol extract obtained from the bark or seed of D. peregrina, which is rich in tannins and other phenols, was evaluated for its antibacterial potential against the pathogenic bacteria associated with diarrhea. The bark extract demonstrated inhibitory effects against S. aureus, Shigella dysenteriae, E. coli, and P. aeruginosa, while the seed extract inhibited all tested strains except for P. aeruginosa [160]. Similarly, the methanol extract of D. tricolor leaves, known for its abundance of tannins and other phenols, exhibited antibacterial activity against both Gram-positive bacteria (Bacillus cereus and S. aureus) and Gram-negative bacteria (Salmonella typhii and Escherichia coli) [161].
Diospyros kaki Thunb., known as the persimmon tree, is originally from Asia, but it is cultivated in various parts of the world, including Mozambique. Different plant parts are well-known and useful as medicinal plants, and the fruit is known as persimmon. This species has been extensively studied, particularly regarding the antimicrobial activity of the tannins isolated from it. In a study conducted by Liu et al. (2019), the antimicrobial effects of persimmon tannins (PTs) extracted from the fruit of D. kaki against methicillin-resistant Staphylococcus aureus (MRSA) were investigated. The persimmon tannins (MIC = 1000 μg/mL) displayed potential mechanisms of inhibitory activity (i.e., the tannins can change the normal morphology of MRSA and cause severe damage to the cell wall and cell membrane) [152]. In addition, the hydrolysate of condensed tannins (composed of a polymer of flavan-3-ols, such as catechin groups) exhibited high bacteriostatic activity in vitro against the M. avium complex (nontuberculous mycobacteria) that causes opportunistic chronic pulmonary infections [63]. Aqueous extract from the D. kaki fruit was tested in vivo, showing interesting antibacterial activities against Gram-negative strains compared to Gram-positive bacteria, justifying its use in traditional medicine for the treatment and/or management of disorders of the digestive system such as diarrhea [162]. The results of another study showed that the condensed tannins extracted from the unripened fruit of D. kaki displayed antibacterial activity against biofilms containing multiple bacteria. It is estimated that intraoral cavity biofilms consist of at least 800 types of bacteria. Therefore, it is suggested that this medicinal plant has a high potential for preventing dental disease and aspiration pneumonitis in geriatric patients and recovering patients when it is added to mouthwash and toothpaste [163].
The in vitro antibacterial potential of D. blancoi was also found against biofilm formation by S. mutans. Both extracts containing tannins and other phenols showed inhibition ranges of 96% for methanol and 95% for ethyl acetate [164].
Recently, Diospyros species rich in tannins have been applied in the development of nanoparticles. For instance, titanium dioxide (TiO2) nanoparticles containing D. ebenum leaf extract exhibit excellent antibacterial activity and potential against Gram-negative bacteria E. coli [165]. Silver nanoparticles (AgNPs) containing aqueous extract from the fruit of D. malabarica have demonstrated antibacterial activity against S. aureus at 500 μg/mL and against E. coli at 1000 ug/mL, with an average zone of inhibition size of 8.4 ± 0.3 mm and 12.1 ± 0.5 mm and 6.1 ± 0.7 mm and 13.1 ± 0.5 mm, respectively [166]. Similarly, biogenic silver nanoparticles demonstrated excellent antibacterial activity against a broad range of bacteria, with the highest antibacterial activity observed against E. faecalis (17.77 mm) and B. subtilis (20 mm), also demonstrating good hemocompatibility against humans and rat red blood cells [167].

Antiviral Activity

No studies were found on the specific activity of tannins isolated from the native Diospyros species in Mozambique. However, a tannin isolated from D. kaki has been demonstrated to have in vitro antiviral activity against the influenza virus, vesicular stomatitis virus, poliovirus, coxsackievirus, adenovirus, rotavirus, feline calicivirus, mouse norovirus, Sendai virus, and Newcastle disease virus [168]. The results of another study involving D. kaki extracts with tannin contents ranging from 0.08 to ≥0.11 mg/mL demonstrated their capacity to inactivate human noroviruses and bacteriophage MS2, both of which are the cause of gastroenteritis and foodborne illnesses worldwide (i.e., the results suggest that the antiviral effect and astringent effects of tannins are likely related to noroviral genome reduction and MS2 inactivation) [169].

Antiparasitic Activity

Species of the genus Diospyros contain a broad spectrum of antimicrobial agents identified using in vitro and/or in vivo methods against strains capable of causing opportunistic infections as well as neglected parasitic diseases. The anthelmintic activity of a D. peregrina fruit extract containing tannins was compared to the standard drug albendazole. The extract was found to be more potent than the selected standard drug at a concentration of 10 mg/mL [170].
According to the WHO, malaria is one of the most widespread neglected diseases in Africa, caused by the parasite Plasmodium and responsible for severe immune complications and deaths. The anti-Plasmodium activity of extracts from various species of the Mozambican Diospyros species has been reported in the literature. Ethyl acetate extract from D. abyssinica leaves showed moderate activity against chloroquine-resistant Plasmodium falciparum (FcB1), while D. mespiliformis, traditionally used to treat malaria, showed potent antimalarial activity in mice infected with Plasmodium berghei and significant inhibition of beta-hematin using an in vitro assay [98].
The antiparasitic activity against Leishmania donovani, Trypanosoma cruzi, and Trypanosoma brucei was confirmed in several studies on Diospyros species [99]. For example, an acetate leaf extract of D. abyssinica and the isodiospyrin and diospyrin marker compounds isolated from the bark by bioguided fractionation showed high anti-L. donovani activity (IC50 = 1.5 g/mL, extract, and IC50 = 0.5 g/mL, isolated compounds) [65].

3. Materials and Methods

This review was conducted according to the criteria described in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 statement (http://www.prisma-statement.org/; accessed on 16 January 2023). For this purpose, the scientific literature data were considered until 10 December 2022.

3.1. Search Strategy

The scientific data were collected using the search engines PubMed, Scopus, Web of Science, and Google Scholar, identifying all scientific papers published between 1 January 1970, and 10 December 2022 using the keywords Diospyros AND antibacterial, Diospyros AND antifungal, Diospyros AND antiparasitic, Diospyros AND antiviral, Diospyros AND medicine, Diospyros AND chemical compounds, Diospyros AND biological activity, and Diospyros AND toxicity.

3.2. Study Selection

As described in Figure 7, a total of 5528 scientific studies were included in the search and initial data collection based on their title and abstract. After eliminating the duplicates, 2071 studies remained, of which 1852 could not be selected due to a lack of information relevant to this work. After the screening, 279 studies reporting on Diospyros were considered eligible for inclusion in this review.

3.2.1. Criteria for Inclusion and Exclusion of Data

Inclusion Criteria

Related to the Diospyros genus, in particular species of the genus Diospyros present in Mozambican Flora;
Abstract or full text in English;
Studies on Diospyros species concerning their medicinal importance.

Exclusion Criteria

Duplicate scientific publications;
Not directly related to medicinal issues and others related but not with species of Mozambican Flora;
Containing irrelevant or incomplete information.

4. Conclusions

Species of the genus Diospyros have been studied worldwide, with a significant number exhibiting pharmacological activity. One referenced example, D. kaki, native to East Asia, NaoXinQing, is part of a patented and officially approved traditional Chinese medicine formula for the treatment of stroke. However, there are no studies integrating data on all Diospyros species present in the flora of Mozambique.
More than 70% of Mozambique’s population uses medicinal plants for primary health care, and a total of 54.8% of the Diospyros species used in the country’s ethnomedicine are also used in other regions of Africa; however, the biological potential of most of them is still largely unknown. For example, 64.5% of these species were not tested for their antibacterial properties, namely D. abyssinica subsp. attenuata, D. abyssinica subsp. Chapmaniorum, D. anitae, D. consolatae, D. consolatae-rotundifolia intermediates, D. dichrophylla, D. ferrea, D. inhacaensis, D. kirkii, D. kirkii-mespiliformis intermediates, D. loureiriana subsp. Loureiriana, D. natalensis subsp. Numulária, D. quiloensis, D. senensis, D. truncatifolia, D. usambarensis subsp. Usambarensis/rufescens, D. villosa var. parvifolia, D. villosa var. villosa, D. whyteana, D. zombensis, and Diospyros sp. no. 1 sensu FZ. On the other hand, several isolated compounds of these species (particularly naphthoquinones and triterpenoids) have also been isolated from other species of the genus Diospyros, showing different biological activities including antiviral activity. However, no antiviral studies were found on the Mozambican species.
Studies on the antifungal potential of Diospyros are still scarce. In fact, the antifungal activity of 98.14% of the species (D. abyssinica subsp. attenuata, D. abyssinica subsp. chapmaniorum, D. anitae, D. bussei, D. consolatae, D. consolatae-rotundifolia intermediates, D. dichrophylla, D. inhacaensis, D. kabuyeana, D. kirkii, D. kirkii-mespiliformis intermediates, D. loureiriana subsp. loureiriana, D. lycioides Desf. subsp. sericea, D. natalensis subsp. natalensis, D. natalensis subsp. numulária, D. quiloensis, D. rotundifolia, D. senensis, D. squarrosa, D. truncatifolia, D. verrucosa, D. villosa var. parvifolia, D. whyteana, D. zombensis, and Diospyros sp. No. 1 sensu FZ) need to be evaluated, as they are traditionally used to treat skin diseases and diseases of the oral cavity, as well as other diseases where opportunistic fungal infections can co-occur. In addition, antiparasitic activities have been studied in other species of the genus Diospyros, however, 97.21% of Mozambican species (D. abyssinica subsp. attenuata, D. abyssinica subsp. chapmaniorum, D. anitae, D. consolatae, D. consolatae-rotundifolia intermediates, D. dichrophylla, D. ferrea, D. inhacaensis, D. kirkii, D. kirkii-mespiliformis intermediates, D. lycioides Desf. subsp. sericea, D. mafiensis, D. natalensis subsp. numularia, D. quiloensis, D. rotundifolia, D. senensis, D. squarrosa, D. truncatifolia, D. villosa var. villosa, D. villosa var. parvifolia, D. whyteana, D. zombensis, and Diospyros. sp. no. 1 sensu FZ) have not yet had their antiparasitic activities studied.
In summary, out of the 31 native and naturalized species in the flora of Mozambique that are used in different regions of Africa, a total of 17 species have not been studied as antimicrobial agents, of which three species, namely D. dichrophylla, D. whyteana, and D. zombensis, have only been studied at the toxicological level. Of the 14 species that have already been the subject of antimicrobial studies, D. abyssinica and D. mespiliformis are the best studied.
This work provides comprehensive information on the chemical, biological, and toxicological studies of the Diospyros species present in the flora of Mozambique, examining their pharmacological potential in detail. Of the Diospyros plant parts, the root is the best-researched and documented. The identified studies confirmed ongoing efforts to improve the understanding of the mechanism of action underlying the biological activity, and in particular, the antimicrobial activity of these species, drawing on their traditional use. In addition, several secondary metabolites of Diospyros are currently being investigated for their potential pharmacological applications. However, it is important to emphasize that most of the available data are in vitro assessments of biological activity. Therefore, further efforts are needed to obtain more comprehensive evidence aimed at strengthening the validity and applicability of the results and ultimately contributing to public health benefits, especially in the face of global antimicrobial resistance.

Author Contributions

Conceptualization, A.R., R.S., J.F.P. and O.S.; investigation, methodology, and data collection, A.R., R.S., J.F.P. and O.S.; data analysis, A.R., R.S., J.F.P., I.B.M.d.S., E.T.G. and O.S.; writing—original draft preparation, A.R., R.S., J.F.P. and O.S.; writing—review and editing, A.R., R.S., J.F.P., I.B.M.d.S., E.T.G. and O.S.; supervision—O.S.; project administration, O.S.; funding acquisition, O.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Foundation for Science and Technology (FCT, Portugal) through the national funds FCT/MCTES to iMed.ULisboa (UIDP/04138/2020).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rauf, A.; Uddin, G.; Patel, S.; Khan, A.; Halim, S.A.; Bawazeer, S.; Ahmad, K.; Muhammad, N.; Mubarak, M.S. Diospyros, an under-Utilized, Multi-Purpose Plant Genus: A Review. Biomed. Pharmacother. 2017, 91, 714–730. [Google Scholar] [CrossRef] [PubMed]
  2. Fareed, N.; El-Kersh, D.M.; Youssef, F.S.; Labib, R.M. Unveiling Major Ethnopharmacological Aspects of Genus Diospyros in Context to Its Chemical Diversity: A Comprehensive Overview. J. Food Biochem. 2022, 46, e14413. [Google Scholar] [CrossRef]
  3. Nematollahi, A.; Aminimoghadamfarouj, N.; Wiart, C. Reviews on 1,4-Naphthoquinones from Diospyros L. J. Asian Nat. Prod. Res. 2012, 14, 80–88. [Google Scholar] [CrossRef] [PubMed]
  4. Mallavadhani, U.V.; Panda, A.K.; Rao, Y.R. Review Article Number 134 Pharmacology and Chemotaxonomy of Diospyros. Phytochemistry 1998, 49, 901–951. [Google Scholar] [CrossRef] [PubMed]
  5. Wallnöfer, B. The Biology and Systematics of Ebenaceae: A Review. Ann. Naturhist. Mus. Wien 2001, 103 Bd, 485–512. [Google Scholar]
  6. White, F. Ebenaceae . Flora Zambesiaca 1983, 7, 269–271. [Google Scholar]
  7. Denk, T.; Bouchal, J.M. Dispersed Pollen and Calyx Remains of Diospyros (Ebenaceae) from the Middle Miocene “Plant Beds” of Søby, Denmark. GFF 2021, 143, 292–304. [Google Scholar] [CrossRef]
  8. Burrows, J.E.; Burrows, S.M.; Lötter, M.C.; Schmidt, E. Trees and Shrubs of Mozambique; Print Matters Heritage: Cape Town, South Africa, 2018; pp. 757–1114. [Google Scholar]
  9. The Plant List (2013). Version 1.1. Published on the Internet. Available online: http://www.theplantlist.org/1.1/cite/ (accessed on 27 January 2021).
  10. The WFO Plant List|World Flora Online. Available online: https://wfoplantlist.org/plant-list (accessed on 1 December 2022).
  11. Flora of Mozambique: Home Page. Available online: https://www.mozambiqueflora.com/ (accessed on 1 December 2022).
  12. Da Silva, M.C.; Izidine, S.; Amude, A.B. A Preliminary Checklist of the Vascular Plants of Mozambique. In Southern Africa Botanical Diversity Network 30; SABONET: Pretoria, South Africa, 2004; pp. 1–192. [Google Scholar]
  13. World Health Organization. WHO Global Report on Traditional and Complementary Medicine 2019; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  14. WHO Traditional Medicine Strategy: 2014–2023. Available online: https://www.who.int/publications/i/item/9789241506096 (accessed on 1 December 2022).
  15. Bandeira, S.O.; Gaspar, F.; Pagula, F.P. African Ethnobotany and Healthcare: Emphasis on Mozambique. Pharm. Biol. 2011, 39, 70–73. [Google Scholar] [CrossRef]
  16. Babula, P.; Adam, V.; Havel, L.; Kizek, R. Noteworthy Secondary Metabolites naphthoquinones—Their Occurrence, Pharmacological Properties and Analysis. Curr. Pharm. Anal. 2009, 5, 47–68. [Google Scholar] [CrossRef]
  17. 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]
  18. Mahapatra, A.; Mativandlela, S.P.N.; Binneman, B.; Fourie, P.B.; Hamilton, C.J.; Meyer, J.J.M.; van der Kooy, F.; Houghton, P.; Lall, N. Activity of 7-methyljuglone Derivatives against Mycobacterium tuberculosis and as Subversive Substrates for Mycothiol Disulfide Reductase. Bioorg. Med. Chem. 2007, 15, 7638–7646. [Google Scholar] [CrossRef] [PubMed]
  19. World Health Organization. WHO Library Cataloguing-in-Publication Data Global Action Plan on Antimicrobial Resistance. Microbe Mag. 2015, 10, 354–355. [Google Scholar]
  20. Antimicrobial Resistance: A Global Threat|UNEP—UN Environment Programme. Available online: https://www.unep.org/explore-topics/chemicals-waste/what-we-do/emerging-issues/antimicrobial-resistance-global-threat (accessed on 14 January 2023).
  21. WHO World Health Statistics 2022: Monitoring Health for the SDGs, Sustainable Development Goals. Available online: https://www.who.int/publications/i/item/9789240051157 (accessed on 14 January 2023).
  22. Kaikabo, A.A.; Suleiman, M.M.; Samuel, B.B.; Eloff, J.N. Antibacterial Activity of Eleven South African Plants Use in Treatment of Diarrhoea in Folkloric Medicine. Afr. J. Tradit. Complement. Altern. Med. 2009, 6, 315–316. [Google Scholar]
  23. Bandeira, S.; Senkoro, A.; Barbosa, F.; Mualassace, D.; Figueiredo, E. The Terrestrial Environment Adjacent to Maputo Bay. In The Maputo Bay Ecosystem; Bandeira Salomão, P.J., Ed.; Western Indian Ocean Marine Science Association: Zanzibar Town, Tanzania, 2014; pp. 239–254. [Google Scholar]
  24. Iwalewa, E.O.; McGaw, L.J.; Naidoo, V.; Eloff, J.N. Inflammation: The Foundation of Diseases and Disorders. A Review of Phytomedicines of South African Origin Used to Treat Pain and Inflammatory Conditions. Afr. J. Biotechnol. 2007, 6, 2868–2885. [Google Scholar] [CrossRef]
  25. Conde, P.; Figueira, R.; Saraiva, S.; Catarino, L.; Romeiras, M.; Duarte, M.C. The Botanic Mission to Mozambique (1942–1948): Contributions to Knowledge of the Medicinal Flora of Mozambique. Hist. Cienc. Saude Manguinhos 2014, 21, 539–585. [Google Scholar] [CrossRef]
  26. Ribeiro, A.; Serrano, R.; Silva, I.B.M.D.; Gomes, E.T.; Pinto, J.F.; Silva, O. Silva Botanical Markers of Diospyros villosa Dried Root for Pharmacognostic Characterization. In Proceedings of the iMed.Ulisboa Meeting, Lisbon, Portugal, 15 May 2021; Book of Abstract: Lisbon, Portugal, 2021; pp. 57–191. [Google Scholar]
  27. Maiga, A.; Malterud, K.E.; Diallo, D.; Paulsen, B.S. Antioxidant and 15-Lipoxygenase Inhibitory Activities of the Malian Medicinal Plants Diospyros abyssinica (Hiern) F. White (Ebenaceae), Lannea velutina A. Rich (Anacardiaceae) and Crossopteryx febrifuga (Afzel) Benth. (Rubiaceae). J. Ethnopharmacol. 2006, 104, 132–137. [Google Scholar] [CrossRef]
  28. Nafiu, M.O.; Salawu, M.O.; Kazeem, M.I. Antioxidant Activity of African Medicinal Plants. In Medicinal Plant Research in Africa; Elsevier: Amsterdam, The Netherlands, 2013; pp. 787–803. [Google Scholar]
  29. Okello, S.V.; Nyunja, R.O.; Netondo, G.W.; Onyango, J.C. Ethnobotanical Study of Medicinal Plants Used by Sabaots of Mt. Elgon Kenya. Afr. J. Tradit. Complement. Altern. Med. 2010, 7, 1–10. [Google Scholar] [CrossRef]
  30. Namukobe, J.; Kasenene, J.M.; Kiremire, B.T.; Byamukama, R.; Kamatenesi-Mugisha, M.; Krief, S.; Dumontet, V.; Kabasa, J.D. Traditional Plants Used for Medicinal Purposes by Local Communities around the Northern Sector of Kibale National Park, Uganda. J. Ethnopharmacol. 2011, 136, 236–245. [Google Scholar] [CrossRef]
  31. Lacroix, D.; Prado, S.; Kamoga, D.; Kasenene, J.; Namukobe, J.; Krief, S.; Dumontet, V.; Mouray, E.; Bodo, B.; Brunois, F. Antiplasmodial and Cytotoxic Activities of Medicinal Plants Traditionally Used in the Village of Kiohima, Uganda. J. Ethnopharmacol. 2011, 133, 850–855. [Google Scholar] [CrossRef]
  32. Tugume, P.; Kakudidi, E.K.; Buyinza, M.; Namaalwa, J.; Kamatenesi, M.; Mucunguzi, P.; Kalema, J. Ethnobotanical Survey of Medicinal Plant Species Used by Communities around Mabira Central Forest Reserve, Uganda. J. Ethnobiol. Ethnomed. 2016, 12, 5. [Google Scholar] [CrossRef]
  33. Timberlake, J.; Golding, J.; Clarke, P. Niassa Botanical Expedition—June 2003. Prep. Soc. Para A Gestão E Desenvolv. Da Reserva Do Niassa Moçambique Biodivers. Found. Afr. 2004, 43, 3–20. [Google Scholar]
  34. Kadavul, K.; Dixit, A.K. Ethnomedicinal Studies of the Woody Species of Kalrayan & Shervarayan Hills, Eastern Chats, Tamil Nadu. Indian J. Tradit. Knowl. 2009, 8, 592–597. [Google Scholar]
  35. Vijayalakshmi, R.; Ravindhran, R. Pharmacognostical Studies on Root of Diospyros ferrea (Willd.) Bakh and Aerva lanata Linn., a Potent Indian Medicinal Plants. Asian J. Pharm. Clin. Res. 2013, 6, 323–327. [Google Scholar]
  36. Vijayalakshmi, R.; Ravindhran, R. HPTLC Method for Quantitative Determination of gallic acid in Ethanolic Root Academic Sciences Asian Journal of Pharmaceutical and Clinical Research. Asian J. Pharm. Clin. Res. 2012, 5, 170–174. [Google Scholar]
  37. Choi, C.W.; Song, S.B.; Oh, J.S.; Kim, Y.H. Antiproliferation Effects of Selected Tanzania Plants. Afr. J. Tradit. Complement. Altern. Med. 2015, 12, 96–102. [Google Scholar] [CrossRef]
  38. Bagla, V.P.; Lubisi, V.Z.; Ndiitwani, T.; Mokgotho, M.P.; Mampuru, L.; Mbazima, V. Antibacterial and Antimetastatic Potential of Diospyros lycioides Extract on Cervical Cancer Cells and Associated Pathogens. Evid.-Based Complement. Altern. Med. 2016, 2016, 1–10. [Google Scholar] [CrossRef]
  39. Miller, J.S. Zulu Medicinal Plants: An Inventory By A. Hutchings with A. H. Scott, G. Lewis, and A. B. Cunningham (University of Zululand). J. Nat. Prod. 1997, 60, 955. [Google Scholar] [CrossRef]
  40. 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]
  41. Cai, L.; Wei, G.-X.; van der Bijl, P.; Wu, C.D. Namibian Chewing Stick, Diospyros lycioides, Contains Antibacterial Compounds against Oral Pathogens. J. Agric. Food Chem. 2000, 48, 909–914. [Google Scholar] [CrossRef]
  42. Mmongoyo, J.A.; Nair, M.G.; Linz, J.E.; Wu, F.; Mugula, J.K.; Dissanayake, A.A.; Zhang, C.; Day, D.M.; Wee, J.M.; Strasburg, G.M. Bioactive Compounds in Diospyros mafiensis Roots Inhibit Growth, Sporulation and Aflatoxin Production by Aspergillus flavus and Aspergillus parasiticus. World Mycotoxin. J. 2017, 10, 237–248. [Google Scholar] [CrossRef]
  43. Adzu, B.; Amos, S.; Muazzam, I.; Inyang, U.S.; Gamaniel, K.S. Neuropharmacological Screening of Diospyros mespiliformis in Mice. J. Ethnopharmacol. 2002, 83, 139–143. [Google Scholar] [CrossRef] [PubMed]
  44. Chinsembu, K.C. Ethnobotanical Study of Plants Used in the Management of HIV/AIDS-Related Diseases in Livingstone, Southern Province, Zambia. Evid.-Based Complement. Altern. Med. 2016, 2016, 1–14. [Google Scholar] [CrossRef] [PubMed]
  45. 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] [PubMed]
  46. 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] [PubMed]
  47. 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]
  48. 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]
  49. 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]
  50. Nworu, C.; Onuigbo, E.; Omeje, J.; Nsirim, K.; Ogbu, J.; Ngwu, M.; Chah, K.; Esimone, C. Anti-Mycobacterial Activity of Root and Leaf Extracts of Anthocleista djalonensis (Loganiaceae) and Diospyros mespiliformis (Ebenaceae). Int. J. Green Pharm. 2009, 3, 201. [Google Scholar] [CrossRef]
  51. 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. South Afr. J. Bot. 2022, 150, 305–312. [Google Scholar] [CrossRef]
  52. Chinsembu, K.C. Plants and Other Natural Products Used in the Management of Oral Infections and Improvement of Oral Health. Acta Trop. 2016, 154, 6–18. [Google Scholar] [CrossRef]
  53. Maroyi, A. Traditional Use of Medicinal Plants in South-Central Zimbabwe: Review and Perspectives. J. Ethnobiol. Ethnomed. 2013, 9, 31. [Google Scholar] [CrossRef] [PubMed]
  54. Marston, A.; Msonthi, J.; Hostettmann, K. Naphthoquinones of Diospyros usambarensis Their Molluscicidal and Fungicidal Activities. Planta Med. 1984, 50, 279–280. [Google Scholar] [CrossRef] [PubMed]
  55. Hamza, O.J.M.; van den Bout-van den Beukel, C.J.P.; Matee, M.I.N.; Moshi, M.J.; Mikx, F.H.M.; Selemani, H.O.; Mbwambo, Z.H.; van der Ven, A.J.A.M.; Verweij, P.E. Antifungal Activity of Some Tanzanian Plants Used Traditionally for the Treatment of Fungal Infections. J. Ethnopharmacol. 2006, 108, 124–132. [Google Scholar] [CrossRef] [PubMed]
  56. Moshi, M.J.; Mbwambo, Z.H. Experience of Tanzanian Traditional Healers in the Management of Non-Insulin Dependent Diabetes Mellitus. Pharm. Biol. 2002, 40, 552–560. [Google Scholar] [CrossRef]
  57. Khan, M.; Kishimba, M.; Locksley, H. Extractives from Ebenaceae: Constituents of the Root and Stem Barks of Diospyros verrucosa. Planta Med. 1987, 53, 498. [Google Scholar] [CrossRef]
  58. Ribeiro, A.; Serrano, R.; da Silva, I.B.M.; Gomes, E.T.; Pinto, J.F.; Silva, O. Diospyros villosa Root Monographic Quality Studies. Plants 2022, 11, 3506. [Google Scholar] [CrossRef]
  59. Aston Philander, L. An Ethnobotany of Western Cape Rasta Bush Medicine. J. Ethnopharmacol. 2011, 138, 578–594. [Google Scholar] [CrossRef]
  60. Verschaeve, L.; Kestens, V.; Taylor, J.L.S.; Elgorashi, E.E.; Maes, A.; van Puyvelde, L.; de Kimpe, N.; van Staden, J. Investigation of the Antimutagenic Effects of Selected South African Medicinal Plant Extracts. Toxicol. Vitr. 2004, 18, 29–35. [Google Scholar] [CrossRef]
  61. Gafner, F.; Chapuis, J.-C.; Msonthi, J.D.; Hostettmann, K. Cytotoxic Naphthoquinones, Molluscicidal Saponins and Flavonols from Diospyros zombensis. Phytochemistry 1987, 26, 2501–2503. [Google Scholar] [CrossRef]
  62. Yoshihira, K.; Tezuka, M.; Takahashi, C.; Natori, S. Four New Naphthoquinone Derivatives from Diospyros spp. Chem. Pharm. Bull 1971, 19, 851–854. [Google Scholar] [CrossRef]
  63. Matsumura, Y.; Kitabatake, M.; Ouji-Sageshima, N.; Yasui, S.; Mochida, N.; Nakano, R.; Kasahara, K.; Tomoda, K.; Yano, H.; Kayano, S.; et al. Persimmon-Derived Tannin Has Bacteriostatic and Anti-Inflammatory Activity in a Murine Model of Mycobacterium avium Complex (MAC) Disease. PLoS ONE 2017, 12, e0183489. [Google Scholar] [CrossRef] [PubMed]
  64. 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]
  65. Krief, S.; Huffman, M.A.; Sévenet, T.; Hladik, C.-M.; Grellier, P.; Loiseau, P.M.; Wrangham, R.W. Bioactive Properties of Plant Species Ingested by Chimpanzees (Pan Troglodytes Schweinfurthii) in the Kibale National Park, Uganda. Am. J. Primatol. 2006, 68, 51–71. [Google Scholar] [CrossRef] [PubMed]
  66. Cantrell, C.L.; Berhow, M.A.; Phillips, B.S.; Duval, S.M.; Weisleder, D.; Vaughn, S.F. Bioactive Crude Plant Seed Extracts from the NCAUR Oilseed Repository. Phytomedicine 2003, 10, 325–333. [Google Scholar] [CrossRef] [PubMed]
  67. Prada, N.J.; Vishnauvardhan, Z.; Baratha, J. Gc-Ms Identification of Bioactive Compounds from Solvent Ex-Tracts of Diospyros ferrea (Willd.) Bakh, Leaf. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2019, 6, 93–98. [Google Scholar]
  68. Kuo, Y.H.; Li, S.Y.; Shen, C.C.; Yang, L.M.; Huang, H.C.; Liao, W.B.; Chang, C.I.; Kuo, Y.H.; Chen, C.F. Cytotoxic Constituents from the Fruit of Diospyros ferrea. Chin. Pharm. J. 1997, 49, 207–216. [Google Scholar]
  69. Tezuka, M.; Takahashi, C.; Kuroyanagi, M.; Satake, M.; Yoshihira, K.; Natori, S. New Naphthoquinones from Diospyros. Phytochemistry 1973, 12, 175–183. [Google Scholar] [CrossRef]
  70. Vijayalakshmi, R.; Ravindhran, R. Comparative Fingerprint and Extraction Yield of Diospyros ferrea (Willd.) Bakh. Root with Phenol Compounds (gallic acid), as Determined by Uv–Vis and Ft–Ir Spectroscopy. Asian Pac. J. Trop. Biomed. 2012, 2, S1367–S1371. [Google Scholar] [CrossRef]
  71. Van der Vijver, L.M.; Gerritsma, K.W. Naphthoquinones of Euclea and Diospyros Species. Phytochemistry 1974, 13, 2322–2323. [Google Scholar] [CrossRef]
  72. Théophile, O.; Christian, K.T.R.; Alain, Y.; Pascal, A.D.C.; Reine, B.G.S.; Diane, B.F.T.; Félicien, A.; Dominique, S.C.K. Natural Chemical Compounds from Plants Extract for Prevention and Treatment of Oral Infections: A Review. Int. J. Biosci. (IJB) 2022, 20, 21–38. [Google Scholar] [CrossRef]
  73. Khan, R.M.; Rwekika, E. 6″,8’-Bisdiosquinone from Diospyros mafiensis. Phytochemistry 1999, 50, 143–146. [Google Scholar] [CrossRef]
  74. Khan, M.R.; Rwekika, E. Naphthoquinones from the Barks of Three Species of the Genus Diospyros. Fitoterapia 1993, 64, 375. [Google Scholar]
  75. Khan, M.R.; Rwekika, E. Triterpenoids from the Leaves of Four Species of Family Ebenaceae. Fitoterapia 1992, 63, 375–376. [Google Scholar]
  76. Bulus, A.; Ben, A.C.; Florence, D.T.; Oluwakanyinsola, A.S.; Ogbaji, J.I. Isolation and Analgesic Property of Lupeol from Diospyros mespiliformis Stem Bark. J. Med. Plants Res. 2015, 9, 813–819. [Google Scholar] [CrossRef]
  77. 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. Z. Für Naturforschung C 2022, 77, 387–393. [Google Scholar] [CrossRef] [PubMed]
  78. 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]
  79. Sharma, V. Brief Review on the Genus Diospyros: A Rich Source of Naphthoquinones. Asian J. Adv. Basic Sci. 2017, 5, 34–53. [Google Scholar]
  80. Khan, M.; Kishimba, M.; Locksley, H. Naphthoquinones from the Root and Stem Barks of Diospyros usambarensis. Planta Med. 1989, 55, 581. [Google Scholar] [CrossRef]
  81. Hook, I.; Mills, C.; Sheridan, H. Bioactive naphthoquinones from Higher Plants. Stud. Nat. Prod. Chem. 2014, 41, 119–160. [Google Scholar] [CrossRef]
  82. Ebbo, A.A.; Mammam, M.; Suleiman, M.M.; Ahmed, A.; Bello, A. Preliminary Phytochemical Screening of Diospyros mespiliformis. Anat. Physiol. 2014, 4, 156–158. [Google Scholar] [CrossRef]
  83. Peyrat, L.-A.; Eparvier, V.; Eydoux, C.; Guillemot, J.-C.; Stien, D.; Litaudon, M. Chemical Diversity and Antiviral Potential in the Pantropical Diospyros Genus. Fitoterapia 2016, 112, 9–15. [Google Scholar] [CrossRef]
  84. 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]
  85. Rauf, A.; Uddin, G.; Khan, H.; Arfan, M.; Siddiqui, B.S. Bioassay-Guided Isolation of Antibacterial Constituents from Diospyros lotus Roots. Nat. Prod. Res. 2016, 30, 426–428. [Google Scholar] [CrossRef]
  86. Ma, C.-Y.; Musoke, S.F.; Tan, G.T.; Sydara, K.; Bouamanivong, S.; Southavong, B.; Soejarto, D.D.; Fong, H.H.S.; Zhang, H.-J. Study of Antimalarial Activity of Chemical Constituents from Diospyros quaesita. Chem. Biodivers 2008, 5, 2442–2448. [Google Scholar] [CrossRef] [PubMed]
  87. Njanpa, C.A.N.; Wouamba, S.C.N.; Yamthe, L.R.T.; Dize, D.; Tchatat, B.M.T.; Tsouh, P.V.F.; Pouofo, M.N.; Jouda, J.B.; Ndjakou, B.L.; Sewald, N.; et al. Bio-Guided Isolation of Anti-Leishmanial Natural Products from Diospyros gracilescens L. (Ebenaceae). BMC Complement. Med. Ther. 2021, 21, 1–12. [Google Scholar] [CrossRef]
  88. Prachayasittikul, S.; Saraban, P.; Cherdtrakulkiat, R.; Ruchirawat, S.; Prachayasittikul, V. New Bioactive Triterpenoids and Antimalarial Activity of Diospyros rubra Lec. EXCLI J. 2010, 9, 1–10. [Google Scholar] [PubMed]
  89. Maitera, O.; Louis, H.; Oyebanji, O.; Anumah, A. Investigation of Tannin Content in Diospyros mespiliformis Extract Using Various Extraction Solvents. J. Anal. Pharm. Res. 2018, 7, 00200. [Google Scholar] [CrossRef]
  90. Maroyi, A. Diospyros lycioides Desf.: Review of Its Botany, Medicinal Uses, Pharmacological Activities and Phytochemistry. Asian Pac. J. Trop. Biomed. 2018, 8, 130. [Google Scholar] [CrossRef]
  91. Prada, N.J.; Vardhan, V.; Reddy, S. Antidiabetic Activity of Methanolic Leaf Extract of Diospyros ferrea (Willd) Bakh. in Streptozotocin Induced Diabetic Rats. Glob. J. Res. Anal. 2017, 6, 323–327. [Google Scholar] [CrossRef]
  92. Marston, A.; Maillard, M.; Hostettmann, K. Search for Antifungal, Molluscicidal and Larvicidal Compounds from African Medicinal Plants. J. Ethnopharmacol. 1993, 38, 209–214. [Google Scholar] [CrossRef]
  93. 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, 1934578X2092318. [Google Scholar] [CrossRef]
  94. Diallo, D.; Marston, A.; Terreaux, C.; Touré, Y.; Smestad Paulsen, B.; Hostettmann, K. Screening of Malian Medicinal Plants for Antifungal, Larvicidal, Molluscicidal, Antioxidant and Radical Scavenging Activities. Phytother. Res. 2001, 15, 401–406. [Google Scholar] [CrossRef] [PubMed]
  95. Dougnon, V.; Hounsa, E.; Agbodjento, E.; Keilah, L.P.; Legba, B.B.; Sintondji, K.; Afaton, A.; Klotoe, J.R.; Baba-Moussa, L.; Bankole, H. Percentage Destabilization Effect of Some West African Medicinal Plants on the Outer Membrane of Various Bacteria Involved in Infectious Diarrhea. Biomed. Res. Int. 2021, 2021, 1–12. [Google Scholar] [CrossRef] [PubMed]
  96. Samie, A.; Tambani, T.; Harshfield, E.; Green, E.; Ramalivhana, J.N.; Bessong, P.O. Antifungal Activities of Selected Venda Medicinal Plants against Candida albicans, Candida krusei and Cryptococcus neoformans Isolated from South African AIDS Patients. Afr. J. Biotechnol. 2010, 9, 2965–2976. [Google Scholar]
  97. Ebbo, A.A.; Sani, D.; Suleiman, M.M.; Ahmed, A.; Hassan, A.Z. Phytochemical Composition, Proximate Analysis and Antimicrobial Screening of the Methanolic Extract of Diospyros mespiliformis Hochst Ex a. Dc (Ebenaceae). Pharmacogn. J. 2019, 11, 362–368. [Google Scholar] [CrossRef]
  98. Olanlokun, J.O.; Adetutu, J.A.; Olorunsogo, O.O. In 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]
  99. Christopher, R.; Mgani, Q.; Nyandoro, S.; Rousseau, A.; Vuuren, S.; Isaacs, M.; Hoppe, H. Antitrypanosomal, Antiplasmodial, and Antibacterial Activities of Extracts from Selected Diospyros and Annonaceae Species. J. Complement. Med. Res. 2018, 7, 161. [Google Scholar] [CrossRef]
  100. Rani, V.S.; Ramana, K.V. Evaluation of Anti-Inflammatory and Analgesic Activities of Diospyros ferrea Leaves. Res. J. Pharm. Biol. Chem. Sci. 2011, 2, 584–588. [Google Scholar]
  101. Ramana, K.V.; Rambabu, P.S.G. Evaluation of Anti-Inflammatory and Analgesic Activities of Diospyros ferrea Root. Adv. Pharmacol. Toxicol. 2010, 11, 37–40. [Google Scholar]
  102. Ito, Y. Antifungal Compounds from Trees of the Genus Diospyros with Complete Assignment of Nuclear Magnetic Resonance Data. Mokuzai Gakkaishi 1995, 41, 694–698. [Google Scholar]
  103. Munissi, J.J.E. Cytotoxic and Antimicrobial Activities of the Constituents of Ten Plant Species from Tanzania. Tanzan. J. Sci. 2019, 45, 44–52. [Google Scholar]
  104. Cirera, J.; da Silva, G.; Serrano, R.; Gomes, E.; Duarte, A.; Silva, O. Antimicrobial Activity of Diospyros villosa Root. Planta Med. 2010, 76, P454. [Google Scholar] [CrossRef]
  105. Begum, S.; Munissi, J.J.E.; Buriyo, A.S.; Makangara, J.J.; Lucantoni, L.; Avery, V.M.; Erdelyi, M.; Nyandoro, S.S. Antiplasmodial, Antimicrobial and Cytotoxic Activities of Extracts from Selected Medicinal Plants Growing in Tanzania. J. Biol. Act. Prod. Nat. 2020, 10, 165–176. [Google Scholar] [CrossRef]
  106. 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]
  107. Adu, O.T.; Naidoo, Y.; Lin, J.; Adu, T.S.; Sivaram, V.; Dewir, Y.H.; El-Banna, A.N. Phytochemical Screening and Biological Activities of Diospyros villosa (L.) De Winter Leaf and Stem-Bark Extracts. Horticulturae 2022, 8, 945. [Google Scholar] [CrossRef]
  108. 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] [PubMed]
  109. Mukhtar, Y.; Aliyu, B.S.; Zakari, S.M.; Aliko, A.A.; Habib, A.S.; Zubairu, S.M.; Bashir, R.A.; Tukur, S.; Jalingo, A.S.; Abubakar, F.S.; et al. Phytochemical, Pharmacognostic and Acute Toxicity Study of Diospyros mespiliformis (African Ebony) Stem Bark. Biosci. J. 2022, 10, 28–40. [Google Scholar]
  110. Elgorashi, E. Screening of Medicinal Plants Used in South African Traditional Medicine for Genotoxic Effects. Toxicol. Lett. 2003, 143, 195–207. [Google Scholar] [CrossRef]
  111. Cirera, J. Contribution to the Pharmacognostic Characterization of Diospyros villosa Root. Master’s Thesis, Universidade de Lisboa, Lisboa, Portugal, 2012. [Google Scholar]
  112. Sanogo, R.; Crisafi, G.; Germanò, M.P.; de Pasquale, R.; Bisignano, G. Evaluation of Malian Traditional Medicines: Screening for Antimicrobial Activity. Ltd. Phytother. Res. 1998, 12, 154–156. [Google Scholar] [CrossRef]
  113. Dangoggo, S.M.; Hassan, L.; Shina, I.S.; Manga, S. Phytochemical Analysis and Antibacterial Screening of Leaves of Diospyros mespiliformis and Ziziphus spina-christi. J. Chem. 2012, 1, 31–37. [Google Scholar]
  114. Peres, M.A.; Macpherson, L.M.D.; Weyant, R.J.; Daly, B.; Venturelli, R.; Mathur, M.R.; Listl, S.; Celeste, R.K.; Guarnizo-Herreño, C.C.; Kearns, C.; et al. Oral diseases: A global public health challenge. Lancet. 2019, 394, 249–260. [Google Scholar] [CrossRef] [PubMed]
  115. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 354 Diseases and Injuries for 195 Countries and Territories, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef]
  116. Douglas, K. Ethnobotanical Medicinal Plants Used as Chewing Sticks among the Kenyan Communities. Br. J. Pharm. Res. 2016, 13, 1–8. [Google Scholar] [CrossRef]
  117. Mbaveng, A.T.; Kuete, V. Review of the Chemistry and Pharmacology of 7-Methyljugulone. Afr. Health Sci. 2014, 14, 201. [Google Scholar] [CrossRef]
  118. Padhye, S.; Dandawate, P.; Yusufi, M.; Ahmad, A.; Sarkar, F.H. Perspectives on Medicinal Properties of Plumbagin and Its Analogs. Med. Res. Rev. 2012, 32, 1131–1158. [Google Scholar] [CrossRef]
  119. Isnansetyo, A.; Putri Handayani, D.; Istiqomah, I.; Arif, A.; Kaneko, T. An Antibacterial Compound Purified from a Tropical Coastal Plant, Diospyros maritima. Biodiversitas 2021, 23, 135–142. [Google Scholar] [CrossRef]
  120. Karkare, S.; Chung, T.T.H.; Collin, F.; Mitchenall, L.A.; McKay, A.R.; Greive, S.J.; Meyer, J.J.M.; Lall, N.; Maxwell, A. The Naphthoquinone diospyrin Is an Inhibitor of DNA Gyrase with a Novel Mechanism of Action. J. Biol. Chem. 2013, 288, 5149–5156. [Google Scholar] [CrossRef] [PubMed]
  121. Kuete, V.; Tangmouo, J.G.; Marion Meyer, J.J.; Lall, N. Diospyrone, crassiflorone and plumbagin: Three Antimycobacterial and Antigonorrhoeal Naphthoquinones from Two Retrieved from Diospyros spp. Int. J. Antimicrob. Agents 2009, 34, 322–325. [Google Scholar] [CrossRef]
  122. Kuete, V.; Ngameni, B.; Tangmouo, J.G.; Bolla, J.-M.; Alibert-Franco, S.; Ngadjui, B.T.; Pagès, J.-M. Efflux Pumps Are Involved in the Defense of Gram-Negative Bacteria against the Natural Products Isobavachalcone and Diospyrone. Antimicrob. Agents Chemother. 2010, 54, 1749–1752. [Google Scholar] [CrossRef]
  123. Tangmouo, J.G.; Lontsi, D.; Ngounou, F.N.; Kuete, V.; Meli, A.L.; Manfouo, R.N.; Kamdem W., H.; Tane, P.; Beng, V.P.; Sondengam, B.L.; et al. Diospyrone, a new coumarinylbinaphthoquinone from Diospyros canaliculata (Ebenaceae): Structure and Antimicrobial Activity. Bull. Chem. Soc. Ethiop. 2005, 19, 81–88. [Google Scholar]
  124. Janeczko, M.; Kubiński, K.; Martyna, A.; Muzyczka, A.; Boguszewska-Czubara, A.; Czernik, S.; Tokarska-Rodak, M.; Chwedczuk, M.; Demchuk, O.M.; Golczyk, H.; et al. 1,4-Naphthoquinone Derivatives Potently Suppress Candida albicans Growth, Inhibit Formation of Hyphae and Show No Toxicity toward Zebrafish Embryos. J. Med. Microbiol. 2018, 67, 598–609. [Google Scholar] [CrossRef]
  125. Dzoyem, J.P.; Tangmouo, J.G.; Lontsi, D.; Etoa, F.X.; Lohoue, P.J. In vitro Antifungal Activity of Extract and Plumbagin from the Stem Bark of Diospyros crassiflora Hiern (Ebenaceae). Phytother. Res. 2007, 21, 671–674. [Google Scholar] [CrossRef] [PubMed]
  126. Dzoyem, J.P.; Kechia, F.A.; Kuete, V.; Pieme, A.C.; Akak, C.M.; Tangmouo, J.G.; Lohoue, P.J. Phytotoxic, Antifungal Activities and Acute Toxicity Studies of the Crude Extract and Compounds from Diospyros canaliculata. Nat. Prod. Res. 2011, 25, 741–749. [Google Scholar] [CrossRef] [PubMed]
  127. Surapuram, V.; Setzer, W.N.; McFeeters, R.L.; McFeeters, H. Antifungal Activity of Plant Extracts against Aspergillus niger and Rhizopus stolonifer. Nat. Prod. Commun. 2014, 9, 1934578X1400901. [Google Scholar] [CrossRef]
  128. Ilaria, C.; Queiroz, E.; Brusotti, G.; Favre-Godal, Q.; Caccialanza, G.; Moundipa, P.; Wolfender, J. Antifungal Compounds Isolated from Diospyros bipindensis. Planta Med. 2012, 78, PI427. [Google Scholar] [CrossRef]
  129. Cesari, I.; Queiroz, E.F.; Favre-Godal, Q.; Marcourt, L.; Caccialanza, G.; Moundipa, P.F.; Brusotti, G.; Wolfender, J.-L. Extensive Phytochemical Investigation of the Polar Constituents of Diospyros bipindensis Gürke Traditionally Used by Baka Pygmies. Phytochemistry 2013, 96, 279–287. [Google Scholar] [CrossRef]
  130. Cesari, I.; Hoerlé, M.; Simoes-Pires, C.; Grisoli, P.; Queiroz, E.F.; Dacarro, C.; Marcourt, L.; Moundipa, P.F.; Carrupt, P.A.; Cuendet, M.; et al. Anti-Inflammatory, Antimicrobial and Antioxidant Activities of Diospyros bipindensis (Gürke) Extracts and Its Main Constituents. J. Ethnopharmacol. 2013, 146, 264–270. [Google Scholar] [CrossRef]
  131. Wang, X.; Habib, E.; León, F.; Radwan, M.M.; Tabanca, N.; Gao, J.; Wedge, D.E.; Cutler, S.J. Antifungal Metabolites from the Roots of Diospyros virginiana by Overpressure Layer Chromatography. Chem. Biodivers 2011, 8, 2331–2340. [Google Scholar] [CrossRef]
  132. Prajoubklang, A.; Sirithunyalug, B.; Charoenchai, P.; Suvannakad, R.; Sriubolmas, N.; Piyamongkol, S.; Kongsaeree, P.; Kittakoop, P. Bioactive Deoxypreussomerins and Dimeric Naphthoquinones from Diospyros ehretioides Fruits: Deoxypreussomerins May Not Be Plant Metabolites But May Be from Fungal epiphytes or endophytes. Chem. Biodivers 2005, 2, 1358–1367. [Google Scholar] [CrossRef]
  133. Olasehinde, G.I.; Ojurongbe, O.; Adeyeba, A.O.; Fagade, O.E.; Valecha, N.; Ayanda, I.O.; Ajayi, A.A.; Egwari, L.O. In vitro Studies on the Sensitivity Pattern of Plasmodium falciparum to Anti-Malarial Drugs and Local Herbal Extracts. Malar. J. 2014, 13, 63. [Google Scholar] [CrossRef]
  134. Hazra, B.; Ghosh, R.; Banerjee, A.; Kirby, G.C.; Warhurst, D.C.; Phillipson, J.D. In vitro Antiplasmodial Effects of diospyrin, a Plant-Derived naphthoquinoid, and a Novel Series of Derivatives. Phytother. Res. 1995, 9, 72–74. [Google Scholar] [CrossRef]
  135. Uliassi, E.; Fiorani, G.; Krauth-Siegel, R.L.; Bergamini, C.; Fato, R.; Bianchini, G.; Carlos Menéndez, J.; Molina, M.T.; López-Montero, E.; Falchi, F.; et al. Crassiflorone Derivatives That Inhibit Trypanosoma brucei glyceraldehyde-3-phosphate dehydrogenase (Tb GAPDH) and Trypanosoma cruzi trypanothione reductase (Tc TR) and Display Trypanocidal Activity. Eur. J. Med. Chem. 2017, 141, 138–148. [Google Scholar] [CrossRef] [PubMed]
  136. Kantamreddi, V.S.S.; Wright, C.W. Investigation of Indian Diospyros Species for Antiplasmodial Properties. Evid.-Based Complement. Altern. Med. 2008, 5, 187–190. [Google Scholar] [CrossRef] [PubMed]
  137. Rajarajeshwari, N.; Ganapaty, S.; Harish Kumar, D.H. In vitro Anthelmintic Activity of Five Rare Species of Diospyros. Int. J. Pharm. Sci. 2010, 2, 445–447. [Google Scholar]
  138. Amar Dev, M.J.; Rajarajeshwari, N.; Ganapaty, S.; Parixit, B.; Brun, R. Antiprotozoal and Anthelmintic Naphthoquinones from Three Unexplored Species of Diospyros. J. Nat. Remedies 2012, 12, 129–134. [Google Scholar]
  139. Yue, X.; Yang, H.; Wang, T.; Dong, S.; Sun, Z.; Wang, Y.; Luo, X.; Chen, B.; Yao, G.; Gao, Y.; et al. Molecules and Medical Function of Diospyros lotus L. Therm. Sci. 2020, 24, 1705–1712. [Google Scholar] [CrossRef]
  140. Ayoub, A.; Singh, J.; Hameed, F.; Mushtaq, M. Evaluation of Secondary Metabolites (Antibacterial and Antioxidant Activity) of Amlok (Diospyros lotus L) Fruit Extracts of Jammu Region. J. Pharm. Res. Int. 2021, 32, 8–19. [Google Scholar] [CrossRef]
  141. Yang, H.-Q.; Chen, G.-H.; Dong, S.; Sun, Z.; Wang, Y.; Luo, X.; Chen, B.; Yao, G.; Gao, Y.; Lv, C.; et al. Chemical Constituents and Medical Function of Leaves of Diospyros lotus L. Therm. Sci. 2020, 24, 1633–1639. [Google Scholar] [CrossRef]
  142. Rouf, R.; Uddin, S.J.; Shilpi, J.A.; Toufiq-Ur-Rahman, M.; Ferdous, M.M.; Sarker, S.D. Anti-Diarrhoeal Properties of Diospyros peregrina in the Castor Oil-Induced Diarrhoea Model in Mice. Ars. Pharm. 2006, 47, 81–89. [Google Scholar]
  143. Dewanjee, S.; Kundu, M.; Maiti, A.; Majumdar, R.; Majumdar, A.; Mandel, S.C. In Vitro Evaluation of Antimicrobial Activity of Crude Extract from Plants Diospyros peregrina, Coccinia grandis and Swietenia macrophylla. Trop. J. Pharm. Res. 2007, 6, 773–778. [Google Scholar] [CrossRef]
  144. Sinha, B.N.; Bansal, S.K.; Pattnaik, A.K. Phytochemical and Antimicrobial Activity of Extracts, Fractions and Betulin, 7-Methyl Juglone Obtained from Diospyros paniculata. J. Nat. Remedies 2009, 9, 99–102. [Google Scholar]
  145. Rao, N.H.; Lakshmidevi, N.; Pammi SV, N.; Kollu, P.; Ganapaty, S.; Lakshmi, P. Green Synthesis of Silver Nanoparticles Using Methanolic Root Extracts of Diospyros paniculata and Their Antimicrobial Activities. Mater. Sci. Eng. C 2016, 62, 553–557. [Google Scholar] [CrossRef]
  146. Hu, J.-F.; Garo, E.; Goering, M.G.; Pasmore, M.; Yoo, H.-D.; Esser, T.; Sestrich, J.; Cremin, P.A.; Hough, G.W.; Perrone, P.; et al. Bacterial Biofilm Inhibitors from Diospyros dendo. J. Nat. Prod. 2006, 69, 118–120. [Google Scholar] [CrossRef]
  147. Yogeeswari, P.; Sriram, D. Betulinic acid and Its Derivatives: A Review on Their Biological Properties. Curr. Med. Chem. 2005, 12, 657–666. [Google Scholar] [CrossRef] [PubMed]
  148. Baglin, I.; Mitaine-Offer, A.C.; Nour, M.; Tan, K.; Cave, C.; Lacaille-Dubois, M.A. A Review of Natural and Modified betulinic, ursolic and echinocystic acid derivatives as Potential Antitumor and Anti-HIV Agents. Mini-Rev. Med. Chem. 2003, 3, 525–539. [Google Scholar] [CrossRef]
  149. Fouokeng, Y.; Feumo Feusso, H.M.; Mbosso Teinkela, J.E.; Siwe Noundou, X.; Wintjens, R.; Isaacs, M.; Hoppe, H.C.; Krause, R.W.M.; Azebaze, A.G.B.; Vardamides, J.C. In vitro Antimalarial, Antitrypanosomal and HIV-1 Integrase Inhibitory Activities of Two Cameroonian Medicinal Plants: Antrocaryon klaineanum (Anacardiaceae) and Diospyros conocarpa (Ebenaceae). South Afr. J. Bot. 2019, 122, 510–517. [Google Scholar] [CrossRef]
  150. Juárez-Méndez, M.T.; Borges-Argáez, R.; Ayora-Talavera, G.; Escalante-Rebolledo, S.E.; Escalante-Erosa, F.; Cáceres-Farfán, M. Diospyros anisandra Phytochemical Analysis and Anti-Hemagglutinin-Neuraminidase Activity on Influenza AH1N1pdm09 Virus. Nat. Prod. Res. 2022, 36, 2666–2672. [Google Scholar] [CrossRef]
  151. Cetina-Montejo, L.; Ayora-Talavera, G.; Borges-Argáez, R. Zeylanone Epoxide Isolated from Diospyros anisandra Stem Bark Inhibits Influenza Virus in vitro. Arch. Virol. 2019, 164, 1543–1552. [Google Scholar] [CrossRef]
  152. Liu, M.; Yang, K.; Wang, J.; Zhang, J.; Qi, Y.; Wei, X.; Fan, M. Young Astringent Persimmon Tannin Inhibits Methicillin-Resistant Staphylococcus aureus Isolated from Pork. LWT 2019, 100, 48–55. [Google Scholar] [CrossRef]
  153. Maugeri, A.; Lombardo, G.E.; Cirmi, S.; Süntar, I.; Barreca, D.; Laganà, G.; Navarra, M. Pharmacology and Toxicology of tannins. Arch. Toxicol. 2022, 96, 1257–1277. [Google Scholar] [CrossRef] [PubMed]
  154. Arakawa, H.; Takasaki, M.; Tajima, N.; Fukamachi, H.; Igarashi, T. Antibacterial Activities of Persimmon Extracts Relate with Their Hydrogen Peroxide Concentration. Biol. Pharm. Bull. 2014, 37, 1119–1123. [Google Scholar] [CrossRef] [PubMed]
  155. Thatoi, H.N.; Panda, S.K.; Rath, S.K.; Dutta, S.K. Antimicrobial Activity and Ethnomedicinal Uses of Some Medicinal Plants from Similipal Biosphere Reserve, Orissa. Asian J. Plant Sci. 2008, 7, 260–267. [Google Scholar] [CrossRef]
  156. Dutta, S.; Panda, S.; Dubey, D. Anticandidal Activity of Diospyros melanoxylon Roxb. Bark from Similipal Biosphere Reserve, Orissa, India. Int. J. Green Pharm. 2010, 4, 102. [Google Scholar] [CrossRef]
  157. Supriya, K.A.; Growther, L. In-vitro Antioxidant and Antibacterial Activity of Different Extracts of Diospyros melanoxylon Roxb. Int. J. Pharm. Sci. Res. 2019, 10, 1820–1827. [Google Scholar]
  158. Kanta Rath, S.; Kumar Patra, J.; Gouda, S.; Kumar Sahoo, S.; Thatoi, H.; Kumar Dutta, S. Evaluation of Antioxidant Potential, Phytochemical Analysis and Chromatographic Separation of Bark Extracts of Diospyros melanoxylon Roxb. J. Biol. Act. Prod. Nat. 2014, 4, 377–390. [Google Scholar] [CrossRef]
  159. Kamaraj, C.; Rahuman, A.A.; Siva, C.; Iyappan, M.; Kirthi, A.V. Evaluation of Antibacterial Activity of Selected Medicinal Plant Extracts from South India against Human Pathogens. Asian Pac. J. Trop. Dis. 2012, 2, S296–S301. [Google Scholar] [CrossRef]
  160. Wangensteen, H.; Klarpås, L.; Alamgir, M.; Samuelsen, A.; Malterud, K. Can Scientific Evidence Support Using Bangladeshi Traditional Medicinal Plants in the Treatment of Diarrhoea? A Review on Seven Plants. Nutrients 2013, 5, 1757–1800. [Google Scholar] [CrossRef]
  161. Vijayan, G.S.; Chandra, J. Effect of Methanolic and Ethyl Acetate Leaf Extract of Diospyros discolor against gram positive and gram negative bacteria. J. Pharm. Sci. 2015, 8, 389–392. [Google Scholar]
  162. Dhawefi, N.; Jedidi, S.; Rtibi, K.; Jridi, M.; Sammeri, H.; Abidi, C.; Zouari, N.; Sebai, H. Antidiarrheal, Antimicrobial, and Antioxidant Properties of the Aqueous Extract of Tunisian Persimmon (Diospyros kaki Thunb.) Fruits. J. Med. Food 2021, 24, 1100–1112. [Google Scholar] [CrossRef]
  163. Tomiyama, K.; Mukai, Y.; Saito, M.; Watanabe, K.; Kumada, H.; Nihei, T.; Hamada, N.; Teranaka, T. Antibacterial Action of a Condensed tannin Extracted from Astringent Persimmon as a Component of Food Addictive Pancil PS-M on Oral Polymicrobial Biofilms. Biomed. Res. Int. 2016, 2016, 1–7. [Google Scholar] [CrossRef]
  164. Tahir, L.; Aslam, A.; Ahmed, S. Antibacterial Activities of Diospyros blancoi, Phoenix Dactylifera and Morus nigra against Dental Caries Causing Pathogens: An in vitro Study. Pak. J. Pharm. Sci. 2017, 30, 163–169. [Google Scholar] [PubMed]
  165. Senthilkumar, S.; Ashok, M.; Kashinath, L.; Sanjeeviraja, C.; Rajendran, A. Phytosynthesis and Characterization of TiO2 Nanoparticles Using Diospyros ebenum Leaf Extract and Their Antibacterial and Photocatalytic Degradation of Crystal Violet. Smart Sci. 2018, 6, 1–9. [Google Scholar] [CrossRef]
  166. Bharadwaj, K.K.; Rabha, B.; Pati, S.; Choudhury, B.K.; Sarkar, T.; Gogoi, S.K.; Kakati, N.; Baishya, D.; Kari, Z.A.; Edinur, H.A. Green Synthesis of Silver Nanoparticles Using Diospyros malabarica Fruit Extract and Assessments of Their Antimicrobial, Anticancer and Catalytic Reduction of 4-Nitrophenol (4-NP). Nanomaterials 2021, 11, 1999. [Google Scholar] [CrossRef]
  167. Polash, S.A.; Hamza, A.; Hossain, M.M.; Tushar, M.H.; Takikawa, M.; Shubhra, R.D.; Saiara, N.; Saha, T.; Takeoka, S.; Sarker, S.R. Diospyros malabarica Fruit Extract Derived Silver Nanoparticles: A Biocompatible Antibacterial Agent. Front. Nanotechnol. 2022, 4, 888444. [Google Scholar] [CrossRef]
  168. Ueda, K.; Kawabata, R.; Irie, T.; Nakai, Y.; Tohya, Y.; Sakaguchi, T. Inactivation of Pathogenic Viruses by Plant-Derived tannins: Strong Effects of Extracts from Persimmon (Diospyros kaki) on a Broad Range of Viruses. PLoS ONE 2013, 8, e55343. [Google Scholar] [CrossRef]
  169. Kamimoto, M.; Nakai, Y.; Tsuji, T.; Shimamoto, T.; Shimamoto, T. Antiviral Effects of Persimmon Extract on Human Norovirus and Its Surrogate, Bacteriophage MS2. J. Food Sci. 2014, 79, M941–M946. [Google Scholar] [CrossRef]
  170. Dewanjee, S.; Maiti, A.; Kundu, M.; Mandal, S.C. Evaluation of Anthelmintic Activity of Crude Extracts of Diospyros peregrina, Coccinia grandis, Schima wallichii. Dhaka Univ. J. Pharm. Sci. 1970, 6, 121–123. [Google Scholar] [CrossRef]
Figure 1. Diospyros rotundifolia: (a) aspect in its natural habitat; (b,c) details of leaf and fruit; (d) transverse view of the fruit with the seeds. Photography by Elsa Gomes.
Figure 1. Diospyros rotundifolia: (a) aspect in its natural habitat; (b,c) details of leaf and fruit; (d) transverse view of the fruit with the seeds. Photography by Elsa Gomes.
Plants 12 02833 g001
Figure 2. Diospyros villosa: (a) Aspect in the natural habitat; (b) cross-section of the root. Photography by Elsa Gomes (a) and Adriana Ribeiro (b).
Figure 2. Diospyros villosa: (a) Aspect in the natural habitat; (b) cross-section of the root. Photography by Elsa Gomes (a) and Adriana Ribeiro (b).
Plants 12 02833 g002
Figure 3. Traditional use of Diospyros species: (a) plant part used; (b) type of disease.
Figure 3. Traditional use of Diospyros species: (a) plant part used; (b) type of disease.
Plants 12 02833 g003
Figure 4. Diospyros representatives identified 1,4-naphthoquinones.
Figure 4. Diospyros representatives identified 1,4-naphthoquinones.
Plants 12 02833 g004
Figure 5. Diospyros identified representative lupan-type triterpenoids.
Figure 5. Diospyros identified representative lupan-type triterpenoids.
Plants 12 02833 g005
Figure 6. Diospyros dichrophylla (Gand.) De Winter: Detail of fruits in nature, Mandevo, Namaacha district, Maputo, 2010. Photography by Elsa Gomes.
Figure 6. Diospyros dichrophylla (Gand.) De Winter: Detail of fruits in nature, Mandevo, Namaacha district, Maputo, 2010. Photography by Elsa Gomes.
Plants 12 02833 g006
Figure 7. PRISMA flowchart of the screening process in the different databases.
Figure 7. PRISMA flowchart of the screening process in the different databases.
Plants 12 02833 g007
Table 1. Species of the genus Diospyros L. present in Mozambican Flora.
Table 1. Species of the genus Diospyros L. present in Mozambican Flora.
First Discription YearScientific Name AuthorCommon Name English/LocalMD 1IUCN 2
NZTMSGIMCDNp
1980D. anitaeF.Whitemalawi star apple/- LC
1911D. busseiGürkecoral star-berry/- NT
1935D. consolataeChiov.-/novolo LC
1963D. dichrophylla(Gand.) De Winterpoison star-apple/- LC
1933D. ferrea(Willd.) Bakh.-/- A
1962D. inhacaensisF.Whitecoastal jackal-berry/dodo LC
1988 D. kabuyeanaF.White -/- LC
1873D. kirkiiHiernlarge-leaved jackal-berry/
cula, fuma, jacualala, mucula, murriba, tendje
LC
1980D. mafiensisF.White-/- NT
1844D. mespiliformisHochst. ex A.DC.african ebony, jackal-berry/
muribariba, mucula, muquéué, murriparipa, mutona, mussuma
LC
1956D. quiloensis(Hiern) F.Whitecrocodile-bark jackal-berry/
midodo, murodo
LC
1873D. rotundifoliaHierndune star-apple/
impapa, mapiti, munhentze
Δ NE
1861D. senensisKlotzschspiny jackal-berry/
matamba, mudalima, tombatica
LC
1861D. squarrosaKlotzschrigid star-berry/cachenz’ere, mpomopo, senzasicana, sicana LC
1980D. truncatifoliaCaveneysquare-leaved star apple/
impope, mpope
LC
1873D. verrucosaHiernwarty star-apple/djacola, mkonhomo, nkalanongo, riparipa LC
1961D. whyteana(Hiern) P.Whitebladder-nut/- LC
1963D. zombensis(B.L.Burtt) F.Whitemalawi star-apple/- LC
1891 D. abyssinica subsp. abyssinica (Hiern) F.White giant diospyros/- LC
1988D. abyssinica subsp. attenuata *(Hiern) F.Whitegiant diospyros/- LC
1980D. abyssinica subsp. chapmaniorum(Hiern) F.Whitegiant diospyros/- LC
1837 D. loureiriana subsp. loureiriana aG.Dondye star-apple, sand star-apple/chipongoti, nhandima LC
1805D. lycioides Desf. subsp. sericea(Bernh.) De Wintereastern blue-bush, red star-apple/chitomatomana, m’dima LC
1968D. natalensis subsp. natalensis *(Harv.) Brenanacorn jackal-berry/- A
2009D. natalensis subsp. nummularia(Harv.) Brenan Jordaanacorn diospyros, acorn jackal-berry, granite jackal-berry **/- A
b D. usambarensis subsp. usambarensis /rufescensF.White-/aboba, kidanko, mpome, nhamudima, popa LC
c D. villosa (L.) var. villosaDe Winterhairy star-apple/nhachibabane, nhaurratane, chicanela, chicumbela, chibabane A
c D. villosa var. parvifolia (De Winter) De Winterhairy star-apple/- A
d D. consolatae-rotundifolia intermediates-- A
d D. kirkii-mespiliformis intermediates-- A
d D. sp. no. 1 sensu FZ-- A
1 Distribution in Mozambique (blue, MD) [10,11]; Common name local (green, MD) [12]: N—Niassa; Z—Zambezia; T—Tete; MS—Manica and Sofala; GI—Gaza–Inhambane; M—Maputo; CD—Cabo Delgado; Np—Nampula; * Not identified; ** Other names—small-leaved jackal berry, Tickey tree; ∇: Gaza; Δ: Sofala. 2 International Union for the Conservation of Nature: LC—least concern; NE—not evaluated; NT—near threatened [B2ab(iii)]; A—absent. WFO Plant List: a D. loureiroana G.Don subsp. loureiriana; b Synonym of D. loureiroana subsp. rufescens (Caveney) Verdc.; c Synonym of D. villosa (L.) De Winter; d not included in the WFO Plant List [10,11]; (-)—not available.
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

Ribeiro, A.; Serrano, R.; da Silva, I.B.M.; Gomes, E.T.; Pinto, J.F.; Silva, O. The Genus Diospyros: A Review of Novel Insights into the Biological Activity and Species of Mozambican Flora. Plants 2023, 12, 2833. https://doi.org/10.3390/plants12152833

AMA Style

Ribeiro A, Serrano R, da Silva IBM, Gomes ET, Pinto JF, Silva O. The Genus Diospyros: A Review of Novel Insights into the Biological Activity and Species of Mozambican Flora. Plants. 2023; 12(15):2833. https://doi.org/10.3390/plants12152833

Chicago/Turabian Style

Ribeiro, Adriana, Rita Serrano, Isabel B. Moreira da Silva, Elsa T. Gomes, João F. Pinto, and Olga Silva. 2023. "The Genus Diospyros: A Review of Novel Insights into the Biological Activity and Species of Mozambican Flora" Plants 12, no. 15: 2833. https://doi.org/10.3390/plants12152833

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop