Next Article in Journal
Characteristics of Hospitalized Elderly Patients with Severe Pneumonia Due to SARS-CoV-2, Vaccinated Against COVID-19
Next Article in Special Issue
Putting Ethnobotany into Practice: In Vitro Antioxidant Potential and Impact on Rat Gastric Smooth Muscle Reactivity of Aqueous Extracts of Marrubium friwaldskyanum Boiss. and Marrubium peregrinum L.
Previous Article in Journal
Literature Review of Prognostic Factors in Secondary Generalized Peritonitis
Previous Article in Special Issue
Comparison of Major Compounds in Essential Oils Steam Distilled from Fresh Plant Material of South African Hop Varieties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Moringa oleifera Lam.: A Nutritional Powerhouse with Multifaceted Pharmacological and Functional Applications

by
Natalina Panova
1,
Anelia Gerasimova
2,
Galia Gentscheva
3,*,
Stoyanka Nikolova
4,
Lubomir Makedonski
2,
Margarita Velikova
5,
Abdessamad Beraich
6,
Abdelmonaem Talhaoui
6,
Nadezhda Petkova
7,
Daniela Batovska
8 and
Krastena Nikolova
1,*
1
Department of Physics and Biophysics, Faculty of Pharmacy, Medical University of Varna, 84 Tzar Osvoboditel, 9000 Varna, Bulgaria
2
Department of Chemistry, Faculty of Pharmacy, Medical University of Varna, 84 Tzar Osvoboditel, 9000 Varna, Bulgaria
3
Department of Chemistry and Biochemistry, Medical University-Pleven, 1 Saint Kliment Ohridski Street, 5800 Pleven, Bulgaria
4
Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 4000 Plovdiv, Bulgaria
5
Department of Physiology and Pathofysiology, Faculty of Medicine, Medical University of Varna, 55 Marin Drinov, 9000 Varna, Bulgaria
6
Laboratory of Environment and Applied Chemistry (LCAE), Team: Physical Chemistry of the Natural Resources and Processes, Department of Chemistry, Faculty of Sciences, Mohamed First University, Oujda 60000, Morocco
7
Department of Organic Chemistry and Inorganic Chemistry, University of Food Technologies, 26 Maritsa Blvd., 4002 Plovdiv, Bulgaria
8
Institute of Chemical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 103, 1113 Sofia, Bulgaria
*
Authors to whom correspondence should be addressed.
Life 2025, 15(6), 881; https://doi.org/10.3390/life15060881
Submission received: 27 March 2025 / Revised: 23 May 2025 / Accepted: 27 May 2025 / Published: 29 May 2025
(This article belongs to the Special Issue Bioactive Natural Compounds: Therapeutic Insights and Applications)

Abstract

:
Moringa oleifera, often referred to as the “miracle tree”, has gained widespread recognition for its exceptional nutritional profile and broad pharmacological potential. This review provides a comprehensive synthesis of the plant’s botanical characteristics, taxonomy, cultivation practices, and biochemical composition. Special emphasis is placed on its rich content of bioactive secondary metabolites-such as flavonoids, alkaloids, phenolic acids, saponins, isothiocyanates, and glucosinolates-which underlie its diverse therapeutic effects. The paper compiles and analyzes evidence from over 200 peer-reviewed studies, documenting antioxidant, anti-inflammatory, antimicrobial, antidiabetic, anticancer, hepatoprotective, neuroprotective, and anti-obesity effects, among others. For instance, leaf extracts have demonstrated potent antioxidant and antidiabetic effects in both animal models and clinical trials, while seed-derived isothiocyanates have shown significant antibacterial and anticancer activity. In addition, clinical and in vivo data support M. oleifera’s role in fertility regulation, cardiovascular protection, and neurodegenerative disease mitigation. Beyond its medicinal applications, the review highlights its growing use in functional foods, dietary supplements, and cosmeceutical products, reflecting its commercial and industrial relevance. By consolidating findings across disciplines, this review underscores the multifaceted value of M. oleifera as a nutraceutical and therapeutic resource.

1. Introduction

Moringa oleifera Lam, commonly known as the “miracle tree”, is a fast-growing, drought-tolerant species native to the Indian subcontinent and now cultivated extensively across tropical and subtropical regions [1]. Its reputation as a multipurpose plant stems from the fact that nearly every part—including leaves, seeds, pods, bark, flowers, and roots—has traditional or modern applications in food, medicine, agriculture, and cosmetics. The plant thrives in marginal soils, requires minimal inputs, and tolerates prolonged dry periods, making it an important candidate for sustainable agriculture in regions facing malnutrition, climate variability, and land degradation [2].
In recent decades, M. oleifera has drawn increasing global attention due to its exceptional nutritional and pharmacological potential. Given the accelerating volume of research on M. oleifera, driven by its broad utility and increasing global relevance, there is a pressing need to critically evaluate and synthesize emerging findings. Such an integrative review will help contextualize recent advances, highlight knowledge gaps, and facilitate targeted investigations across scientific, clinical, and agricultural domains. The leaves, in particular, are valued for their high content of proteins, essential amino acids, vitamins (A, B-complex, C, and E), and minerals such as calcium, potassium, iron, and magnesium [2]. These properties have led to its incorporation into feeding programs, food fortification strategies, and dietary supplements, particularly in low-resource settings.
Phytochemical analyses of M. oleifera have identified a broad range of bioactive compounds, including flavonoids, phenolic acids, glucosinolates, carbamates, tannins, and saponins [3,4,5]. These constituents contribute to the plant’s wide spectrum of pharmacological activities, such as antioxidant, anti-inflammatory, antimicrobial, antidiabetic, anticancer, hepatoprotective, cardioprotective, and neuroprotective effects [2,3,4,5,6]. Extracts from various plant parts have shown promising results in the prevention and management of chronic diseases, including diabetes, cancer, and neurodegenerative disorders [7]. Additional studies have reported antiepileptic, wound-healing, and immunomodulatory properties [6,8]. Moreover, the high oleic acid content of the leaves supports their use in cosmeceutical formulations, providing moisturizing, emollient, and skin-protective effects [9].
As a result of these combined attributes, M. oleifera has become a focal point in the development of functional foods, herbal therapeutics, and natural personal care products. Its inclusion in climate-resilient cropping systems and its use in reforestation, soil rehabilitation, and carbon sequestration initiatives further enhance its significance on both the health and environmental fronts.
Despite the growing number of reviews on M. oleifera L., most have concentrated on specific, narrowly defined themes. For example, Berg and Kuipers systematically assessed its antibacterial properties [10], while Chiș et al. focused on the anti-inflammatory mechanisms of key bioactive compounds such as quercetin, kaempferol, and isothiocyanates [11]. Other authors reviewed its nutraceutical and cosmeceutical applications [12], Abdelwanis et al. addressed its industrial and agricultural value [13], particularly in Egypt [14,15], and Elsadek et al. explored its regenerative and osteogenic effects in oral medicine [14]. Singh et al. examined its anticancer potential from a phytochemical and pharmacological perspective [15], while Anzano et al. provided a broader overview of its botany and medicinal applications [16]. However, an integrated and updated synthesis that bridges these domains is still lacking.
This review addresses that gap by offering a comprehensive, multidisciplinary analysis of M. oleifera, covering its botanical identity, taxonomy, cultivation strategies, phytochemical diversity, and biological activities, with particular emphasis on its role as a functional food crop and its agronomic adaptability beyond tropical regions. By consolidating recent findings from diverse research fields, we aim to support researchers, agricultural practitioners, and product developers in harnessing the full nutritional, therapeutic, and ecological potential of M. oleifera in both developing and developed contexts.

2. Methodology

This review is based on a comprehensive literature search conducted to collect and analyze scientific publications related to M. oleifera from 2010 up to February 2025. Databases including PubMed, Scopus, Science Direct, Web of Science, and Google Scholar were used to identify relevant studies. The search terms included combinations of keywords such as “Moringa oleifera”, “nutritional composition”, “phytochemistry”, “pharmacological activity”, “bioactive compounds”, “antioxidant”, “anti-inflammatory”, “traditional medicine”, and “functional foods”.
Only peer-reviewed journal articles published in English were included. Review articles and original research papers describing the botanical, nutritional, chemical, pharmacological, or commercial aspects of M. oleifera were considered. Studies based on in vitro, in vivo, or clinical evaluations were prioritized when discussing pharmacological effects. Articles were excluded if they were duplicate entries, non-peer-reviewed materials, or outside the scope of the review.
The selected studies were critically evaluated and organized thematically to provide a structured synthesis of the current knowledge on M. oleifera, with attention to both traditional uses and modern applications.

3. Botanical Identity, Taxonomy, and Cultivation

3.1. Botanical Description

M. oleifera is a fast-growing, deciduous tree that typically reaches up to 8 m in height. It has a twisted trunk with smooth, dark gray to yellow bark and an umbrella-shaped crown. The compound leaves are bipinnate or tripinnate, with ellipsoid leaflets that are dark green on the upper surface and pale green underneath, measuring up to 2.5 cm in length. The plant produces fragrant, creamy-white flowers arranged in loose inflorescences up to 15 cm long. Its fruit is a long, three-sided capsule commonly referred to as a “drumstick”, measuring up to 90 mm in length and 12 mm in width, containing light brown seeds that germinate within a week under suitable conditions [17].
Common names for M. oleifera vary widely across countries and languages, reflecting its broad geographic distribution and cultural importance [18,19]. These are presented in Table 1.

3.2. Taxonomy

The taxonomic classification of M. oleifera is summarized in Table 2, [20,21,22].
The species of the genus Moringa include: M. oleifera, M. concanensis, M. drouhardii, M. arborea, M. borziana, M. hildebrandtii, M. longituba, M. pygmaea, M. rivae, M. ruspoliana, M. ovalifolia, M. peregrina, and M. stenopetala.

3.3. Cultivation and Agronomical Practices

3.3.1. Growing Conditions

M. oleifera is a highly adaptable species that thrives in tropical and subtropical regions, growing well from sea level up to 1000 m in elevation. It tolerates a wide range of temperatures—from 12 to 40 °C—and can withstand short-term extremes as low as 1–3 °C or as high as 48 °C [23]. It grows in diverse soils—including sandy, loamy, and semi-arid types—but performs best in well-drained soils with near-neutral pH, tolerating pH values between 5.0 and 9.0. It is also capable of surviving in nutrient-poor and degraded soils, making it suitable for cultivation in areas impacted by land degradation or climate stress [24].

3.3.2. Cultivation

Propagation of M. oleifera can be achieved through seeds, hardwood cuttings, or nursery-grown transplants. Seeds germinate within two weeks when sown at shallow depths (up to 2 cm), whereas hardwood cuttings (1–2 m long, 4–16 cm thick) taken from mature trees during the rainy season root easily under moist conditions [24]. Trees grown from seeds develop deeper root systems, which enhance drought resilience, while cuttings establish more rapidly but produce shallower roots [23,25]. Germination and vegetative growth are optimal at day/night temperatures of approximately 30/20 °C [26].
Due to its rapid growth—up to 3 m in just three months—M. oleifera supports multiple harvest cycles annually. Pruning or pollarding is commonly practiced promoting lateral branching, facilitating harvest, and improving regrowth. Moderate pruning, in particular, has been shown to significantly increase leaf biomass compared to light or heavy pruning [26].
Planting density is tailored to the production goal:
  • Planting density is usually tailored to the production objective. For example, for intensive leaf production: spacing is 10–20 cm, with harvest every 35–45 days, and requires irrigation and fertilization; for semi-intensive systems: spacing is approximately 50 × 100 cm, with harvest every 50–60 days and moderate inputs; for agroforestry systems: 2–4 m between rows, designed for low-input integration into wider farming systems.
  • Yields vary widely depending on genotype, climate, and spacing, with intensive plantations producing between 40 and 580 metric tons of fresh biomass per hectare per year [23]. Shoots are typically harvested at 0.5–1 m height to stimulate regrowth, while harvesting individual leaves—although faster—may reduce vigor over time.
For seed production, wider spacing (2.5–3 m) is recommended to maximize pod development. Pods mature about three months after flowering and must be harvested promptly to avoid seed loss. A single tree can yield 15,000–25,000 seeds per year, depending on cultivar and environmental conditions [27,28].
Beyond traditional tropical zones, M. oleifera has demonstrated excellent adaptability to temperate climates. In Bulgaria, Indian-developed PKM1 and PKM2 cultivars have been successfully introduced under monitored greenhouse and field conditions, allowing leaf harvests at a plant height of ~50 cm [24,29,30,31,32]. Similarly, trials in Portugal and Spain have shown the species can be cultivated in Mediterranean climates, provided frost protection is in place during colder months [33,34]. In Portugal, M. oleifera has been successfully cultivated under temperate conditions, demonstrating good agronomic performance in Mediterranean climates. Its high carbon dioxide sequestration capacity-reported to be up to 20 times greater than that of typical vegetation—further supports its potential as a sustainable crop in climate-smart agriculture [34].

4. Nutritional Profile

M. oleifera leaves are widely acknowledged for their exceptional nutritional richness, containing over 90 phytonutrients, including essential amino acids, proteins, vitamins, and minerals. Among these, vitamins have been extensively studied across different leaf forms—fresh, dried, and powdered. Leaf powder, commonly available in health food stores, is typically derived from shade- or sun-dried leaves [35].
Fresh M. oleifera leaves are particularly rich in vitamins E, A, and C. Vitamin E is the most abundant, with concentrations reaching 448 mg/100 g in raw leaves—significantly higher than any other vitamin. Although some loss occurs during drying, powdered leaves still retain a substantial amount (113 mg/100 g). In comparison, fresh leaves contain approximately four times more vitamin A than carrots and seven times more vitamin C than oranges [35]. In addition, M. oleifera leaves provide notable amounts of B vitamins, including pyridoxine (B6), nicotinic acid (B3), and folic acid (B9), which support various metabolic and cellular functions [36]. Table 3 summarizes the concentrations of key vitamins across raw, dried, and powdered leaf forms [37].
The vitamin content of M. oleifera also shows considerable geographical variation. For instance, vitamin C levels range from 0.04% to 0.12%, with the highest values reported in samples from the United Kingdom, Jordan, and South Africa, and undetectable levels in samples from Nigeria [38]. A similar trend is observed for tocopherol content in M. oleifera oil: the highest concentrations of α- and β-tocopherol have been reported in samples from Ethiopia and Kenya, while γ-tocopherol levels show considerable variability, ranging from 4.47 to 93.70 mg/kg [39,40,41,42,43,44]. For example, Pluhackova et al. reported 2.6 mg/kg of β-tocopherol in Ethiopian samples [39], whereas Lalas et al. found a significantly higher level of 15.5 mg/kg in Kenyan samples [41]. Such variation is likely attributable to differences in plant genotype, environmental conditions, and harvest timing (Figure 1).
The vitamin C content of M. oleifera leaves has been studied in different countries [38]. The data are presented in Figure 2. The relative content varies in the range of 0.04 to 0.12%. The highest content is in the United Kingdom, followed by Jordan and South Africa. There is no vitamin C in samples from Nigeria.
Figure 2 summarizes the results for vitamins and tocopherols in fresh and dried M. oleifera leaves [45]: The vitamin C content in both fresh and dried leaves is nearly identical. However, in the case of vitamin E, the concentrations of α- and β-tocopherols are 8.3, 3, and 2.6 times higher in dried leaves compared to fresh ones (Figure 3).
M. oleifera leaves are also an excellent source of essential minerals (Table 4), particularly calcium and potassium, both critical for muscle contraction and nerve signaling. High calcium levels support bone health, while magnesium plays a key role in energy metabolism. The leaves also contain notable amounts of iron, essential for oxygen transport and red blood cell formation. According to Gopalan et al. [46], M. oleifera leaves provide three times more potassium than bananas, four times more calcium than milk, and twice the protein content of yogurt. Dried leaves are particularly rich in iron, 25 times more than spinach [36].
A comparative review by Kashyap et al. [37] further confirmed that dried leaves contain the highest concentrations of phosphorus, magnesium, iron, and potassium, along with significantly greater levels of fats, fiber, carbohydrates, and caloric content than fresh leaves and extract powders. This enhanced nutrient density underscores the value of dried M. oleifera leaves as a concentrated dietary supplement.
The protein content of dried M. oleifera leaves reaches approximately 29.4 g/100 g, making them a superior plant-based protein source, exceeding even chia seeds in protein density [30]. Their caloric value ranges from 205 to 350 kcal per 100 g, depending on processing and origin (Figure 4).
Amino acid profiling has revealed that different plant parts of M. oleifera exhibit distinct profiles: leucine is predominant in both the stem (5.12 g/100 g) and the root (5.33 g/100 g), while arginine is most abundant in the leaves (8.22 g/100 g) [31], underscoring the plant’s nutritional diversity.
The data presented in Figure 5 indicate that the ratio of mineral content differs among fresh leaves, raw seeds, powdered leaves, and pods. In fresh leaves, copper and zinc are predominant, whereas in powdered (dried) leaves, calcium, iron, and magnesium are present in the highest concentrations. Raw seeds are particularly rich in phosphorus.

5. Phytochemical Composition and Associated Bioactivities

The phytochemical composition of M. oleifera varies significantly across different plant parts and extraction methods (Table 5). Aqueous and alcoholic extracts of leaves, seeds, flowers, roots, and pods have yielded a wide spectrum of secondary metabolites with established or potential bioactivity. The major classes of compounds include flavonoids, carbamates, glucosinolates, and phenolic acids, each exhibiting organ-specific profiles and contributing to distinct therapeutic effects [3,49,50].
Figure 6 compares the total content of phenolic acids and flavonoids in plants from different countries. The data is from research by [38].
Extracts from Madagascar have the highest flavonoid content (around 30%), followed by the ones from South Africa and Colombia. For phenolic acids, there is no such sharply defined boundary. In 2024, [49] investigated the phytochemical composition of M. oleifera leaves, bark, and pods in polar solvents. Figure 7, Figure 8 and Figure 9 present the results of their studies using water, ethanol, and methanol extracts.
The study by [49] shows that the concentration of secondary metabolites changes when different solvents are used. The aqueous extract from all parts of the plant yields the maximum amounts of alkaloids, tannins, phenols, terpenoids, and sterols. Plant pod extracts contain the most alkaloids, while bark extracts contain the most tannins. All the aforementioned compounds have the highest content in the extracts from the leaves. Lu et al. [50] demonstrate that anthocyanin concentration is highest in the bark (52.80 mg/g), followed by the flowers (40.90 mg/g) and the leaves (9.40 mg/g) [38]. Figure 10 presents the phenolic and flavonoid content of M. Oleifera leaves, flowers, and bark. The flowers have the highest antioxidant activity (AOA) using the DPPH method (405 µg/mL), followed by the leaves (610 µg/mL) and finally the bark (890 µg/mL).
Flavonoids are primarily found in the flowers and leaves of the plant. The main flavonoids in the leaves are myricetin, rutin, apigenin, quercetin, and kaempferol [51]. These compounds contribute significantly to the antioxidant activity of the extracts. Wang et al. discovered that using a reflux condenser to extract these compounds from leaves at a subcritical temperature could increase the extraction by 26.7% [52]. The seeds are rich in α-tocopherols, γ-tocopherols, and δ-tocopherols [53,54]. In addition to the aforementioned chemicals, the leaves also contain linolenic acid, myristic acid, oleic acid, palmitic acid, α-linolenic acid, and α-linolenic acid [48]. Other authors expand the fatty acid composition to include eicosanoid acid, erucic acid, and palmitic acid [53].

6. Therapeutic Potential of M. oleifera Lam.

6.1. Antioxidant Activity

6.1.1. In Vitro Antioxidant Activity

Moringa oleifera contains a wide range of antioxidant compounds, including flavonoids, phenolic acids, tannins, carbamates, and isothiocyanates [24,55,56]. Its polyphenol content has been reported to surpass that of commonly consumed vegetables such as spinach, broccoli, and cabbage [57]. Numerous in vitro studies support the antioxidant potential of extracts derived from various plant parts, with the highest activity consistently attributed to the leaves due to their rich polyphenolic and flavonoid content. Lower antioxidant activity has been observed in the stems, roots, and seeds [58,59,60,61,62,63]. However, variations in assay methods, extraction solvents, and growing conditions often influence reported results [61] (Figure 11).
Table 6 presents a comparative overview of antioxidant activity across different plant parts and solvent systems. Leaf extracts showed the strongest antioxidant potential in all tested assays, particularly when extracted with ethyl acetate (IC50 = 5.72 µg/mL) or crude methanol (35.42 µg/mL). In contrast, extracts from stems and roots showed weaker activity, and those from seeds were generally the least effective. Solvent polarity also played a crucial role: medium-polar solvents like ethyl acetate were highly effective at concentrating flavonoids, whereas methanol-based extracts, while broader in scope, were often less potent. Notably, dry leaves demonstrated strong antioxidant performance in both FRAP and ORAC assays, suggesting their suitability for nutraceutical applications. Nonetheless, differences in extraction protocols and assay designs caution against direct cross-study comparisons.
Collectively, these findings underscore the prominence of M. oleifera leaves as the most antioxidant-rich plant part and highlight the critical importance of solvent selection in maximizing extract potency. Methodological discrepancies, such as differences in plant origin and assay calibration, must be considered when interpreting absolute values.

6.1.2. Cellular and In Vivo Evidence for Antioxidant Activity

Oxidative stress, characterized by the excessive accumulation of reactive oxygen species (ROS), disrupts cellular homeostasis and contributes to cellular damage and death. Studies have shown that M. oleifera leaf extract enhances antioxidant defenses both in vitro and in vivo. In a mouse C2C12 myoblast model treated with hydrogen peroxide (H2O2), leaf extract significantly increased total antioxidant capacity and improved the redox status by elevating the ratio between reduced glutathione (GSH) and oxidized glutathione (GSSG), a key marker of intracellular redox balance. This intervention also enhanced cell viability and regenerative capacity following oxidative injury [38].
Flavonoids such as quercetin are largely responsible for these effects. In A549 lung epithelial cells stimulated with lipopolysaccharide (LPS), quercetin reduced pro-inflammatory cytokines (TNF-α, IL-1, IL-6), suppressed NF-κB nuclear translocation, and downregulated NOX2 expression—a major ROS-generating enzyme—at both mRNA and protein levels [66].
In vivo, the administration of aqueous leaf extract mitigated Abamectin-induced oxidative disturbances in mice, restoring acetylcholinesterase and glutathione S-transferase activities in the brain and elevating reduced GSH levels in both brain tissue and erythrocytes [67].

6.2. Anti-Inflammatory Activity

Numerous in vitro and in vivo studies confirm the anti-inflammatory activity of M. oleifera. The anti-inflammatory effects of the plant are associated with the content of flavonoids (quercetin, kaempferol), phenolic acids (e.g., chlorogenic acid), isothiocyanates, tannins, and saponins. A detailed description of the phytochemicals contained in the plant and the mechanisms through which they inhibit inflammation has been presented by [11]. The biologically active compounds suppress inflammatory processes through several mechanisms. Many of the antioxidant compounds in the plant, such as quercetin and kaempferol, also contribute to its anti-inflammatory effects, highlighting the interconnected nature of these activities. The above-mentioned flavonoids inhibit key pro-inflammatory enzymes like cyclooxygenase (COX) and lipoxygenase and downregulate cytokines such as IL-1β, IL-6, PGE2, and TNF-α in RAW 264.7 macrophages [11]. β-sitosterol (BSS) is a widespread phytosterol found in many plant species, its anti-inflammatory activity has been investigated in formulations containing M. oleifera-derived BSS. In vitro studies demonstrate that BSS nanoparticles can suppress the secretion of inflammatory mediators such as TNF-α, IL-1β, and ROS in keratinocytes and macrophages [68].
A hydroethanolic extract of M. oleifera flowers reduced NO, PGE2, and cytokine production in LPS-stimulated macrophages, primarily through inhibition of the NF-κB signaling pathway [69,70]. Glucosinolates and their hydrolysis products, such as moringin, also contribute significantly to the anti-inflammatory effects [71,72]. Leaf tea preparations rich in moringin were shown to inhibit inducible nitric oxide synthase (iNOS) expression in LPS-stimulated mouse macrophages [73].
The anti-inflammatory potential of M. oleifera has been widely validated in animal models. Topical application of MIC (an isothiocyanate from seeds) and MSE (seed extract) in a TPA-induced mouse ear edema model significantly reduced pro-inflammatory cytokines, including IL-6, monocyte chemoattractant protein-1 (MCP-1), and keratinocyte chemoattractant (KC) [74].
In rat paw edema models, both aqueous and ethanolic leaf extracts reduced inflammation induced by carrageenan and egg albumin, showing efficacy comparable to ibuprofen and diclofenac [75,76]. Ezeamuzie et al. initially reported a 35–45% reduction in paw edema using root and leaf extracts [75].
In a streptozotocin-induced diabetic rat model, treatment with methanolic leaf extract led to a marked decrease in hepatic and renal levels of IL-1, IL-12, IL-18, and NF-κB, while also exhibiting antioxidant and anti-apoptotic effects [77].
Additional research demonstrated a bronchodilator and anti-asthmatic effect of methanolic leaf extract in ovalbumin-sensitized guinea pigs, as evidenced by a reduction in white blood cell count and histamine levels in lung tissue [78].
Beyond inflammation, M. oleifera extracts have shown analgesic effects, demonstrated using the hot plate and acetic acid writhing methods. Extracts from leaves, seeds, bark, and roots have been effective, with some studies suggesting dose-dependent anti-migraine potential [79,80,81].
These in vivo findings highlight the multi-faceted anti-inflammatory potential of M. oleifera extracts, especially from leaves and seeds. A summary of selected in vivo studies, extract types, and their effects is presented in Table 7.

6.3. Antimicrobial (Antibacterial, Antifungal) Activity

Among the different parts of M. oleifera, the leaves and seeds exhibit the highest antibacterial efficacy. Their extracts are active against a broad spectrum of Gram-positive and Gram-negative bacteria, underscoring the plant’s potential as a natural source of antibacterial agents. These antimicrobial effects are mediated through multiple mechanisms, including disruption of bacterial cell membranes, inhibition of key metabolic enzymes, interference with quorum sensing and biofilm formation, and induction of oxidative stress [61,82,83,84,85,86].
Moringa seed powder has demonstrated antibacterial activity against several pathogens, with the strongest effects observed against Salmonella spp. [87]. Flavonoids such as kaempferol and quercetin, identified in seed extracts, contribute significantly to this activity: kaempferol inhibited Klebsiella pneumoniae, Pseudomonas aeruginosa, and Streptococcus pneumoniae, while quercetin was most effective against Staphylococcus aureus [84]. Additionally, isothiocyanates isolated from seeds exhibited strong activity against Gram-positive bacteria, including S. aureus, S. epidermidis, and Bacillus subtilis [88]. Moringa extracts have also been shown to inhibit biofilm formation, a critical factor in persistent infections, particularly those involving S. aureus [86].
The antibacterial efficacy of M. oleifera can be further enhanced through combination with conventional antibiotics. Studies have demonstrated synergistic effects, particularly with ethanol leaf extracts. Ashraf et al. [89] reported that combining M. oleifera extracts with antibiotics such as gentamicin, ampicillin, enrofloxacin, and oxytetracycline significantly improved activity against E. coli, S. aureus, and Klebsiella spp., highlighting its potential to combat antibiotic resistance.
Nanotechnology-based formulations of M. oleifera further enhance its antibacterial properties. Zinc oxide nanoparticles synthesized using M. oleifera leaf extract (MOL: ZnO NPs) exhibited strong inhibitory effects against Pseudomonas spp. and B. cereus, primarily through membrane disruption mechanisms [90]. Similarly, silver nanoparticles (Ag-NPs) derived from M. oleifera showed superior antibacterial activity compared to crude aqueous extracts, suggesting that nanoparticle delivery systems can improve the bioavailability and potency of plant-derived antimicrobials [91].
In the context of oral health, M. oleifera extracts from seeds, roots, and leaves have demonstrated inhibitory activity against common oral pathogens such as Streptococcus mutans and S. aureus. Ethanol leaf extracts showed the greatest efficacy and have been incorporated into formulations like toothpaste and mouthwash [92]. Aqueous extracts have also been effective against bacteria associated with orthopedic wound infections, supporting their potential use in topical antimicrobial applications [93]. Clinically, M. oleifera bark extract significantly improved outcomes in patients with urinary tract infections, with a 67% recovery rate compared to 47% in the group receiving standard medications, highlighting its antibacterial and anti-inflammatory therapeutic potential [94].
Overall, the antibacterial activity of M. oleifera is well-supported by in vitro and clinical evidence. Its phytochemicals, particularly flavonoids and isothiocyanates, exert potent, multi-target effects against many pathogens. The plant’s ability to enhance conventional antibiotic efficacy, inhibit biofilms, and perform comparably to standard therapies in clinical settings positions it as a promising candidate for the development of plant-based antibacterial formulations. Continued investigation into standardized extraction methods and bioactive synergy will be essential to advance its integration into medical and pharmaceutical applications.
M. oleifera has demonstrated promising antifungal activity against a broad spectrum of human and phytopathogenic fungi (Table 8). Extracts from various plant parts, particularly the leaves, seeds, and oils, have shown efficacy against dermatophytes (e.g., Trichophyton, Microsporum, Epidermophyton spp.), yeasts (e.g., Candida spp., Saccharomyces cerevisiae), and molds (F. solani, F. oxysporum).
Aqueous and ethanolic leaf extracts inhibited C. tropicalis and S. cerevisiae, although early studies noted limited activity against C. albicans [95]. However, more recent investigations report significant antifungal effects against C. albicans, with ethanolic extracts yielding larger inhibition zones than aqueous preparations [96]. In vitro tests have also confirmed the efficacy of ethanol extracts against dermatophytes such as T. rubrum, T. mentagrophytes, E. floccosum, and M. canis [97].
M. oleifera oil has shown antifungal activity against multiple Candida species (e.g., C. albicans, C. dubliniensis, C. glabrata, C. kefyr, C. krusei, C. lusitaniae) isolated from stool samples of autistic children, suggesting therapeutic potential for gastrointestinal candidiasis [98]. Additionally, extracts from various plant parts have displayed fungicidal effects against several phytopathogenic fungi, underscoring potential applications as natural biofungicides in agriculture [99].
A notable mechanism of antifungal action involves chitin-binding proteins (CBPs) isolated from M. oleifera seeds. Mo-CBP3, for example, inhibited the growth of Fusarium solani, F. oxysporum, Colletotrichum musae, and C. gloeosporioides in vitro [100]. This protein demonstrated both fungistatic and fungicidal effects against F. solani, depending on the concentration used [101]. Additional CBPs, including Mo-CBP2 and Mo-CBP4, have shown potent activity against Candida spp. and dermatophytes. Mo-CBP4, in particular, reduced the severity of T. mentagrophytes infection in a murine dermatophytosis model by increasing membrane permeability, inducing reactive oxygen species (ROS), and damaging fungal cell walls [102,103].
Collectively, these findings highlight the potential of M. oleifera extracts and seed-derived CBPs as novel antifungal agents for both medical and agricultural applications.
Table 8. Antibacterial and antifungal effects of M. oleifera.
Table 8. Antibacterial and antifungal effects of M. oleifera.
TypePart of the PlantDoseReferences
Antibacterial effects
S. typhiLeaves (ethanolic extract)800 mg/mL[83]
S. aureus, E. faecalis, B. subtilis, S. typhi, E. coliLeaves (petroleum ether extract)62.5, 125, 250, 10,000 μg[59]
S. aureus, B. subtilis, E. coli, P. aeruginosaSeeds (aqueous and methanolic extracts)EC 5, 10, 20, 40%[83]
S. aureus, V. parahaemolyticus, E. faecalis, A. caviaeLeaves (aqueous and ethanolic extracts)400 μL (20 g/180 mL)[85]
Salmonella sp., E. coli, S. aureusSeed (powder)0.017 g/mL[87]
E. coli, K. pneumoniae, P. aeruginosa, S. pneumoniae, S. aureus.Seeds (methanolic extract)2, 4, 6 mg/mL[84]
S. aureus, S. epidermidis, B. subtilisIsothiocyanates isolated from seeds1, 10 mg/mL[88]
S. aureusLeaves (aqueous and saline extracts)100, 200 μg/mL[86]
K. pneumoniae, E. coli, S. aureus.Leaves (ethanolic extract)1000–3906 mg/mL[89]
L. monocytogenesMoringin, isolated from seeds0.124 mg/mL[104]
E. coli, S. sciuri, S. aureus, S. typhi, S. enterica, P. aeruginosaLeaves
(ethanolic extract,
aqueous extract)

0.04–0.42 mg/mL
0.03–0.33 mg/mL
[91]
S. aureus, S. mutansSeeds, roots, leaves (ethanolic, acetate, ethyl-acetate extracts)400 mg/mL[92]
E. coli, S. typhi, S. aureus, Enterococcus sp., P. aeruginosaLeaves (aqueous and methanolic extracts)30 mg/mL[93]
Antifungal effects
Yeasts (Candida species, etc.)
C. albicansLeaves (ethanolic and aqueous extracts)100, 200, 300, 400, 500 µg/mL[96]
C. albicans, C. dulblinesis, C. glabarata, C. kefyr, C. krusei, C. lusitania.Seeds (oil)1.0%[98]
C. albicans, C. parapsilosis, C. krusei, C. tropicalis.Seeds (purified protein, Mo-CBP2)0.32 mg/g[102]
Dermatophytes
T. rubrum, T. mentagrophytes, E. xoccosum, M. canisLeaves
(crude essential oil)
(ethanolic extract)
Seed extract

0.2. 0.4, 0.6, 1.6 mg/mL
2.5 mg/mL
2.5 mg/mL
[97]
E. floccosum and T. rubrumIsothiocyanates isolated from seed extract1, 10 mg/mL[88]
Phytopathogenic fungi
Fusarium solani, F. oxysporum, C. musae and C. gloesporioidesProtein Mo-CBP3 isolated from seeds0.05 mg/mL[100,101]
T. mentagrophytesProtein Mo-CBP4 isolated from seeds5, 10, 20 mg/g[103]

6.4. Antiviral Activity

M. oleifera has demonstrated promising antiviral activity in both in vitro and in vivo studies, attributed to its rich content of bioactive phytochemicals (Table 9). Extracts and isolated compounds from various parts of the plant have shown inhibitory effects against a wide range of viruses, including Hepatitis B virus (HBV), Influenza A virus subtype H1N1 (A/H1N1), Herpes Simplex Virus Type 1 (HSV-1), SARS-CoV-2, Foot and Mouth Disease Virus (FMDV), and Newcastle Disease Virus (NDV).
Aqueous leaf extract inhibited HBV replication in Huh7 liver cells expressing HBV genotypes, demonstrating its antiviral potential at the cellular level [105]. In vivo, oral administration of aqueous extract significantly limited the development of skin lesions in HSV-1-infected mice and enhanced immune response by increasing interferon-gamma (IFN-γ) production [106].
In the context of SARS-CoV-2, M. oleifera has attracted substantial attention due to its phytochemical diversity. Computational studies have identified 22 bioactive compounds with predicted affinity for viral targets. Flavonoids such as rutin, myricetin, quercetin, apigenin, and especially ellagic acid demonstrated strong binding to the spike (S) protein of SARS-CoV-2, suggesting potential to block viral entry [107]. Additionally, kaempferol-3-O-rutinoside and vitexin formed stable complexes with the main protease (Mpro), a critical enzyme in viral replication [108]. Similar inhibitory effects were observed for quercetin-3-rhamnoside, myricetin-3-rutinoside, and rutin [109].
These findings support the hypothesis that M. oleifera extracts may interfere with key stages of viral infection and replication. Moreover, the water-soluble nature of many of these compounds favors intestinal absorption and possibly even passage through the blood-brain barrier—enhancing their potential as orally administered antiviral agents.
Table 9. Antiviral activity of M. oleifera extracts against various viral pathogens.
Table 9. Antiviral activity of M. oleifera extracts against various viral pathogens.
Virus TypePlant Part
(Extract Type)
Dose/EC50/EC90References
Influenza Virus (H1N1)Seeds
(ethanolic)
EC50 = 1.27 µM;
EC90 = 5.30 µM
[105]
Herpes Simplex Virus Type 1 (HSV-1)Leaves300 mg/kg (in vivo)[106]
Hepatitis B Virus (HBV)Leaves
(aqueous)
30, 45, and 60 µg/mL[105]
Foot and Mouth Disease Virus (FMDV)Leaves
(ethanolic)
1.6, 6.12, 25, 50, 100, 200 µg/mL[110]
Newcastle Disease Virus (NDV)Leaves
(methanolic)
200 mg/kg (in vivo)[111]

6.5. Anticancer Effects

M. oleifera exhibits promising anticancer potential due to its rich composition of bioactive compounds that modulate oxidative stress, promote apoptosis, and interfere with tumor-promoting signaling pathways.

6.5.1. In Vitro and Mechanistic Evidence

Numerous in vitro studies have demonstrated the cytotoxic effects of M. oleifera extracts against a variety of cancer cell lines, including cervical (HeLa) [112], leukemia and hepatocarcinoma (HepG2) [113], colon [114], lung adenocarcinoma (A549) [115], breast and colorectal cancer [116].
Bioactive fractions from leaves, seeds, and bark have shown the ability to trigger apoptosis, modulate oxidative balance, and suppress tumor cell proliferation. For instance, ethyl acetate leaf extract rich in phenolics inhibited melanoma cell growth via caspase-dependent and -independent pathways [117], while an alkaloid extract upregulated caspase-3 and -9 in A549 lung cells [118]. In PC3 prostate cancer cells, methanolic leaf extract enhanced caspase-3 activity by downregulating the Hedgehog pathway [119].
M. oleifera compounds such as quercetin, kaempferol, and benzyl isothiocyanate have been shown to induce cell cycle arrest by regulating cyclins and cyclin-dependent kinases, thereby limiting cancer cell division [114,118,120,121].
Moreover, inhibition of key oncogenic signaling cascades—such as NF-κB, PI3K/AKT, JAK2/STAT3, and MAPK—has been reported following treatment with M. oleifera extracts, highlighting their role in suppressing tumor growth and metastasis [116,118,120]
Antioxidant-rich compounds in M. oleifera also contribute to DNA protection by reducing oxidative damage. Phenolic-rich leaf extracts counteracted DNA damage in tumor KB cells [122,123], while aqueous extracts induced oxidative stress and cell cycle arrest (sub-G0 phase) in colon cancer lines, demonstrating a dual role depending on the cellular context [114].
There is also emerging evidence of epigenetic modulation. Moringa isothiocyanate (MIC-1) was shown to reverse aberrant DNA CpG methylation during carcinogenic transformation in JB6 cells, suggesting its potential to influence gene expression patterns linked to tumorigenesis [124].

6.5.2. In Vivo Studies and Cancer Chemopreventive Potential

In vivo models further support M. oleifera’s anticancer potential. A flower-derived trypsin inhibitor (MoFTI) reduced tumor growth and angiogenesis in a murine sarcoma model at doses of 15 or 30 mg/kg [125]. In a colon cancer mouse model, dietary supplementation with boiled M. oleifera pods (1.5–6.0%) over 15 weeks significantly reduced tumor incidence, with 3% being the most effective dose [124].
In a breast cancer model, M. oleifera extract mitigatedoxorubicin-induced toxicity by reducing inflammation and oxidative stress, demonstrating a protective role during chemotherapy [126,127]. However, combination treatment with seed extract and chemotherapy in triple-negative breast cancer unexpectedly led to the upregulation of pro-angiogenic genes, indicating that therapeutic interactions may vary depending on disease context and treatment strategy [127].
To date, no clinical trials have confirmed the anticancer efficacy of M. oleifera in humans, and further translational research is needed to assess its safety, bioavailability, and potential synergistic effects with conventional therapies.

6.6. Hepatoprotective Effects

M. oleifera has demonstrated significant hepatoprotective activity in various animal models of chemically induced liver injury. Experimental studies using toxic agents such as carbon tetrachloride (CCl4), bisphenol-A, cadmium, paracetamol, acetaminophen, and lead consistently show that M. oleifera extracts can prevent or mitigate liver damage through both antioxidant and anti-inflammatory mechanisms.
Administration of M. oleifera leaf extracts led to a marked reduction in serum liver enzymes—alanine transaminase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP)—which are biomarkers of hepatic injury. Histopathological analyses corroborated these findings, revealing improved hepatic architecture and reduced hepatocellular necrosis in treated animals [128,129].
In CCl4-induced liver damage, leaf extract significantly lowered levels of bilirubin, glutamate pyruvate transferase (GPT), glutamate oxaloacetate transferase (GOT), ALP, and lysosomal enzymes, with corresponding improvements in hepatocyte morphology [130]. Similarly, rats exposed to cadmium and treated with M. oleifera (500 mg/kg for 28 days) exhibited normalized liver enzymes and reduced lipid peroxidation [131].
Mechanistically, the hepatoprotective effects of M. oleifera are attributed to its ability to suppress oxidative stress, inhibit pro-inflammatory cytokines (e.g., TNF-α and IL-1β), and modulate the NF-κB signaling pathway [130,131,132,133]. For instance, aqueous leaf extract (100–200 mg/kg) significantly attenuated acetaminophen-induced hepatotoxicity by restoring hepatic glutathione levels and reducing serum transaminases, nitrite, and malondialdehyde (MDA) content [131,134]. In another study, microencapsulated M. oleifera oil reduced hepatic oxidative stress and inflammation in rats fed a high-fat diet. Additionally, the hepatoprotective action of the leaf extract in rats with levofloxacin-induced liver damage was linked to its antioxidant properties, as evidenced by the normalization of liver enzymes (ALT, AST, GGT) and restoration of antioxidant balance [135].

6.7. Antidiabetic Effects

M. oleifera has demonstrated antidiabetic potential in both experimental animal models and human studies. Extracts from its leaves and seeds have been shown to reduce blood glucose levels, improve lipid profiles, and mitigate diabetes-induced tissue damage. These effects are attributed to its rich content of bioactive compounds with antioxidant and anti-inflammatory properties, which modulate key biochemical pathways involved in glucose and lipid metabolism [136,137].
In vitro studies indicate that M. oleifera leaf extracts inhibit carbohydrate-digesting enzymes such as α-glucosidase and pancreatic lipase, thereby reducing glucose absorption and improving insulin sensitivity in adipocyte models (3T3-L1 cells) [138]. The inhibition of α-amylase and α-glucosidase enzymes is a recognized strategy to reduce postprandial hyperglycemia, especially in patients with type 2 diabetes [139].
In vivo, administration of M. oleifera leaf extract (100 or 200 mg/kg orally) for eight weeks in streptozotocin (STZ)-induced diabetic rats significantly reduced blood glucose and oxidative stress markers. It also improved renal histology and downregulated pro-inflammatory markers such as TGF-β1 and collagen IV expression in diabetic nephropathy [140]. Aljazzaf et al. further showed that combined methanolic extracts of leaves and seeds (500 mg/kg) administered for 13 months to alloxan-induced diabetic mice improved antioxidant status, lipid profiles, and histopathological outcomes in liver and kidney tissues [141].
The stem extract also exhibited protective effects on pancreatic β-cells by modulating oxidative stress and cell signaling pathways, reinforcing the therapeutic relevance of non-leaf tissues [142].
Clinical studies support these findings. In postmenopausal women, supplementation with M. oleifera leaf powder over three months led to a reduction in fasting blood glucose levels [143]. A randomized placebo-controlled trial demonstrated that prediabetic individuals taking dry leaf powder capsules exhibited significant decreases in fasting glucose and HbA1c levels compared to controls [144]. In obese individuals with type 2 diabetes, 40-day supplementation with leaf powder reduced serum glucose and LDL cholesterol levels [145].
Together, these results suggest that M. oleifera offers promising therapeutic value in diabetes management, potentially serving as an adjunct to conventional treatments.

6.8. Cardiovascular Effects

The cardioprotective potential of M. oleifera has been increasingly supported by experimental evidence, much of which is linked to its antioxidant, hypoglycemic, and hypolipidemic properties. These effects are attributed to a variety of phytochemicals—including quercetin, apigenin, and lupeol—known to modulate oxidative stress and vascular function.
In vivo studies have shown that supplementation with M. oleifera seed powder exerts antihypertensive effects in spontaneously hypertensive rats, with improvements in blood pressure regulation and cardiac function [146,147]. Similarly, in isoproterenol-induced myocardial injury in Wistar rats, chronic administration of leaf extracts normalized serum biochemical parameters, reduced lipid peroxidation, and mitigated histopathological damage to cardiac tissue [148].
Cardioprotective effects were also observed in a model of potassium bromate-induced cardiac dysfunction, where treatment with aqueous leaf extract restored serum levels of AST, ALT, and ALP, markers of cardiac and hepatic stress [149].
Mechanistic insights suggest that the cardiodepressive effects of M. oleifera may involve the activation of muscarinic M2 receptors, leading to reduced cardiac contractility and enhanced vasodilation, ultimately lowering peripheral resistance and blood pressure [150].
Together, these findings support the potential application of M. oleifera extracts as adjunct agents in managing cardiovascular conditions such as hypertension, myocardial injury, and ischemic heart disease.

6.9. Neuroprotective Effects

Neurodegenerative disorders, including Alzheimer’s disease, Parkinson’s disease, and dementia, are commonly associated with progressive neuronal loss caused by oxidative stress, neuroinflammation, and mitochondrial dysfunction. Emerging evidence from in vitro and in vivo studies suggests that M. oleifera exhibits promising neuroprotective potential against such conditions, as well as acute injuries like stroke and neurotoxic damage [151].
These effects are largely attributed to the high levels of polyphenolic compounds in M. oleifera, which exhibit potent antioxidant activity. In human neuroblastoma SH-SY5Y cells, leaf extract significantly reduced oxidative stress by lowering lipid peroxidation, enhancing endogenous antioxidant enzyme activity, and preserving mitochondrial function, all while maintaining low cytotoxicity [152,153]. In vivo, moringa tree extracts protected neural tissue in models of ischemic stroke and pesticide-induced neurotoxicity [151,152,153,154,155,156].
Seed extracts, rich in glucosinolates such as niazimicin (a thiocarbamate glycoside), have demonstrated protective effects in neurodegenerative models. In rotenone-induced Parkinson’s disease in mice, moringa seed extract reduced motor dysfunction and histologically preserved dopaminergic neurons [157]. Similarly, niazimicin-containing ethanol seed fractions alleviated oxidative damage and behavioral deficits in an aluminum chloride-induced dementia model [158].
In a rat model of hyperhomocysteinemia-induced Alzheimer’s disease, methanolic leaf extract administered at 400 mg/kg/day improved outcomes both as a preventive and curative agent. It reduced oxidative stress, inhibited tau hyperphosphorylation, lowered amyloid-β production, and mitigated neurodegenerative pathology, supporting a multifaceted neuroprotective mechanism involving antioxidative and anti-inflammatory pathways [159].
Beyond biochemical and histological protection, M. oleifera leaf extracts have also been shown to improve cognitive performance. In several models—including age-related dementia, scopolamine-induced amnesia, and maternal protein deficiency moringa supplementation improved learning, memory, and spatial navigation abilities [155,160,161,162].

6.10. Gastrointestinal Protective Effects

M. oleifera demonstrates notable gastroprotective properties. In a rat model of bisphenol A-induced gastric ulcers, oral administration of leaf extract (200 mg/kg for four weeks) significantly reduced ulceration, accompanied by potent antioxidant, anti-apoptotic, and anti-inflammatory effects [163]. Aqueous seed extract (2000 mg/kg for 14 days) also protected against indomethacin-induced gastric ulcers, though its effect was not as strong as that of the reference drug cimetidine [164]. In a mouse model of ulcerative colitis, oral administration of a polysaccharide isolated from M. oleifera alleviated intestinal damage and suppressed the release of pro-inflammatory cytokines [165]. The antibacterial, anti-inflammatory, and ulcer-protective properties of M. oleifera contribute to its anti-diarrheal effect, which has been documented in models of bacterial gastroenteritis [166,167].

6.11. Anti-Obesity Effects

Obesity is a chronic metabolic condition marked by excessive fat accumulation and is closely associated with an increased risk of type 2 diabetes, cardiovascular disease, stroke, and certain cancers. M. oleifera has been extensively investigated for its anti-obesity effects, which are primarily attributed to its rich profile of bioactive compounds with antioxidant, anti-inflammatory, and lipid-lowering properties. These compounds exert their effects by improving lipid metabolism, reducing adiposity, modulating hormone levels, and enhancing the expression of genes involved in fat oxidation.
In high-fat diet (HFD)-induced obese rats, oral administration of M. oleifera methanolic extract (200 and 400 mg/kg for 12 weeks) significantly reduced body weight, total cholesterol, and triglyceride levels, while increasing HDL-C and antioxidant enzyme activity [168]. Similar results were reported in HFD-obese mice, where fermented M. oleifera extract reduced hepatic lipid accumulation and downregulated pro-inflammatory cytokine mRNA expression [169]. Additionally, ethanolic leaf extract (5.6 or 11.2 mg/20 g body weight/day for 7 weeks) prevented hematological disturbances in HFD-fed mice, including improvements in hemoglobin levels and white blood cell profiles [170].
At the molecular level, M. oleifera extracts suppressed the expression of obesity-related hormones such as leptin and resistin, while increasing adiponectin levels—an effect comparable to the lipid-lowering drug Simvastatin [171]. Other studies demonstrated that M. oleifera ethanol extract decreased adiposity index, blood glucose, insulin resistance markers, and downregulated lipogenic enzymes like fatty acid synthase and HMG-CoA reductase. It simultaneously enhanced the expression of PPARα and MC4R, facilitating β-oxidation and reducing fat accumulation in hepatic and adipose tissues [172].
In guinea pigs fed a high-cholesterol diet, dietary inclusion of moringa leaves (2 or 3.5 g/day) significantly reduced hepatic cholesterol and triglyceride levels, inflammatory cytokines (IL-1β, IL-10, IFN-γ), and histologically evident liver steatosis, without altering plasma lipid profiles. These findings suggest modulation of hepatic lipid synthesis pathways as a key mechanism [173].
Human clinical trials, although still limited, support these findings. In a 12-week randomized controlled trial with 40 overweight hyperlipidemic subjects, supplementation with M. oleifera leaf powder resulted in reductions in body weight, BMI, waist circumference, blood pressure, triglycerides, and LDL cholesterol [174]. Similarly, a double-blind, placebo-controlled study in overweight women (400 mg ethanolic extract/day for 8 weeks) showed significant decreases in BMI, total cholesterol, and LDL levels compared to placebo [172].
Together, these data from preclinical and clinical studies indicate that M. oleifera may be a promising natural adjunct for managing obesity and associated metabolic disturbances through multi-targeted mechanisms.

6.12. Effects on Fertility

M. oleifera has been traditionally used in ethnomedicine across parts of Africa and Asia to influence reproductive function. Scientific studies have confirmed that the plant exhibits both fertility-enhancing and antifertility effects, depending on factors such as plant part, dose, extract type, sex of the subject, and physiological context.

6.12.1. Fertility-Enhancing Effects

The antioxidant, anti-inflammatory, and androgen-supporting properties of M. oleifera contribute to improved male reproductive health. These effects are largely attributed to the plant’s bioactive constituents, which protect against oxidative damage, improve testicular architecture, and restore hormonal balance.
In animal models, M. oleifera leaf extract has ameliorated testicular toxicity caused by melamine, improving sperm quality, testicular weight, follicle-stimulating hormone (FSH), and testosterone levels [175]. In rats with experimentally induced cryptorchidism, aqueous extract (400–800 mg/kg, administered orally) reduced germ cell apoptosis, oxidative stress, and heat shock protein 70 expression, preserving testicular integrity [176].
In a high-fructose diet rat model, M. oleifera aqueous leaf extract (300 mg/kg) reversed hepatic insulin resistance, improved serum testosterone levels, and upregulated genes involved in steroidogenesis (e.g., StAR and 3β-HSD), further supporting its reproductive benefits [177].

6.12.2. Antifertility and Abortifacient Effects

Conversely, M. oleifera has demonstrated antifertility effects, particularly in female animal models. Studies have reported abortifacient activity, impaired pregnancy outcomes, and toxic effects on offspring, particularly with seed extracts.
In pregnant Wistar rats, lipid-rich aqueous-methanol seed extracts caused hepatic and renal toxicity in offspring and restricted uterine development in females, while lipid-free extracts at doses below 300 mg/kg showed no adverse reproductive outcomes [178]. An aqueous extract (175 mg/kg) caused complete abortion in pregnant rats, reflecting its ethnomedical use in rural India as an early-stage abortifacient [179]. However, at lower doses (30 mg/kg), the same extract ameliorated postpartum depression symptoms without abortive effects, improving maternal behavior and lactation [180].
Further evidence shows that ethanol extract (250–500 mg/kg) inhibited implantation in decidualized female rats, indicating anti-implantation activity [181]. A dietary study in rabbits revealed sex-dependent hormonal modulation: while male rabbits showed improved fertility, increased FSH and LH, and enhanced semen quality, female rabbits exhibited reduced FSH, LH, and estrogen, with elevated progesterone at higher doses [182].
These findings suggest that M. oleifera modulates reproductive function in a complex, context-dependent manner. While its use may benefit male fertility and protect against reproductive toxins, caution is warranted regarding its use during pregnancy due to its potential abortifacient effects. Further studies are needed to clarify the safe therapeutic window and underlying mechanisms of these divergent effects.

6.13. Effects on Bones

In vivo and in vitro studies have demonstrated the positive effects of M. oleifera leaf extract on bone health, including stimulation of osteoblastic cell proliferation, bone matrix formation and mineralization, and reduction of bone resorption. In rabbit models with critical-sized mandibular bone defects, the application of M. oleifera leaf extract in combination with beta-tricalcium phosphate enhanced bone regeneration. Histological analyses revealed increased new bone formation and a higher number of osteoblasts in the treated groups compared to controls. [14], In the ovariectomized rat model (a postmenopausal osteoporosis model), supplementation with M. oleifera leaf powder significantly increased bone mineral density and improved bone microstructure. The effects were associated with the modulation of gut microbiota and the MAPK signaling pathway, suggesting a multifaceted mechanism of action [183]. A recent study identified bioactive peptides derived from M. oleifera leaves with osteoporosis-modulatory properties. Cellular assays showed that one of these, DPYLGK, significantly inhibits bone resorption and promotes bone formation [184]. Another in vitro study reported a biphasic dose-dependent effect of M. oleifera leaf extract on the growth activity of osteoblast-like SaOS-2 cells, with the lower doses (25 and 50 μg/mL) enhancing bone formation, and 100 and 200 μg/mL doses inhibiting the proliferation of the cells [185].

7. Toxicity Studies

The safety profile of M. oleifera has been widely assessed in both in vitro and in vivo models to evaluate potential cytotoxicity and systemic toxicity associated with various extracts. While most studies indicate that M. oleifera is relatively safe at moderate doses, high concentrations or prolonged use may lead to toxicological effects, depending on the extract type, dose, and biological system used.

7.1. In Vitro Cytotoxicity

Several in vitro studies have examined the cytotoxic effects of M. oleifera extracts on cell lines:
  • Methanolic leaf extract applied to HBF4 cells for 24 h caused noticeable changes at doses ≥1000 μg/mL, including increased cell size. The concentration affecting viability was ≥700 μg/mL [186].
  • Aqueous leaf extract showed cytotoxic effects on A549 lung cancer cells, with 30% and 15% cell death at 400 μg/mL and 500 μg/mL, respectively [187].
  • Essential oil from seeds tested on HeLa, HepG2, MCF-7, Caco-2, and L929 cell lines (0.15–1 mg/mL) showed the highest cytotoxicity in HeLa, HepG2, and MCF-7 cells, suggesting a dose- and cell type-specific toxicity [170].

7.2. In Vivo Toxicity in Animal Models

Zhang et al. administered aqueous-methanol leaf extract (2000 mg/kg) to Wistar rats. Blood markers (ALT, AST, total bilirubin) did not indicate acute liver damage, suggesting the lethal dose (LD50) exceeds 2000 mg/kg in females [188].
Adedapo et al. found that aqueous leaf extract (1600 mg/100 g) lowered serum albumin levels in rats, indicating potential hepatic stress [189]. Subacute toxicity tests with doses of 250, 500, and 1500 mg/kg showed an LD50 value of approximately 1585 mg/kg [190]. Asiedu-Gyekye et al. tested doses up to 5000 mg/kg and advised limiting daily intake to under 70 g of the extract to avoid the potential accumulation of toxic macro- and microelements [191].
Ajibade et al. reported no mortality at 3000 mg/100 g body weight, but acute toxicity occurred at 4000 mg/kg, with deaths recorded at 5000 mg/kg [192].

7.3. Safety Considerations for Human Use

Despite high-dose toxicity in animal models, M. oleifera extracts (particularly fruits and leaves) have shown favorable safety profiles in mutagenicity and genotoxicity assessments. Siddiqui et al. reported that fruit extracts were non-mutagenic and safe for potential use in anticancer formulations based on toxicity assays in liver cancer cells [193].

8. Moringa oleifera and Its Application in Dietary Supplements

8.1. Commercial Use in Dietary Supplements

Due to its rich profile of bioactive compounds, including flavonoids, phenolic acids, isothiocyanates, vitamins, and minerals, M. oleifera has become increasingly popular in the formulation of dietary supplements (Table 10). These products are marketed globally for their diverse health-promoting properties, ranging from antioxidant and anti-inflammatory effects to blood sugar regulation and immune system support. Supplements typically utilize leaf extracts, but preparations from seeds, bark, root, and fruit are also available, each offering specific therapeutic benefits depending on the extract type and composition.

8.2. Applications in Functional Foods and Nutritional Enrichment

Moringa oleifera is widely utilized in the development of functional foods due to its exceptional nutritional value. In African and Asian cuisines, various parts of the plant are consumed: the pods and seeds are cooked in stews, the leaves are brewed as tea or added to soups, and the roots are used as a spice with a horseradish-like flavor.
In Europe, particularly in Bulgaria, powdered M. oleifera leaves are becoming increasingly popular as a dietary supplement. They are incorporated into smoothies, juices, and cooked dishes to enhance micronutrient intake and support immune function. The dried leaf powder is also used for its content of essential fatty acids and bioactive polyphenols.
Experimental applications in food technology have demonstrated that incorporating M. oleifera into staple foods can enhance their nutritional quality. For example, adding moringa seed powder to wheat bread increases its protein, carbohydrate, and vitamin A content, while also extending shelf life by reducing fungal contamination [26,194]. However, while moringa enrichment improves the fiber and mineral content of baked goods such as biscuits and muffins, it may also affect organoleptic properties, altering taste, color, texture, and vitamin retention [30,195,196].

9. Cosmetic Applications

M. oleifera is increasingly used in cosmetic formulations due to its rich profile of bioactive compounds with antioxidant, anti-inflammatory, and anti-aging properties. The plant’s seed oil is particularly valued for its high content of essential fatty acids, including oleic, linoleic, and palmitic acids, which help nourish and hydrate the skin. Leaf extracts are also incorporated for their antioxidant polyphenols and vitamins (A, C, and E), which protect the skin from oxidative damage and environmental stressors.
Isothiocyanates derived from M. oleifera (MITC) have demonstrated potential in anti-aging applications. When encapsulated in nanoliposomes, MITC showed increased skin permeability (up to 71.4%), enhancing dermal absorption and photoprotection [197]. Kumalaningsih and colleagues developed a UV-protective whitening cream combining seed oil, leaf extract, and red rice-derived zinc oxide (ZnO), offering both antioxidant protection and UV-blocking activity [198].
M. oleifera extracts and oils from seeds, leaves, and other parts have been approved by the European Commission’s COSING database (2020) for use in cosmetic products. These include facial cleansers, micellar water, moisturizers, body lotions, eye creams, and anti-aging serums manufactured globally in Europe, North America, and Asia (Table 11).

10. Preparation of Moringa Formulations

M. oleifera formulations include granules, tablets, capsules, topical applications, and plant extracts incorporated into nanoparticles. Tablets commonly utilize binding agents such as corn starch, gelatin, and microcrystalline cellulose. Gelatin is the most suitable binding agent for the tablets, providing the lowest brittleness, appropriate strength, and easy disintegration.
Many components in plant extracts have poor solubility and low bioavailability. To address these challenges, the literature includes various modern formulations. To improve the water solubility of isothiocyanates, modified amphiphilic hyaluronic acid and ceramide nanoliposomes have been used to enhance the permeation ability of the substance through the skin [205]. Studies have also reported coating gelatin nanoparticles with M. oleifera extracts to enhance repellent activity [170]. Silver nanoparticles combined with leaf extract have shown anticancer activity against HTC116 and SW480 cancer cells [206]. For external applications intended to promote rapid wound healing, a hydrogel containing a hexane extract from the plant’s seeds has been recommended. Research indicates that tablets formulated with M. oleifera gum facilitate 91% drug release, making them a promising carrier for colon-targeted drug delivery [207].

11. Existing Patents on Compositions with Extracts from Different Parts of M. oleifera

In 2009, Jiaheng Zhang patented a formulation derived from the leaves and seeds of M. oleifera. The invention claims that it can be used as an adjunct therapy for treating fever, bronchitis, eye and ear infections, scurvy, skin infections, and arthritis, as well as for lowering blood pressure and blood sugar. Additionally, it is said to help alleviate fatigue and anxiety and improve sleep [208]. In 2004, Ranjitsinh Solanki patented a composition of 11 herbs intended for cancer treatment, with M. oleifera included at a concentration of 8–12%. The patented formula was effective against squamous cell carcinoma and lung cancer cells [209].
In 2005, Suman Preet Singh Khanuja patented a glycoside isolated from the plant’s pods, called niaziridine. The invention enhances the activity of antibiotics, such as rifampicin, tetracycline, and ampicillin. The patent claims that this biomolecule increases the absorption of certain drugs and vitamins through gastrointestinal membranes, thereby improving their bioavailability [210]. In 2011, Richard Gomez patented a dietary supplement containing M. oleifera, Curcuma longa, and piperine, which has been shown to aid in weight reduction [209]. Anti-adipogenic and lipolytic compositions have also been developed, containing extracts from Curcuma longa (10%) and M. oleifera (30–70%). It has been demonstrated that the supplement can combat obesity. A patent was published in 2013 [210].

12. Interactions Between M. oleifera and Pharmaceutical Drugs

12.1. Synergistic Effects

Due to resistance to chemotherapy drugs, cancer cell recurrence is often observed. It is known that some plant extracts have antitumor and immunoregulatory effects with relatively low toxicity [4]. They show that the chemotherapy drug doxorubicin, combined with M. oleifera leaf extracts, has a synergistic effect in inhibiting the growth of HeLa cells. Compounds with biological activity in the methanol extract from the bark can enhance the action of antibiotics, helping to overcome resistance in multidrug-resistant organisms. Vankwani, S. et al. demonstrated a significant synergistic effect between M. oleifera stem bark extract and the antibiotic ampicillin against methicillin-resistant S. aureus [209]. Abu-Hussien et al. reported that the combination of essential oil from M. oleifera seeds with essential oils from cinnamon (Cinnamomum verum) and black cumin (Nigella sativa), significantly increased antibacterial activity against Staphylococcus aureus, and the inhibitory concentration values for the combination were lower compared to the individual oils [210]. In an animal model, the combination of M. oleifera extract with fluoxetine, a selective serotonin reuptake inhibitor, produced enhanced antidepressant-like effects compared to either treatment alone. This suggests a potential synergistic interaction, although further research is needed to confirm these findings in humans [211].

12.2. Pharmacokinetic Drug Interactions

  • Antimalarial drugs. An experimental study demonstrated that co-administration of M. oleifera leaf extract with chloroquine resulted in antagonistic effects, potentially due to the inhibition of chloroquine absorption. This interaction may reduce the drug efficacy in treating malaria, highlighting the need for caution when combining Moringa with certain antimalarial drugs [212].
  • Cytochrome P450 enzyme inhibition. In vitro studies have shown that M. oleifera extracts can inhibit cytochrome P450 enzymes, particularly CYP3A4 and CYP2D6, which are involved in the metabolism of many drugs. The degree of inhibition varied depending on the extract type and concentration, with methanolic leaf extracts showing more potent effects [213].
  • Antihypertensive drugs. Compared to standard antihypertensive drugs, M. oleifera leaf extract administered alone to spontaneously hypertensive rats reduced blood pressure. However, when combined with these drugs, no synergistic effects were observed. This suggests that the concurrent use of Moringa with antihypertensive medications may not enhance therapeutic outcomes [214].

13. Conclusions

M. oleifera is a plant of exceptional nutritional and pharmacological significance, known for its rich profile of vitamins, minerals, amino acids, and bioactive secondary metabolites. Extensive in vitro and in vivo studies have demonstrated its antioxidant, anti-inflammatory, antimicrobial, antidiabetic, hepatoprotective, neuroprotective, and anticancer properties, supporting its integration into food, medicinal, and cosmetic formulations. Its utility as a functional food and dietary supplement is further enhanced by its safety profile and wide-ranging biological activities. Moreover, M. oleifera is a fast-growing, drought-resistant species that thrives in poor soils, making it an ecologically sustainable crop suitable for cultivation in arid and semi-arid regions. Its resilience and low agricultural input requirements make it especially valuable for improving food security and supporting local economies in resource-limited settings. Future research should focus on standardizing extraction methods, elucidating underlying molecular mechanisms, and conducting well-designed clinical trials to validate efficacy and safety. Integrating M. oleifera into public health strategies and evidence-based therapeutics holds great promise for addressing nutritional deficiencies and managing chronic diseases on a global scale.

Author Contributions

A.G., K.N. and N.P. (Natalina Panova)—designed and conceived the project. M.V., L.M., D.B. and N.P. —initial draft preparation, A.T. and A.B.—visualization and data curation, K.N., project administration, K.N., D.B., A.G., N.P. (Nadezhda Petkova), G.G. and S.N.—writing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study is financed by the European Union-Next Generation EU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, the project No. BG-RRP-2.004-0009-C02 Special thanks to the Medical University-Varna and European Union for the financial support provided for the publication of the paper.

Data Availability Statement

Datasets from the time of this study are available from the respective authors upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ROSreactive oxygen species
GSHreduced glutathione
GSSGoxidized glutathione
ALTalanine aminotransferase
ASTaspartate aminotransferase
GGTgamma-glutamyl transferase
COXcyclooxygenase
BSSβ-sitosterol
MOMoringa oleifera
LPSlipopolysaccharide
DNADeoxyribonucleic Acid
iNOSnitric oxide synthase
MOLM. oleifera leaf
Mo-CBPMoringa oleifera-Chitin-binding proteins
NDVNewcastle disease virus
HSV-1Herpes Simplex Virus Type 1
ALPalkaline phosphatase
TNFtumor necrosis factor
p.osorally
STZstreptozotocin
LDLLow-density lipoprotein
HFDfollicle-stimulating hormone
mRNAMessenger Ribonucleic acid

References

  1. Fuglie, L.J. Producing Food Without Pesticides: Local Solutions to Crop Pest Control in West Africa; Church World Service: Dakar, Senegal, 1998; pp. 1–158. [Google Scholar]
  2. Pareek, A.; Pant, M.; Gupta, M.M.; Kashania, P.; Ratan, Y.; Jain, V.; Pareek, A.; Chuturgoon, A.A. Moringa oleifera: An Updated Comprehensive Review of Its Pharmacological Activities, Ethnomedicinal, Phytopharmaceutical Formulation, Clinical, Phytochemical, and Toxicological Aspects. Int. J. Mol. Sci. 2023, 24, 2098. [Google Scholar] [CrossRef] [PubMed]
  3. Gharsallah, K.; Rezig, L.; Rajoka, M.S.R.; Mehwish, H.M.; Ali, M.A.; Chew, S.C. Moringa oleifera: Processing, Phytochemical Composition, and Industrial Applications. S. Afr. J. Bot. 2023, 160, 180–193. [Google Scholar] [CrossRef]
  4. Liu, R.; Liu, J.; Huang, Q.; Liu, S.; Jiang, Y. Moringa oleifera: A Systematic Review of Its Botany, Traditional Uses, Phytochemistry, Pharmacology and Toxicity. J. Pharm. Pharmacol. 2022, 74, 296–320. [Google Scholar] [CrossRef] [PubMed]
  5. Camilleri, E.; Blundell, R. A Comprehensive Review of the Phytochemicals, Health Benefits, Pharmacological Safety and Medicinal Prospects of Moringa oleifera. Heliyon 2024, 10, e27807. [Google Scholar] [CrossRef]
  6. Mali, S.; Bendre, S.; Patil, S. Overview of Pharmacognostical and Pharmacological Properties of Moringa oleifera. Asian J. Pharm. Technol. 2022, 12, 77–83. [Google Scholar] [CrossRef]
  7. Kou, X.; Li, B.; Olayanju, J.B.; Drake, J.M.; Chen, N. Nutraceutical or Pharmacological Potential of Moringa oleifera Lam. Nutrients 2018, 10, 343. [Google Scholar] [CrossRef]
  8. Malage, A.; Jadhav, S.; Yogekar, T.; Sharma, S. Phytochemical and Pharmacological Characteristics of Moringa oleifera: A Review. Asian J. Pharm. Clin. Res. 2020, 13, 5–8. [Google Scholar] [CrossRef]
  9. Anwar, F.; Ashraf, M.; Bhanger, M.I. Interprovenance Variation in the Composition of Moringa oleifera Oilseeds from Pakistan. J. Am. Oil Chem. Soc. 2005, 82, 45–51. [Google Scholar] [CrossRef]
  10. Berg, J.; Kuipers, S. The Antibacterial Action of Moringa oleifera: A Systematic Review. S. Afr. J. Bot. 2022, 151, 224–233. [Google Scholar] [CrossRef]
  11. Chiș, A.; Noubissi, P.A.; Pop, O.-L.; Mureșan, C.I.; Tagne, M.A.F.; Kamgang, R.; Fodor, A.; Sitar-Tăut, A.-V.; Cozma, A.; Orășan, O.H.; et al. Bioactive Compounds in Moringa oleifera: Mechanisms of Action, Focus on Their Anti-Inflammatory Properties. Plants 2024, 13, 20. [Google Scholar] [CrossRef]
  12. Klimek-Szczykutowicz, M.; Gaweł-Bęben, K.; Rutka, A.; Blicharska, E.; Tatarczak-Michalewska, M.; Kulik-Siarek, K.; Kukula-Koch, W.; Malinowska, M.A.; Szopa, A. Moringa oleifera (Drumstick Tree)—Nutraceutical, Cosmetological and Medicinal Importance: A Review. Front. Pharmacol. 2024, 15, 1288382. [Google Scholar] [CrossRef] [PubMed]
  13. Abdelwanis, F.M.; Abdelaty, H.S.; Saleh, S.A. Exploring the Multifaceted Uses of Moringa oleifera: Nutritional, Industrial and Agricultural Innovations in Egypt. Discov. Food 2024, 4, 146. [Google Scholar] [CrossRef]
  14. Elsadek, N.A.; Aboukhadr, M.A.; Kamel, F.R.; Mostafa, H.M.; El-Kimary, G.I. Moringa oleifera Leaf Extract Promotes the Healing of Critical Sized Bone Defects in the Mandibles of Rabbits. BDJ Open 2024, 10, 22. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, J.; Gautam, D.N.S.; Sourav, S.; Sharma, R. Role of Moringa oleifera Lam. in Cancer: Phytochemistry and Pharmacological Insights. Food Front. 2023, 4, 164–206. [Google Scholar] [CrossRef]
  16. Anzano, A.; Ammar, M.; Papaianni, M.; Grauso, L.; Sabbah, M.; Capparelli, R.; Lanzotti, V. Moringa oleifera Lam.: A Phytochemical and Pharmacological Overview. Horticulturae 2021, 7, 409. [Google Scholar] [CrossRef]
  17. Olson, M.E.; Fahey, J.W. Moringa oleifera: Un árbol multiusos para las zonas tropicales secas. Rev. Mex. Biodivers. 2011, 82, 1071–1082. [Google Scholar] [CrossRef]
  18. Albrecht, J. Tree Seed Handbook of Kenya; GTZ Forestry Seed Center: Nairobi, Kenya, 1993. [Google Scholar]
  19. Ambasta, S.P. The Useful Plants of India; Publications & Information Directorate, CSIR: New Delhi, India, 1986. [Google Scholar]
  20. Nadkarni, K.M. Indian Materia Medica, 3rd ed.; Popular Prakashan: Bombay, India, 2000; p. 811. [Google Scholar]
  21. Paikra, B.K.; Dhongade, H.K.J.; Gidwani, B. Phytochemistry and Pharmacology of Moringa oleifera Lam. J. Pharmacopunct. 2017, 20, 194–200. [Google Scholar]
  22. Mallenakuppe, R.; Homabalegowda, H.; Gouri, M.D.; Basavaraju, P.S.; Chandrashekharaiah, U.B. History, Taxonomy and Propagation of Moringa oleifera: A Review. Int. J. Life Sci. 2019, 5, 2322–2327. [Google Scholar] [CrossRef]
  23. Velázquez-Zavala, M.; Peón-Escalante, I.E.; Zepeda-Bautista, R.; Jiménez-Arellanes, M.A. Moringa oleifera Lam.: Potential Uses in Agriculture, Industry and Medicine. Rev. Chapingo Ser. Hortic. 2016, 22, 95–116. [Google Scholar] [CrossRef]
  24. Leone, A.; Spada, A.; Battezzati, A.; Schiraldi, A.; Aristil, J.; Bertoli, S. Cultivation, Genetic, Ethnopharmacology, Phytochemistry and Pharmacology of Moringa oleifera Leaves: An Overview. Int. J. Mol. Sci. 2015, 16, 12791–12835. [Google Scholar] [CrossRef]
  25. Swatia, A.; Virk, A.K.; Kumari, C.; Ali, A.; Garg, P.; Thakur, P.; Attri, C.; Ulshrestha, S. Moringa oleifera—A Never Die Tree: An Overview. Asian J. Pharm. Clin. Res. 2018, 11, 53. [Google Scholar] [CrossRef]
  26. Abdoun, K.; Alsagan, A.; Altahir, O.; Suliman, G.; Al-Haidary, A.; Alsaiady, M. Cultivation and Uses of Moringa oleifera as Non-Conventional Feedstuff in Livestock Production: A Review. Life 2023, 13, 63. [Google Scholar] [CrossRef]
  27. Mahmood, K.T.; Mugal, T.; Haq, I.U. Moringa oleifera: A Natural Gift—A Review. J. Pharm. Sci. Res. 2010, 2, 775–781. [Google Scholar]
  28. Eke, M.O.; Elechi, J.O.G.; Bello, F. Effect of Fortification of Defatted Moringa oleifera Seed Flour on Consumers’ Acceptability and Nutritional Characteristics of Wheat Bread. Eur. Food Sci. Eng. 2022, 3, 18–25. [Google Scholar] [CrossRef]
  29. Patil, S.V.; Mohite, B.V.; Marathe, K.R.; Salunkhe, N.S.; Marathe, V.; Patil, V.S. Moringa Tree, Gift of Nature: A Review on Nutritional and Industrial Potential. Curr. Pharmacol. Rep. 2022, 8, 262–280. [Google Scholar] [CrossRef]
  30. Milla, P.G.; Peñalver, R.; Nieto, G. Health Benefits of Uses and Applications of Moringa oleifera in Bakery Products. Plants 2021, 10, 318. [Google Scholar] [CrossRef]
  31. Olaofe, O.; Adeyeye, E.I.; Ojugbo, S. Comparative Study of Proximate, Amino Acids and Fatty Acids of Moringa oleifera Tree. Elixir Appl. Chem. 2013, 54, 12543–12554. [Google Scholar]
  32. Hedhili, A.; Lubbers, S.; Bou-Maroun, E.; Griffon, F.; Akinyemi, B.E.; Husson, F.; Valentin, D. Moringa oleifera-Supplemented Biscuits: Nutritional Values and Consumer Segmentation. S. Afr. J. Bot. 2021, 138, 406–414. [Google Scholar] [CrossRef]
  33. Monteiro, J.; Scotti-Campos, P.; Pais, I.; Figueiredo, A.C.; Viegas, D.; Reboredo, F. Elemental Composition, Total Fatty Acids, Soluble Sugar Content and Essential Oils of Flowers and Leaves of Moringa oleifera Cultivated in Southern Portugal. Heliyon 2022, 8, e12647. [Google Scholar] [CrossRef]
  34. Trigo, C.; Castelló, M.L.; Ortolá, M.D.; García-Mares, F.J.; Soriano, M.D. Moringa oleifera: An Unknown Crop in Developed Countries with Great Potential for Industry and Adapted to Climate Change. Foods 2020, 10, 31. [Google Scholar] [CrossRef]
  35. Tafesse, A.; Goshu, D.; Gelaw, F.; Ademe, A. Food and Nutrition Security Impacts of Moringa: Evidence from Southern Ethiopia. Cogent Food Agric. 2020, 6, 1733330. [Google Scholar] [CrossRef]
  36. Fuglie, L.J. The Miracle Tree: Moringa oleifera: Natural Nutrition for the Tropics; Church World Service: Dakar, Senegal, 1999; p. 172. [Google Scholar]
  37. Kashyap, P.; Kumar, S.; Riar, C.S.; Jindal, N.; Baniwal, P.; Guiné, R.P.F.; Correia, P.M.R.; Mehra, R.; Kumar, H. Recent Advances in Drumstick (Moringa oleifera) Leaves Bioactive Compounds: Composition, Health Benefits, Bioaccessibility, and Dietary Applications. Antioxidants 2022, 11, 402. [Google Scholar] [CrossRef] [PubMed]
  38. Ceci, R.; Maldini, M.; La Rosa, P.; Sireno, L.; Antinozzi, C.; Olson, M.E.; Dimauro, I.; Duranti, G. The Effect of Moringa oleifera Leaf Extract on C2C12 Myoblast Proliferation and Redox Status Under Oxidative Insult. Antioxidants 2024, 13, 1460. [Google Scholar] [CrossRef] [PubMed]
  39. Pluhackova, H.; Weichtova, L.; Lojkova, L.; Nemec, P.; Svoboda, Z.; Bosko, R.; Pernica, M.; Benesova, K. The Content of Bioactive Compounds in Moringa oleifera and Moringa stenopetala Species Grown in Ethiopia. S. Afr. J. Bot. 2023, 155, 355–360. [Google Scholar] [CrossRef]
  40. Tsaknis, J.; Lalas, S.; Gergis, V.; Dourtoglou, V.; Spiliotis, V. Characterization of Moringa oleifera Variety Mbololo Seed Oil of Kenya. J. Agric. Food Chem. 1999, 47, 4495–4499. [Google Scholar] [CrossRef]
  41. Lalas, S.; Tsaknis, J. Characterization of Moringa oleifera Seed Oil Variety “Periyakulam 1”. J. Food Compos. Anal. 2002, 15, 65–77. [Google Scholar] [CrossRef]
  42. Anwar, F.; Bhanger, M.I. Analytical Characterization of Moringa oleifera Seed Oil Grown in Temperate Regions of Pakistan. J. Agric. Food Chem. 2003, 51, 6558–6563. [Google Scholar] [CrossRef]
  43. Salama, M.A.; Owon, M.A.; Osman, M.F.; Esmail, A.I.; Matthäus, B. Characterization of Egyptian Moringa oleifera Lipids (Whole Seeds and Kernels). J. Food Dairy Sci. Mansoura Univ. 2018, 9, 379–384. [Google Scholar] [CrossRef]
  44. Ozcan, M.M.; Ghafoor, K.; Al Juhaimi, F.; Ahmed, I.A.M.; Babiker, E.E. Effect of Cold-Press and Soxhlet Extraction on Fatty Acids, Tocopherols and Sterol Contents of the Moringa Seed Oils. S. Afr. J. Bot. 2019, 124, 333–337. [Google Scholar] [CrossRef]
  45. Dhakar, R.C.; Maurya, S.D.; Pooniya, B.K.; Bairwa, N.; Gupta, M.; Sanwarmal. Moringa: The Herbal Gold to Combat Malnutrition. Chron. Young Sci. 2011, 2, 119–125. [Google Scholar] [CrossRef]
  46. Gopalan, C.; Sastri, B.R.; Balasubramanian, S. Nutritive Value of Indian Foods; National Institute of Nutrition, Indian Council of Medical Research: Hyderabad, India, 1989. [Google Scholar]
  47. Islam, Z.; Islam, S.M.R.; Hossen, F.; Mahtab-Ul-Islam, K.; Hasan, M.R.; Karim, R. Moringa oleifera Is a Prominent Source of Nutrients with Potential Health Benefits. Int. J. Food Sci. 2021, 2021, 6627265. [Google Scholar] [CrossRef] [PubMed]
  48. Dai, J.; Tao, L.; Shi, C.; Yang, S.; Li, D.; Sheng, J.; Tian, Y. Fermentation Improves Calcium Bioavailability in Moringa oleifera Leaves and Prevents Bone Loss in Calcium-Deficient Rats. Food Sci. Nutr. 2020, 8, 3692–3703. [Google Scholar] [CrossRef] [PubMed]
  49. Sivapragasam, G.; Devi, T.G.; Ragavan, N.D.; Suryadevara, N.; Sridewi, N.; Al Obaid, S.; Alharbi, S.A.; Arulselvan, P. A Comparative Phytochemical Characterization of Moringa oleifera Plant Parts by Different Solvent Extraction. Indian J. Pharm. Educ. Res. 2024, 58, 648–659. [Google Scholar] [CrossRef]
  50. Lu, Y.; Zhang, M.; Meng, X.; Wan, H.; Zhang, J.; Tian, J.; Hao, S.; Jin, K.; Yao, Y. Photoperiod and Shading Regulate Coloration and Anthocyanin Accumulation in the Leaves of Malus Crabapples. Plant Cell Tiss. Organ Cult. 2015, 121, 619–632. [Google Scholar] [CrossRef]
  51. Moyo, B.; Masika, P.J.; Hugo, A.; Muchenje, V. Nutritional Characterization of Moringa oleifera Lam. Leaves. Afr. J. Biotechnol. 2011, 10, 12925–12933. [Google Scholar] [CrossRef]
  52. Wang, Y.; Gao, Y.; Ding, H.; Liu, S.; Han, X.; Gui, J.; Liu, D. Subcritical Ethanol Extraction of Flavonoids from Moringa oleifera Leaf and Evaluation of Antioxidant Activity. Food Chem. 2017, 218, 152–158. [Google Scholar] [CrossRef]
  53. Ijarotimi, O.S.; Adeoti, O.A.; Ariyo, O. Comparative Study on Nutrient Composition, Phytochemical, and Functional Characteristics of Raw, Germinated, and Fermented Moringa oleifera Seed Flour. Food Sci. Nutr. 2013, 1, 452–463. [Google Scholar] [CrossRef]
  54. Vergara-Jimenez, M.; Almatrafi, M.M.; Fernandez, M.L. Bioactive Components in Moringa oleifera Leaves Protect Against Chronic Disease. Antioxidants 2017, 6, 91. [Google Scholar] [CrossRef]
  55. Hassan, M.A.; Xu, T.; Tian, Y.; Zhong, Y.; Ali, F.A.Z.; Yang, X.; Lu, B. Health Benefits and Phenolic Compounds of Moringa oleifera Leaves: A Comprehensive Review. Phytomedicine 2021, 93, 153771. [Google Scholar] [CrossRef]
  56. Xu, Y.-B.; Chen, G.-L.; Guo, M.-Q. Antioxidant and Anti-Inflammatory Activities of the Crude Extracts of Moringa oleifera from Kenya and Their Correlations with Flavonoids. Antioxidants 2019, 8, 296. [Google Scholar] [CrossRef]
  57. Pakade, V.; Cukrowska, E.; Chimuka, L. Comparison of Antioxidant Activity of Moringa oleifera and Selected Vegetables in South Africa. S. Afr. J. Sci. 2013, 109, 5. [Google Scholar] [CrossRef]
  58. Gómez-Martínez, M.; Ascacio-Valdés, J.A.; Flores-Gallegos, A.C.; González-Domínguez, J.; Gómez-Martínez, S.; Aguilar, C.N.; Morlett-Chávez, J.A.; Rodríguez-Herrera, R. Location and Tissue Effects on Phytochemical Composition and in Vitro Antioxidant Activity of Moringa oleifera. Ind. Crops Prod. 2020, 151, 112439. [Google Scholar] [CrossRef]
  59. Elangovan, M.; Dhanarajan, M.S.; Rajalakshmi, A.; Jayachitra, A.; Mathi, P.; Bhogireddy, N. Analysis of Phytochemicals, Antibacterial and Antioxidant Activities of Moringa oleifera Lam. Leaf Extract—An In Vitro Study. Int. J. Drug Dev. Res. 2014, 6, 173–180. [Google Scholar]
  60. Herman-Lara, E.; Rodríguez-Miranda, J.; Ávila-Manrique, S.; Dorado-López, C.; Villalva, M.; Jaime, L.; Santoyo, S.; Martínez-Sánchez, C.E. In Vitro Antioxidant, Anti-Inflammatory Activity and Bioaccessibility of Ethanolic Extracts from Mexican Moringa oleifera Leaf. Foods 2024, 13, 2709. [Google Scholar] [CrossRef]
  61. Shih, M.-C.; Chang, C.-M.; Kang, S.-M.; Tsai, M.-L. Effect of Different Parts (Leaf, Stem and Stalk) and Seasons (Summer and Winter) on the Chemical Compositions and Antioxidant Activity of Moringa oleifera. Int. J. Mol. Sci. 2011, 12, 6077–6088. [Google Scholar] [CrossRef]
  62. Karagiorgou, I.; Grigorakis, S.; Lalas, S.; Makris, D. Polyphenolic Burden and In Vitro Antioxidant Properties of Moringa oleifera Root Extracts. J. Herbmed Pharmacol. 2015, 5, 33–38. [Google Scholar]
  63. Luqman, S.; Srivastava, S.; Kumar, R.; Maurya, A.K.; Chanda, D. Experimental Assessment of Moringa oleifera Leaf and Fruit for Its Antistress, Antioxidant, and Scavenging Potential Using In Vitro and In Vivo Assays. Evid.-Based Complement. Altern. Med. 2012, 2012, 519084. [Google Scholar] [CrossRef]
  64. Unuigbe, C.A.; Okeri, H.A.; Erharuyi, O.; Oghenero, E.E.; Obamedo, D.A. Phytochemical and Antioxidant Evaluation of Moringa oleifera (Moringaceae) Leaf and Seed. J. Pharm. Bioall. Resour. 2014, 11, 51–57. [Google Scholar] [CrossRef]
  65. Peñalver, R.; Martínez-Zamora, L.; Lorenzo, J.M.; Ros, G.; Nieto, G. Nutritional and Antioxidant Properties of Moringa oleifera Leaves in Functional Foods. Foods 2022, 11, 1107. [Google Scholar] [CrossRef]
  66. Sul, O.J.; Ra, S.W. Quercetin Prevents LPS-Induced Oxidative Stress and Inflammation by Modulating NOX2/ROS/NF-κB in Lung Epithelial Cells. Molecules 2021, 26, 6949. [Google Scholar] [CrossRef]
  67. Boudjema, K.; Chouala, K.; Khelef, Y.; Chenna, H.; Badraoui, R.; Boumendjel, M.; Boumendjel, A.; Messarah, M. Antioxidant Effects of Moringa oleifera Against Abamectin-Induced Oxidative Stress in the Brain and Erythrocytes of Rats. Chem. Biodivers. 2024, 22, e202402709. [Google Scholar] [CrossRef] [PubMed]
  68. Liao, P.C.; Lai, M.H.; Hsu, K.P.; Kuo, Y.H.; Chen, J.; Tsai, M.-C.; Li, C.-X.; Yin, X.J.; Jeyashoke, N.; Chao, L.K.P. Identification of β-Sitosterol as in Vitro Anti-Inflammatory Constituent in Moringa oleifera. J. Agric. Food Chem. 2018, 66, 10748–10759. [Google Scholar] [CrossRef] [PubMed]
  69. Tan, W.S.; Arulselvan, P.; Karthivashan, G.; Fakurazi, S. Moringa oleifera Flower Extract Suppresses the Activation of Inflammatory Mediators in Lipopolysaccharide-Stimulated RAW 264.7 Macrophages via NF-κB Pathway. Mediat. Inflamm. 2015, 2015, 720171. [Google Scholar] [CrossRef] [PubMed]
  70. Sailaja, B.S.; Hassan, S.; Cohen, E.; Tmenova, I.; Farias-Pereira, R.; Verzi, M.P.; Raskin, I. Moringa Isothiocyanate-1 Inhibits LPS-Induced Inflammation in Mouse Myoblasts and Skeletal Muscle. PLoS ONE 2022, 17, e0279370. [Google Scholar] [CrossRef]
  71. Miękus, N.; Marszałek, K.; Podlacha, M.; Iqbal, A.; Puchalski, C.; Świergiel, A.H. Health Benefits of Plant-Derived Sulfur Compounds, Glucosinolates, and Organosulfur Compounds. Molecules 2020, 25, 3804. [Google Scholar] [CrossRef]
  72. Cheenpracha, S.; Park, E.-J.; Yoshida, W.Y.; Barit, C.; Wall, M.; Pezzuto, J.M.; Chang, L.C. Potential Anti-Inflammatory Phenolic Glycosides from the Medicinal Plant Moringa oleifera Fruits. Bioorg. Med. Chem. 2010, 18, 6598–6602. [Google Scholar] [CrossRef]
  73. Cuellar-Núñez, M.L.; Loarca-Piña, G.; Berhow, M.; Gonzalez De Mejia, E. Glucosinolate-Rich Hydrolyzed Extract from Moringa oleifera Leaves Decreased the Production of TNF-α and IL-1β Cytokines and Induced ROS and Apoptosis in Human Colon Cancer Cells. J. Funct. Foods 2020, 75, 104270. [Google Scholar] [CrossRef]
  74. Wolff, K.; Robinson, K.; Suh, N.; Michniak-Kohn, B.; Goedken, M.; Polunas, M.; Raskin, I. Isothiocyanate-Rich Moringa Seed Extract Reduces Skin Inflammation in Mouse Ear Edema Model. Phytomed. Plus 2023, 3, 100479. [Google Scholar] [CrossRef]
  75. Ezeamuzie, I.C.; Ambakederemo, A.W.; Shode, F.O.; Ekwebelem, S.C. Antiinflammatory Effects of Moringa oleifera Root Extract. Int. J. Pharmacogn. 1996, 34, 207–212. [Google Scholar] [CrossRef]
  76. Rakesh, S.; Singh, V.J. Anti-Inflammatory Activity of Moringa oleifera Leaf and Pod Extracts against Carrageenan Induced Paw Edema in Albino Mice. J. Pharm. Sci. Innov. 2011, 1, 22–24. [Google Scholar]
  77. Oguntibeju, O.O.; Aboua, G.Y.; Omodanisi, E.I. Effects of Moringa oleifera on Oxidative Stress, Apoptotic and Inflammatory Biomarkers in Streptozotocin-Induced Diabetic Animal Model. S. Afr. J. Bot. 2020, 129, 354–365. [Google Scholar] [CrossRef]
  78. Suresh, S.; Chhipa, A.S.; Gupta, M.; Lalotra, S.; Sisodia, S.S.; Baksi, R.; Nivsarkar, M. Phytochemical Analysis and Pharmacological Evaluation of Methanolic Leaf Extract of Moringa oleifera Lam. in Ovalbumin Induced Allergic Asthma. S. Afr. J. Bot. 2020, 130, 484–493. [Google Scholar] [CrossRef]
  79. Kumbhare, M.; Sivakumar, T. Anti-Inflammatory and Analgesic Activity of Stem Bark of Moringa oleifera. Pharmacologyonline 2011, 3, 641–650. [Google Scholar]
  80. Upadhye, K.P.; Rangari, V.D.; Mathur, V.B. Antimigraine Activity Study of Moringa oleifera Leaf Juice. Int. J. Green Pharm. 2012, 6, 204. [Google Scholar] [CrossRef]
  81. Adeneye, A. Herbal Pharmacotherapy of Hypertension. Phytother. Manag. Diabetes Hypertens. 2016, 2, 3–71. [Google Scholar] [CrossRef]
  82. Gupta, L.; M., T.; S., A.S.; Nayabaniya, A. Phytochemical Screening and In Vitro Evaluation of Antibacterial and Antioxidant Properties of Moringa oleifera Linn Leaf Extract. Res. J. Pharm. Technol. 2023, 16, 4512–4518. [Google Scholar] [CrossRef]
  83. Saadabi, A.M.; Zaid, I.E.A. An In Vitro Antimicrobial Activity of Moringa oleifera L. Seed Extracts Against Different Groups of Microorganisms. Aust. J. Basic Appl. Sci. 2011, 5, 129–134. [Google Scholar]
  84. Akintelu, S.A.; Folorunso, A.S.; Oyebamiji, A.K. Phytochemical and Antibacterial Investigation of Moringa oleifera Seed: Experimental and Computational Approaches. Eclética Química 2021, 46, 17–25. [Google Scholar] [CrossRef]
  85. Peixoto, J.R.O.; Silva, G.C.; Costa, R.A.; de Sousa Fontenelle, J.R.; Vieira, G.H.F.; Filho, A.A.F.; Vieira, R.H.S.F. In Vitro Antibacterial Effect of Aqueous and Ethanolic Moringa Leaf Extracts. Asian Pac. J. Trop. Med. 2011, 4, 201–204. [Google Scholar] [CrossRef]
  86. De Oliveira, A.M.; Da Silva Férnandes, M.; De Abreu Filho, B.A.; Gomes, R.G.; Bergamasco, R. Inhibition and Removal of Staphylococcal Biofilms Using Moringa oleifera Lam. Aqueous and Saline Extracts. J. Environ. Chem. Eng. 2018, 6, 2011–2016. [Google Scholar] [CrossRef]
  87. Aboagye, G.; Navele, M.; Essuman, E. Protocols for Assessing Antibacterial and Water Coagulation Potential of Moringa oleifera Seed Powder. MethodsX 2021, 8, 101283. [Google Scholar] [CrossRef] [PubMed]
  88. Padla, E.P.; Solis, L.T.; Levida, R.M.; Shen, C.C.; Ragasa, C.Y. Antimicrobial Isothiocyanates from the Seeds of Moringa oleifera Lam. Z. Für Naturforschung C 2012, 67, 557–564. [Google Scholar] [CrossRef] [PubMed]
  89. Ashraf, Q.; Aqib, A.I.; Majeed, H.; Haq, S.U.; Nisar, M.F.; Usman, M.; Muneer, A.; Batool, S.; Ataya, F.S. Effective Synergism of Moringa oleifera with Antibiotics, Body Growth, and Decoction Replacement. S. Afr. J. Bot. 2024, 175, 628–636. [Google Scholar] [CrossRef]
  90. Shashikala, B.S.; Ashwini, K.R.; Lavanya, D.R.; Jessica, Y.; Ushasri, P.; Sunitha, D.V.; Shobha, G.; Dsouza, P.P.P.; Naidu, K.C.B. Moringa oleifera Leaf-Based Zinc Oxide Nanoparticles: Green Synthesis, Characterization, and Their Antibacterial Investigations. Inorg. Chem. Commun. 2024, 170, 113355. [Google Scholar] [CrossRef]
  91. Ahmed, M.; Marrez, D.A.; Abdelmoeen, N.M.; Mahmoud, E.A.; Ali, M.A.S.; Decsi, K.; Tóth, Z. Proximate Analysis of Moringa oleifera Leaves and the Antimicrobial Activities of Successive Leaf Ethanolic and Aqueous Extracts Compared with Green Chemically Synthesized Ag-NPs and Crude Aqueous Extract against Some Pathogens. Int. J. Mol. Sci. 2023, 24, 3529. [Google Scholar] [CrossRef]
  92. Elgamily, H.; Moussa, A.; Elboraey, A.; El-Sayed, H.; Al-Moghazy, M.; Abdalla, A. Microbiological Assessment of Moringa oleifera Extracts and Its Incorporation in Novel Dental Remedies against Some Oral Pathogens+. Open Access Maced. J. Med. Sci. 2016, 4, 585–590. [Google Scholar] [CrossRef]
  93. Oluduro, A.O. Evaluation of Antimicrobial Properties and Nutritional Potentials of Moringa oleifera Lam. Leaf in South-Western Nigeria. Malays. J. Microbiol. 2012, 8, 59–67. [Google Scholar] [CrossRef]
  94. Maurya, S.; Singh, A. Clinical Efficacy of Moringa oleifera Lam. Stems Bark in Urinary Tract Infections. Int. Sch. Res. Not. 2014, 2014, 1–7. [Google Scholar] [CrossRef]
  95. Patel, P.; Patel, N.; Dhara, P.; Desai, S.; Meshram, D. Phytochemical Analysis and Antifungal Activity of Moringa oleifera. Int. J. Pharm. Pharm. Sci. 2014, 6, 144–147. [Google Scholar]
  96. Abbas, H.; Atiyah, M. Anti-Fungal Activities of Aqueous and Alcoholic Leaf Extracts of Moringa oleifera Lam. on Candida albicans Isolated from Diabetic Foot Infections. AIP Conf. Proc. 2023, 2414, 020008. [Google Scholar] [CrossRef]
  97. Chuang, P.; Lee, C.; Chou, J.; Murugan, M.; Shieh, B.; Chen, H. Anti-Fungal Activity of Crude Extracts and Essential Oil of Moringa oleifera Lam. Bioresour. Technol. 2007, 98, 232–236. [Google Scholar] [CrossRef] [PubMed]
  98. Nessma, E.; Metwally, M.A.; Samah, H. Assessment of Oil and Seed Extracts of Moringa oleifera for Promising Anticandidal Activity in Autistic Children. Egypt. J. Bot. 2022, 62, 825–835. [Google Scholar] [CrossRef]
  99. El Mohamedy, R.S.R.; Abdalla, A.M. Evaluation of Antifungal Activity of Moringa oleifera Extracts as Natural Fungicide against Some Plant Pathogenic Fungi In-Vitro. J. Agric. Technol. 2014, 10, 963–982. [Google Scholar]
  100. Gifoni, J.M.; Oliveira, J.T.A.; Oliveira, H.D.; Batista, A.B.; Pereira, M.L.; Gomes, A.S.; Oliveira, H.P.; Grangeiro, T.B.; Vasconcelos, I.M. A Novel Chitin-Binding Protein from Moringa oleifera Seed with Potential for Plant Disease Control. Pept. Sci. 2012, 98, 406–415. [Google Scholar] [CrossRef]
  101. Batista, A.B.; Oliveira, J.T.A.; Gifoni, J.M.; Pereira, M.L.; Almeida, M.G.G.; Gomes, V.M.; Da Cunha, M.; Ribeiro, S.F.F.; Dias, G.B.; Beltramini, L.M.; et al. New Insights into the Structure and Mode of Action of Mo-CBP3, an Antifungal Chitin-Binding Protein of Moringa oleifera Seeds. PLoS ONE 2014, 9, e111427. [Google Scholar] [CrossRef]
  102. Neto, J.X.S.; Pereira, M.L.; Oliveira, J.T.A.; Rocha-Bezerra, L.C.B.; Lopes, T.D.P.; Costa, H.P.S.; Sousa, D.O.B.; Rocha, B.A.M.; Grangeiro, T.B.; Freire, J.E.C.; et al. A Chitin-Binding Protein Purified from Moringa oleifera Seeds Presents Anticandidal Activity by Increasing Cell Membrane Permeability and Reactive Oxygen Species Production. Front. Microbiol. 2017, 8, 980. [Google Scholar] [CrossRef]
  103. Lopes, T.D.P.; Souza, P.F.N.; da Costa, H.P.S.; Pereira, M.L.; da Silva Neto, J.X.; de Paula, P.C.; Brilhante, R.S.N.; Oliveira, J.T.A.; Vasconcelos, I.M.; Sousa, D.O.B. Mo-CBP4, a Purified Chitin-Binding Protein from Moringa oleifera Seeds, Is a Potent Antidermatophytic Protein: In Vitro Mechanisms of Action, in Vivo Effect against Infection, and Clinical Application as a Hydrogel for Skin Infection. Int. J. Biol. Macromol. 2020, 149, 432–442. [Google Scholar] [CrossRef]
  104. Wen, Y.; Li, W.; Su, R.; Yang, M.; Zhang, N.; Li, X.; Li, L.; Sheng, J.; Tian, Y. Multi-Target Antibacterial Mechanism of Moringin from Moringa oleifera Seeds Against Listeria monocytogenes. Front. Microbiol. 2022, 13, 925291. [Google Scholar] [CrossRef]
  105. Feustel, S.; Ayón-Pérez, F.; Sandoval-Rodriguez, A.; Rodríguez-Echevarría, R.; Contreras-Salinas, H.; Armendáriz-Borunda, J.; Sánchez-Orozco, L.V. Protective Effects of Moringa oleifera on HBV Genotypes C and H Transiently Transfected Huh7 Cells. J. Immunol. Res. 2017, 2017, 6063850. [Google Scholar] [CrossRef]
  106. Kurokawa, M.; Wadhwani, A.; Kai, H.; Hidaka, M.; Yoshida, H.; Sugita, C.; Watanabe, W.; Matsuno, K.; Hagiwara, A. Activation of Cellular Immunity in Herpes Simplex Virus Type 1-Infected Mice by the Oral Administration of Aqueous Extract of Moringa oleifera Lam. Leaves. Phytother. Res. 2016, 30, 797–804. [Google Scholar] [CrossRef]
  107. Souza, H.C.A.; Souza, M.D.A.; Sousa, C.S.; Viana, E.K.A.; Alves, S.K.S.; Marques, A.O.; Ribeiro, A.S.N.; de Sousa do Vale, V.; Islam, M.T.; de Miranda, J.A.L.; et al. Molecular Docking and ADME-TOX Profiling of Moringa oleifera Constituents against SARS-CoV-2. Adv. Respir. Med. 2023, 91, 464–485. [Google Scholar] [CrossRef] [PubMed]
  108. Mathpal, S.; Sharma, P.; Joshi, T.; Joshi, T.; Pande, V.; Chandra, S. Screening of Potential Bio-Molecules from Moringa oleifera against SARS-CoV-2 Main Protease Using Computational Approaches. J. Biomol. Struct. Dyn. 2022, 40, 9885–9896. [Google Scholar] [CrossRef] [PubMed]
  109. Sen, D.; Bhaumik, S.; Debnath, P.; Debnath, S. Potentiality of Moringa oleifera against SARS-CoV-2: Identified by a Rational Computer Aided Drug Design Method. J. Biomol. Struct. Dyn. 2022, 40, 7517–7534. [Google Scholar] [CrossRef]
  110. Imran, I.; Altaf, I.; Ashraf, M.; Javeed, A.; Munir, N.; Bashir, R. In Vitro Evaluation of Antiviral Activity of Leaf Extracts of Azadirachta indica, Moringa oleifera, and Morus alba against the Foot and Mouth Disease Virus on BHK-21 Cell Line. Sci. Asia 2016, 42, 392. [Google Scholar] [CrossRef]
  111. Eze, D.C.; Okwor, E.C.; Okoye, J.O.A.; Onah, D.N. Immunologic Effects of Moringa oleifera Methanolic Leaf Extract in Chickens Infected with Newcastle Disease Virus (Kudu 113) Strain. Afr. J. Pharm. Pharmacol. 2013, 7, 2231–2237. [Google Scholar] [CrossRef]
  112. Nadumane, V.; Nair, S. Anticancer, Cytotoxic Potential of Moringa oleifera Extracts on HeLa Cell Line. Jundishapur J. Nat. Pharm. Prod. 2011, 2, 138. [Google Scholar] [CrossRef]
  113. Khalafalla, M.M.; Abdellatef, E.; Dafalla, H.M.; Nassrallah, A.A.; Aboul-Enein, K.M.; Lightfoot, D.A.; El-Deeb, F.E.; El-Shemy, H.A. Active Principle from Moringa oleifera Lam Leaves Effective against Two Leukemias and a Hepatocarcinoma. Afr. J. Biotechnol. 2010, 9, 8467–8471. [Google Scholar] [CrossRef]
  114. Reda, F.; Borjac, J.; Fakhouri, R.; Usta, J. Cytotoxic Effect of Moringa oleifera on Colon Cancer Cell Lines. Acta Hortic. 2017, 1158, 269–278. [Google Scholar] [CrossRef]
  115. Jung, I.L. Soluble Extract from Moringa oleifera Leaves with a New Anticancer Activity. PLoS ONE 2014, 9, e95492. [Google Scholar] [CrossRef]
  116. Al-Asmari, A.K.; Albalawi, S.M.; Athar, M.T.; Khan, A.Q.; Al-Shahrani, H.; Islam, M. Moringa oleifera as an Anti-Cancer Agent against Breast and Colorectal Cancer Cell Lines. PLoS ONE 2015, 10, e0135814. [Google Scholar] [CrossRef]
  117. Do, B.H.; Hoang, N.S.; Nguyen, T.P.T.; Ho, N.Q.C.; Le, T.L.; Doan, C.C. Phenolic Extraction of Moringa oleifera Leaves Induces Caspase-Dependent and Caspase-Independent Apoptosis through the Generation of Reactive Oxygen Species and the Activation of Intrinsic Mitochondrial Pathway in Human Melanoma Cells. Nutr. Cancer 2021, 73, 869–888. [Google Scholar] [CrossRef] [PubMed]
  118. Xie, J.; Peng, L.; Yang, M.; Jiang, W.; Mao, J.; Shi, C.; Tian, Y.; Sheng, J. Alkaloid Extract of Moringa oleifera Lam. Exerts Antitumor Activity in Human Non-Small-Cell Lung Cancer via Modulation of the JAK2/STAT3 Signaling Pathway. Evid.-Based Complement. Altern. Med. 2021, 2021, 5591687. [Google Scholar] [CrossRef] [PubMed]
  119. Khan, F.; Pandey, P.; Ahmad, V.; Upadhyay, T.K. Moringa oleifera Methanolic Leaves Extract Induces Apoptosis and G0/G1 Cell Cycle Arrest via Downregulation of Hedgehog Signaling Pathway in Human Prostate PC-3 Cancer Cells. J. Food Biochem. 2020, 44, e13338. [Google Scholar] [CrossRef]
  120. Berkovich, L.; Earon, G.; Ron, I.; Rimmon, A.; Vexler, A.; Lev-Ari, S. Moringa oleifera Aqueous Leaf Extract Down-Regulates Nuclear Factor-kappaB and Increases Cytotoxic Effect of Chemotherapy in Pancreatic Cancer Cells. BMC Complement. Altern. Med. 2013, 13, 212. [Google Scholar] [CrossRef]
  121. Sultan, R.; Ahmed, A.; Wei, L.; Saeed, H.; Islam, M.; Ishaq, M. The Anticancer Potential of Chemical Constituents of Moringa oleifera Targeting CDK-2 Inhibition in Estrogen Receptor Positive Breast Cancer Using in-Silico and in Vitro Approaches. BMC Complement. Med. Ther. 2023, 23, 396. [Google Scholar] [CrossRef]
  122. Sreelatha, S.; Padma, P. Modulatory Effects of Moringa oleifera Extracts against Hydrogen Peroxide-Induced Cytotoxicity and Oxidative Damage. Hum. Exp. Toxicol. 2011, 30, 1359–1368. [Google Scholar] [CrossRef]
  123. Wang, C.; Wu, R.; Sargsyan, D.; Zheng, M.; Li, S.; Yin, R.; Su, S.; Raskin, I.; Kong, A.-N. CpG Methyl-Seq and RNA-Seq Epigenomic and Transcriptomic Studies on the Preventive Effects of Moringa Isothiocyanate in Mouse Epidermal JB6 Cells Induced by the Tumor Promoter TPA. J. Nutr. Biochem. 2019, 68, 69–78. [Google Scholar] [CrossRef]
  124. Kraiphet, S.; Butryee, C.; Rungsipipat, A.; Budda, S.; Rattanapinyopituk, K. Apoptosis Induced by Moringa oleifera Lam. Pod in Mouse Colon Carcinoma Model. Comp. Clin. Pathol. 2018, 27, 21–30. [Google Scholar] [CrossRef]
  125. Patriota, L.L.D.S.; Ramos, D.D.B.M.; Dos Santos, A.C.L.A.; Silva, Y.A.; Gama, E.; Silva, M.; Torres, D.J.L.; Procópio, T.F.; De Oliveira, A.M.; Coelho, L.C.B.B.; et al. Antitumor Activity of Moringa oleifera (Drumstick Tree) Flower Trypsin Inhibitor (MoFTI) in Sarcoma 180-Bearing Mice. Food Chem. Toxicol. 2020, 145, 111691. [Google Scholar] [CrossRef]
  126. Laftah, A.H.; Alhelfi, N.; Al Salait, S.K.; Altemimi, A.B.; Tabandeh, M.R.; Tsakali, E.; Van Impe, J.F.M.; Abd El-Maksoud, A.A.; Abedelmaksoud, T.G. Mitigation of Doxorubicin-Induced Liver Toxicity in Mice Breast Cancer Model by Green Tea and Moringa oleifera Combination: Targeting Apoptosis, Inflammation, and Oxidative Stress. J. Funct. Foods 2025, 124, 106626. [Google Scholar] [CrossRef]
  127. Zunica, E.R.M.; Yang, S.; Coulter, A.; White, C.; Kirwan, J.P.; Gilmore, L.A. Moringa oleifera Seed Extract Concomitantly Supplemented with Chemotherapy Worsens Tumor Progression in Mice with Triple Negative Breast Cancer and Obesity. Nutrients 2021, 13, 2923. [Google Scholar] [CrossRef] [PubMed]
  128. Toson, E.; Elbakry, K.; Serag, M.; Aboser, M. Hepatoprotective Effect of Moringa oleifera Leaves Extract against Carbon Tetrachloride-Induced Liver Damage in Rats. World J. Pharm. Pharm. Sci. 2016, 5, 76–89. [Google Scholar] [CrossRef]
  129. Kumar, P.; Singh, K.; Kumar, A. Hepatoprotective Studies on Aerial Parts of Moringa oleifera Lam. on Carbon Tetrachloride Induced Liver Cell Damage in Albino Rats. Ann. Biol. Res. 2010, 1, 27–35. [Google Scholar]
  130. Toppo, R.; Roy, B.K.; Gora, R.H.; Baxla, S.L.; Kumar, P. Hepatoprotective Activity of Moringa oleifera against Cadmium Toxicity in Rats. Vet. World 2015, 8, 537–540. [Google Scholar] [CrossRef]
  131. Fotio, A.L.; Nguepi, M.S.D.; Tonfack, L.B.; Temdie, R.J.G.; Nguelefack, T.B. Acetaminophen Induces Liver Injury and Depletes Glutathione in Mice Brain: Prevention by Moringa oleifera Extract. S. Afr. J. Bot. 2020, 129, 317–323. [Google Scholar] [CrossRef]
  132. Abd-Elnaby, Y.A.; El Sayed, I.E.; AbdEldaim, M.A.; Badr, E.A.; Abdelhafez, M.M.; Elmadbouh, I. Anti-Inflammatory and Antioxidant Effect of Moringa oleifera against Bisphenol-A-Induced Hepatotoxicity. Egypt. Liver J. 2022, 12, 57. [Google Scholar] [CrossRef]
  133. El-Fiky, S.A.; Hassan, N.A. Protective Role of Moringa oleifera Leaf Extract as a Natural Antioxidant against Toxic Effect of Paracetamol on Male Mice. World J. Pharm. 2018, 7, 115–135. [Google Scholar]
  134. Abdel Fattah, M.E.; Sobhy, H.M.; Reda, A.; Abdelrazek, H.M.A. Hepatoprotective Effect of Moringa oleifera Leaves Aquatic Extract against Lead Acetate-Induced Liver Injury in Male Wistar Rats. Environ. Sci. Pollut. Res. 2020, 27, 43028–43043. [Google Scholar] [CrossRef]
  135. Farid, A.S.; Hegazy, A.M. Ameliorative Effects of Moringa oleifera Leaf Extract on Levofloxacin-Induced Hepatic Toxicity in Rats. Drug Chem. Toxicol. 2020, 43, 616–622. [Google Scholar] [CrossRef]
  136. Owens, F.S.; Dada, O.; Cyrus, J.W.; Adedoyin, O.O.; Adunlin, G. The Effects of Moringa oleifera on Blood Glucose Levels: A Scoping Review of the Literature. Complement. Ther. Med. 2020, 50, 102362. [Google Scholar] [CrossRef]
  137. Jaiswal, D.; Rai, P.K.; Mehta, S.; Chatterji, S.; Shukla, S.; Rai, D.K.; Sharma, G.; Sharma, B.; Khair, S.; Watal, G. Role of Moringa oleifera in Regulation of Diabetes-Induced Oxidative Stress. Asian Pac. J. Trop. Med. 2013, 6, 426–432. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, G.-L.; Xu, Y.-B.; Wu, J.-L.; Li, N.; Guo, M.-Q. Hypoglycemic and Hypolipidemic Effects of Moringa oleifera Leaves and Their Functional Chemical Constituents. Food Chem. 2020, 333, 127478. [Google Scholar] [CrossRef] [PubMed]
  139. Hossain, U.; Das, A.K.; Ghosh, S.; Sil, P.C. An Overview on the Role of Bioactive α-Glucosidase Inhibitors in Ameliorating Diabetic Complications. Food Chem. Toxicol. 2020, 145, 111738. [Google Scholar] [CrossRef]
  140. Thongrung, R.; Pannangpetch, P.; Senggunprai, L.; Sangkhamanon, S.; Boonloh, K.; Tangsucharit, P. Moringa oleifera Leaf Extract Ameliorates Early Stages of Diabetic Nephropathy in Streptozotocin-Induced Diabetic Rats. J. Appl. Pharm. Sci. 2023, 13, 158–166. [Google Scholar] [CrossRef]
  141. Aljazzaf, B.; Regeai, S.; Elghmasi, S.; Alghazir, N.; Balgasim, A.; Hdud Ismail, I.M.; Eskandrani, A.A.; Shamlan, G.; Alansari, W.S.; Al-Farga, A.; et al. Evaluation of Antidiabetic Effect of Combined Leaf and Seed Extracts of Moringa oleifera (Moringaceae) on Alloxan-Induced Diabetes in Mice: A Biochemical and Histological Study. Oxid. Med. Cell. Longev. 2023, 2023, 9136217. [Google Scholar] [CrossRef]
  142. Jiang, X.; Tang, J.; Gao, Z.; Chen, C.; Li, W.; Peng, L.; Li, H.; Zhao, H.; Zhao, Y. Exploring the Protection on Islet β-Cells and Hypoglycemic Effect of Moringa oleifera Stem Based on Network Pharmacology and Experimental Validation. J. Funct. Foods 2024, 122, 106499. [Google Scholar] [CrossRef]
  143. Kushwaha, S.; Chawla, P.; Kochhar, A. Effect of Supplementation of Drumstick (Moringa oleifera) and Amaranth (Amaranthus tricolor) Leaves Powder on Antioxidant Profile and Oxidative Status among Postmenopausal Women. J. Food Sci. Technol. 2014, 51, 3464–3469. [Google Scholar] [CrossRef]
  144. Gómez-Martínez, S.; Díaz-Prieto, L.; Castro, I.; Jurado, C.; Iturmendi, N.; Martín-Ridaura, M.; Calle, N.; Dueñas, M.; Picón, M.; Marcos, A.; et al. Moringa oleifera Leaf Supplementation as a Glycemic Control Strategy in Subjects with Prediabetes. Nutrients 2021, 14, 57. [Google Scholar] [CrossRef]
  145. Kumar, K.P.; Mandapaka, R.T. Effect of Moringa oleifera on Blood Glucose, LDL Levels in Type II Diabetic Obese People. Innov. J. Med. Health Sci. 2013, 3, 23–25. [Google Scholar]
  146. Randriamboavonjy, J.I.; Loirand, G.; Vaillant, N.; Lauzier, B.; Derbré, S.; Michalet, S.; Pacaud, P.; Tesse, A. Cardiac Protective Effects of Moringa oleifera Seeds in Spontaneous Hypertensive Rats. Am. J. Hypertens. 2016, 29, 873–881. [Google Scholar] [CrossRef]
  147. Li, Y.-J.; Ji, Q.-Q.; Wang, Z.; Shen, L.-H.; He, B. Moringa oleifera Seeds Mitigate Myocardial Injury and Prevent Ventricular Failure Induced by Myocardial Infarction. Am. J. Transl. Res. 2020, 12, 4511–4521. [Google Scholar] [PubMed]
  148. Nandave, M.; Ojha, S.K.; Joshi, S.; Kumari, S.; Arya, D.S. Moringa oleifera Leaf Extract Prevents Isoproterenol-Induced Myocardial Damage in Rats: Evidence for an Antioxidant, Antiperoxidative, and Cardioprotective Intervention. J. Med. Food 2009, 12, 47–55. [Google Scholar] [CrossRef] [PubMed]
  149. Oseni, O.; Ogunmoyole, T.; Idowu, K. Lipid Profile and Cardioprotective Effects of Aqueous Extract of Moringa oleifera (Lam) Leaf on Bromate-Induced Cardiotoxicity in Wistar Albino Rats. Eur. J. Adv. Res. Biol. Life Sci. 2015, 3, 52–66. [Google Scholar]
  150. Odii, E.; Okoro, J.; Okorie, N.; Chukwuka, C.; Okoye, C.; Okeke, E.; Ejere, V.; Eyo, J. Antihypertensive Effect of Moringa oleifera (Moringaceae) Methanolic Leaf Extract (MoMLE) on Cricetomys gambianus (Muridae). Trop. J. Nat. Prod. Res. 2022, 6, 1695–1700. [Google Scholar] [CrossRef]
  151. Ghimire, S.; Subedi, L.; Acharya, N.; Gaire, B.P. Moringa oleifera: A Tree of Life as a Promising Medicinal Plant for Neurodegenerative Diseases. J. Agric. Food Chem. 2021, 69, 14358–14371. [Google Scholar] [CrossRef] [PubMed]
  152. González-Burgos, E.; Ureña-Vacas, I.; Sánchez, M.; Gómez-Serranillos, M.P. Nutritional Value of Moringa oleifera Lam. Leaf Powder Extracts and Their Neuroprotective Effects via Antioxidative and Mitochondrial Regulation. Nutrients 2021, 13, 2203. [Google Scholar] [CrossRef]
  153. Hashim, J.F.; Vichitphan, S.; Boonsiri, P.; Vichitphan, K. Neuroprotective Assessment of Moringa oleifera Leaves Extract against Oxidative-Stress-Induced Cytotoxicity in SH-SY5Y Neuroblastoma Cells. Plants 2021, 10, 889. [Google Scholar] [CrossRef]
  154. Kirisattayakul, W.; Wattanathorn, J.; Tong-Un, T.; Muchimapura, S.; Wannanon, P.; Jittiwat, J. Cerebroprotective Effect of Moringa oleifera against Focal Ischemic Stroke Induced by Middle Cerebral Artery Occlusion. Oxid. Med. Cell. Longev. 2013, 2013, 951415. [Google Scholar] [CrossRef]
  155. Sutalangka, C.; Wattanathorn, J.; Muchimapura, S.; Thukham-mee, W. Moringa oleifera Mitigates Memory Impairment and Neurodegeneration in Animal Model of Age-Related Dementia. Oxid. Med. Cell. Longev. 2013, 2013, 695936. [Google Scholar] [CrossRef]
  156. Idoga, E.; Ambali, S.; Ayo, J. Assessment of Antioxidant and Neuroprotective Activities of Methanol Extract of Moringa oleifera Lam. Leaves in Subchronic Chlorpyrifos-Intoxicated Rats. Comp. Clin. Pathol. 2018, 27, 917–925. [Google Scholar] [CrossRef]
  157. Raza, C.; Mohsin, S.; Faheem, M.; Hanif, U.; Alkhathlan, H.Z.; Shaik, M.R.; Riaz, H.A.; Anjum, R.; Jurrat, H.; Khan, M. In Vivo Study of Moringa oleifera Seed Extracts as Potential Sources of Neuroprotection against Rotenone-Induced Neurotoxicity. Plants 2024, 13, 1479. [Google Scholar] [CrossRef] [PubMed]
  158. Abdelsayed, E.M.; Medhat, D.; Mandour, Y.M.; Hanafi, R.S.; Motaal, A.A. Niazimicin: A Thiocarbamate Glycoside from Moringa oleifera Lam. Seeds with a Novel Neuroprotective Activity. J. Food Biochem. 2021, 45, e13992. [Google Scholar] [CrossRef]
  159. Mahaman, Y.A.R.; Huang, F.; Wu, M.; Wang, Y.; Wei, Z.; Bao, J.; Salissou, M.T.M.; Ke, D.; Wang, Q.; Liu, R.; et al. Moringa oleifera Alleviates Homocysteine-Induced Alzheimer’s Disease-Like Pathology and Cognitive Impairments. J. Alzheimer’s Dis. 2018, 63, 1141–1159. [Google Scholar] [CrossRef]
  160. Mahaman, Y.A.R.; Feng, J.; Huang, F.; Salissou, M.T.M.; Wang, J.; Liu, R.; Zhang, B.; Li, H.; Zhu, F.; Wang, X. Moringa oleifera Alleviates Aβ Burden and Improves Synaptic Plasticity and Cognitive Impairments in APP/PS1 Mice. Nutrients 2022, 14, 4284. [Google Scholar] [CrossRef] [PubMed]
  161. Onasanwo, S.A.; Adamaigbo, V.O.; Adebayo, O.G.; Eleazer, S.E. Moringa oleifera-Supplemented Diet Protect against Cortico-Hippocampal Neuronal Degeneration in Scopolamine-Induced Spatial Memory Deficit in Mice: Role of Oxido-Inflammatory and Cholinergic Neurotransmission Pathway. Metab. Brain Dis. 2021, 36, 2445–2460. [Google Scholar] [CrossRef]
  162. Kambuno, N.T.; Putra, A.G.A.; Louisa, M.; Wuyung, P.E.; Timan, I.S.; Silaen, O.S.M.; Sukria, H.A.; Supali, T. Moringa oleifera Leaf Extract Improves Cognitive Function in Rat Offspring Born to Protein-Deficient Mothers. Biomedicines 2025, 13, 346. [Google Scholar] [CrossRef]
  163. Abo-Elsoud, R.A.E.A.; Abdelaziz, S.A.M.; Eldaim, M.A.A.; Hazzaa, S.M. Moringa oleifera Alcoholic Extract Protected Stomach from Bisphenol A–Induced Gastric Ulcer in Rats via Its Anti-Oxidant and Anti-Inflammatory Activities. Environ. Sci. Pollut. Res. 2022, 29, 68830–68841. [Google Scholar] [CrossRef]
  164. Chedi, B.F.L.; Garba, K.; Shuai’bu, A.B.; Basheer, A.Z.; Chedi, B. Evaluation of the Antiulcer Activity of Aqueous Seed Extract of Moringa oleifera Lamarck (Moringaceae). Trop. J. Nat. Prod. Res. 2018, 2, 140–144. [Google Scholar]
  165. Mohamed Husien, H.; Peng, W.; Su, H.; Zhou, R.; Tao, Y.; Huang, J.; Liu, M.; Bo, R.; Li, J. Moringa oleifera Leaf Polysaccharide Alleviates Experimental Colitis by Inhibiting Inflammation and Maintaining Intestinal Barrier. Front. Nutr. 2022, 9, 1055791. [Google Scholar] [CrossRef]
  166. Woldeyohannes, M.G.; Eshete, G.T.; Abiye, A.A.; Hailu, A.E.; Huluka, S.A.; Tadesse, W.T. Antidiarrheal and Antisecretory Effect of 80% Hydromethanolic Leaf Extract of Moringa stenopetala Baker f. in Mice. Biochem. Res. Int. 2022, 2022, 5768805. [Google Scholar] [CrossRef]
  167. Adji, A.S.; Atika, N.; Kusbijantoro, Y.B.; Billah, A.; Putri, A.; Handajani, F. A Review of Leaves and Seeds Moringa oleifera Extract: The Potential Moringa oleifera as Antibacterial, Anti-Inflammatory, Antidiarrhoeal, and Antiulcer Approaches to Bacterial Gastroenteritis. Open Access Maced. J. Med. Sci. 2022, 10, 305–313. [Google Scholar] [CrossRef]
  168. Mabrouki, L.; Rjeibi, I.; Taleb, J.; Zourgui, L. Cardiac Ameliorative Effect of Moringa oleifera Leaf Extract in High-Fat Diet-Induced Obesity in Rat Model. BioMed Res. Int. 2020, 2020, 6583603. [Google Scholar] [CrossRef] [PubMed]
  169. Joung, H.; Kim, B.; Park, H.; Lee, K.; Kim, H.-H.; Sim, H.-C.; Do, H.-J.; Hyun, C.-K.; Do, M.-S. Fermented Moringa oleifera Decreases Hepatic Adiposity and Ameliorates Glucose Intolerance in High-Fat Diet-Induced Obese Mice. J. Med. Food 2017, 20, 439–447. [Google Scholar] [CrossRef] [PubMed]
  170. Alia, F.; Syamsunarno, M.; Sumirat, V.; Ghozali, M.; Atik, N. The Haematological Profiles of High Fat Diet Mice Model with Moringa oleifera Leaves Ethanol Extract Treatment. BioMed Pharmacol. J. 2019, 12, 2143–2149. [Google Scholar] [CrossRef]
  171. Ahmed, H.; Metwally, F.; Rashad, H.; Zaazaa, A.; Ezzat, S.; Salama, M. Moringa oleifera Offers a Multi-Mechanistic Approach for Management of Obesity in Rats. Int. J. Pharm. Sci. Rev. Res. 2014, 29, 98–106. [Google Scholar]
  172. Ezzat, S.M.; El Bishbishy, M.H.; Aborehab, N.M.; Salama, M.M.; Hasheesh, A.; Motaal, A.A.; Rashad, H.; Metwally, F.M. Upregulation of MC4R and PPAR-α Expression Mediates the Anti-Obesity Activity of Moringa oleifera Lam. in High-Fat Diet-Induced Obesity in Rats. J. Ethnopharmacol. 2020, 251, 112541. [Google Scholar] [CrossRef]
  173. Almatrafi, M.M.; Vergara-Jimenez, M.; Murillo, A.G.; Norris, G.H.; Blesso, C.N.; Fernandez, M.L. Moringa Leaves Prevent Hepatic Lipid Accumulation and Inflammation in Guinea Pigs by Reducing the Expression of Genes Involved in Lipid Metabolism. Int. J. Mol. Sci. 2017, 18, 1330. [Google Scholar] [CrossRef]
  174. Munir, M.; Khan, I.; Almutairi, N.S.; Almutairi, A.H.; Khan, B.; Mehboob, N. Effect of Moringa Leaves Powder on Body Weight, Glycemic Status, Lipid Profile, and Blood Pressure in Overweight Individuals with Hyperlipidemia. Ital. J. Food Sci. 2025, 37, 210–219. [Google Scholar] [CrossRef]
  175. Mansour, M.F.; Arisha, A.H.; Al-Gamal, M.; Yahia, R.; Elsayed, A.A.; Saad, S.; El-Bohi, K.M. Moringa oleifera Leaves Extract Ameliorates Melamine-Induced Testicular Toxicity in Rats. Adv. Anim. Vet. Sci. 2020, 8, 47–54. [Google Scholar] [CrossRef]
  176. Tekayev, M.; Bostancieri, N.; Saadat, K.A.S.M.; Turker, M.; Yuncu, M.; Ulusal, H.; Cicek, H.; Arman, K. Effects of Moringa oleifera Lam Extract (MOLE) in the Heat Shock Protein 70 Expression and Germ Cell Apoptosis on Experimentally Induced Cryptorchid Testes of Rats. Gene 2019, 688, 140–150. [Google Scholar] [CrossRef]
  177. Muhammad, N.; Ibrahim, K.G.; Ndhlala, A.R.; Erlwanger, K.H. Moringa oleifera Lam. Prevents the Development of High Fructose Diet-Induced Fatty Liver. S. Afr. J. Bot. 2020, 129, 32–39. [Google Scholar] [CrossRef]
  178. Attah, A.F.; Akindele, O.O.; Nnamani, P.O.; Jonah, U.J.; Sonibare, M.A.; Moody, J.O. Moringa oleifera Seed at the Interface of Food and Medicine: Effect of Extracts on Some Reproductive Parameters, Hepatic and Renal Histology. Front. Pharmacol. 2022, 13, 816498. [Google Scholar] [CrossRef] [PubMed]
  179. Sethi, N.; Nath, D.; Shukla, S.C.; Dyal, R. Abortifacient Activity of a Medicinal Plant Moringa oleifera. Anc. Sci. Life 1988, 7, 172–174. [Google Scholar] [PubMed]
  180. Hajar, A.; Abdelmounaim, B.; Hamid, K.; Jaouad, L.; Abdelfattah, A.B.; Majda, B.; Loubna, E.Y.; Mohammed, L.; Rachida, A.; Abderrahman, C. Developmental Toxicity of Moringa oleifera and Its Effect on Postpartum Depression, Maternal Behavior and Lactation. S. Afr. J. Bot. 2024, 171, 257–266. [Google Scholar] [CrossRef]
  181. Agrawal, S.S.; Vishal, D.; Sumeet, G.; Shekhar, C.; Ashish, N.; Parul, D.; Ankita, S.; Prakash, A.; Prakash, T.; Kumar, P.; et al. Antifertility Activity of Ethanol Leaf Extract of Moringa oleifera Lam in Female Wistar Rats. Indian J. Pharm. Sci. 2018, 80, 565–570. [Google Scholar] [CrossRef]
  182. Ajuogu, P.K.; Mgbere, O.O.; Bila, D.S.; McFarlane, J.R. Hormonal Changes, Semen Quality and Variance in Reproductive Activity Outcomes of Post Pubertal Rabbits Fed Moringa oleifera Lam. Leaf Powder. J. Ethnopharmacol. 2019, 233, 80–86. [Google Scholar] [CrossRef]
  183. Hu, X.-H.; Yang, X.-Y.; Lian, J.; Chen, Y.; Zheng, C.-Y.; Tao, S.-Y.; Liu, N.-N.; Liu, Q.; Jiang, G.-J. Moringa oleifera Leaf Attenuate Osteoporosis in Ovariectomized Rats by Modulating Gut Microbiota Composition and MAPK Signaling Pathway. BioMed Pharmacother. 2023, 161, 114434. [Google Scholar] [CrossRef]
  184. Men, D.; Dai, J.; Yuan, Y.; Jiang, H.; Wang, X.; Wang, Y.; Tao, L.; Sheng, J.; Tian, Y. Exploration of Anti-Osteoporotic Peptides from Moringa oleifera Leaf Proteins by Network Pharmacology, Molecular Docking, Molecular Dynamics and Cellular Assay Analyses. J. Funct. Foods 2024, 116, 106144. [Google Scholar] [CrossRef]
  185. Khan, M.I.; Siddiqui, S.; Barkat, M.d.A.; Alhodieb, F.S.; Ashfaq, F.; Barkat, H.A.; Alanezi, A.A.; Arshad, M. Moringa oleifera Leaf Extract Induces Osteogenic-like Differentiation of Human Osteosarcoma SaOS2 Cells. J. Tradit. Complement. Med. 2022, 12, 608–618. [Google Scholar] [CrossRef]
  186. El-Sherbiny, G.M.; Alluqmani, A.J.; Elsehemy, I.A.; Kalaba, M.H. Antibacterial, Antioxidant, Cytotoxicity, and Phytochemical Screening of Moringa oleifera Leaves. Sci. Rep. 2024, 14, 30485. [Google Scholar] [CrossRef]
  187. Bhattacharya, A.; Agrawal, D.; Sahu, P.; Kumar, S.; Mishra, S.; Patnaik, S. Analgesic Effect of Ethanolic Leaf Extract of Moringa oleifera on Albino Mice. Indian J. Pain 2014, 28, 89–94. [Google Scholar] [CrossRef]
  188. Zhang, M.; Hettiarachchy, S.N.; Horax, R.; Kannan, A.; Praisoody, M.D.A.; Muhundan, A.; Mallangi, C.R. Phytochemicals, Antioxidant and Antimicrobial Activity of Hibiscus sabdariffa, Centella asiatica, Moringa oleifera and Murraya koenigii Leaves. J. Med. Plants Res. 2011, 5, 6672–6680. [Google Scholar] [CrossRef]
  189. Adedapo, A.A.; Mogbojuri, O.M.; Emikpe, B.O. Safety Evaluations of the Aqueous Extract of the Leaves of Moringa oleifera in Rats. J. Med. Plants Res. 2009, 3, 586–591. [Google Scholar]
  190. Awodele, O.; Oreagba, I.A.; Odoma, S.; da Silva, J.A.T.; Osunkalu, V.O. Toxicological Evaluation of the Aqueous Leaf Extract of Moringa oleifera Lam. (Moringaceae). J. Ethnopharmacol. 2012, 139, 330–336. [Google Scholar] [CrossRef]
  191. Asiedu-Gyekye, I.J.; Frimpong-Manso, S.; Awortwe, C.; Antwi, D.A.; Nyarko, A.K. Micro- and Macroelemental Composition and Safety Evaluation of the Nutraceutical Moringa oleifera Leaves. J. Toxicol. 2014, 2014, 786979. [Google Scholar] [CrossRef]
  192. Ajibade, T.O.; Arowolo, R.; Olayemi, F.O. Phytochemical Screening and Toxicity Studies on the Methanol Extract of the Seeds of Moringa oleifera. J. Combin. Theory Med. 2013, 10, 11–16. [Google Scholar] [CrossRef]
  193. Hernandez-Aguilar, C.; Dominguez-Pacheco, A.; Valderrama-Bravo, C.; Cruz-Orea, A.; Ortiz, E.M.; Ivanov, R.; Ordonez-Miranda, J. Photoacoustic characterization of wheat bread mixed with Moringa oleifera. Curr. Res. Food Sci. 2021, 4, 521–531. [Google Scholar] [CrossRef]
  194. Giuberti, G.; Bresciani, A.; Cervini, M.; Frustace, A.; Marti, A. Moringa oleifera L. leaf powder as ingredient in gluten-free biscuits: Nutritional and physicochemical characteristics. Eur. Food Res. Technology 2021, 247, 687–694. [Google Scholar] [CrossRef]
  195. Paz, R.D.; Landázuri, A.C.; Vernaza, M.G. Development of a cereal-based product using residual Moringa oleifera Lam. seed powder biomass and pseudo-plastic behavior of the dough mixtures. Nutr. Food Sci. 2020, 51, 594–603. [Google Scholar] [CrossRef]
  196. Athira Nair, D.; James, T.J.; Sreelatha, S.L.; Kariyil, B.J.; Nair, S.N. Moringa oleifera (Lam.): A natural remedy for ageing? Nat. Product Res. 2020, 35, 6216–6222. [Google Scholar] [CrossRef]
  197. Kumalaningsih, S.; Arwani, M. Nutritious pure herbal whitening suxcream processed from seed and leaf of Moringa oleifera fortified with Red Rice. Pharmacogn. J. 2018, 10, 77–80. [Google Scholar] [CrossRef]
  198. Available online: https://florafarma.com/ (accessed on 26 May 2025).
  199. Available online: https://us.nuxe.com/ (accessed on 26 May 2025).
  200. Available online: https://www.thebodyshop.bg/moringa (accessed on 26 May 2025).
  201. Available online: https://makeup.bg/product/531425/ (accessed on 26 May 2025).
  202. Available online: https://incidecoder.com/ (accessed on 26 May 2025).
  203. Available online: https://thebalm.com/ (accessed on 26 May 2025).
  204. Althomali, A.; Daghestani, M.H.; Almukaynizi, F.B.; Al-Zahrani, S.A.; Awad, M.A.; Merghani, N.M.; Bukhari, W.I.; Ibrahim, E.M.; Alzahrani, S.M.; Altowair, N.; et al. Anti-Colon Cancer Activities of Green-Synthesized Moringa Oleifera–AgNPs against Human Colon Cancer Cells. Green Process. Synth. 2022, 11, 545–554. [Google Scholar] [CrossRef]
  205. Singhal, A.K.; Jarald, E.E.; Showkat, A.; Daud, A. In Vitro Evaluation of Moringa Oleifera Gum for Colon-Specific Drug Delivery. Int. J. Pharm. Investig. 2012, 2, 48–51. [Google Scholar] [CrossRef]
  206. Zhang, J. Moringaceae Tablet and Its Producing Method. CN 1935165, 22 September 2005. [Google Scholar]
  207. Gomez, R. Dietary Supplement Composition. WO2011098819, 18 August 2011. [Google Scholar]
  208. Gokaraju, G.R. Synergistic Phytochemical Composition for the Treatment of Obesity. US 8541383 B2, 24 September 2013. [Google Scholar]
  209. Vankwani, S.; Ansari, A.; Azhar, A.; Galani, S. Synergistic evaluation of Moringa oleifera extract and ß-lactam antibiotic to restore the susceptibility of methicillin-resistant Staphylococcus aureus. Mol. Biol. Rep. 2022, 49, 421–432. [Google Scholar] [CrossRef]
  210. Abu-Hussien, S.H.; Nasry, A.R.; Samy, Z.; El-Sayed, S.M.; Bakry, A.; Ebeed, N.; Elhariry, H.; ElNoby, T. Synergistic Antimicrobial Activity of Essential Oils Mixture of Moringa Oleifera, Cinnamomum Verum and Nigella Sativa against Staphylococcus Aureus Using L-Optimal Mixture Design. AMB Express 2025, 15, 15. [Google Scholar] [CrossRef]
  211. Kaur, G.; Invally, M.; Sanzagiri, R.; Buttar, H.S. Evaluation of the Antidepressant Activity of Moringa Oleifera Alone and in Combination with Fluoxetine. J. Ayurveda Integr. Med. 2015, 6, 273–279. [Google Scholar] [CrossRef]
  212. Olawoye, O.S.; Adeagbo, B.A.; Bolaji, O.O. Moringa Oleifera Leaf Powder Alters the Pharmacokinetics of Amodiaquine in Healthy Human Volunteers. J. Clin. Pharm. Ther. 2018, 43, 626–632. [Google Scholar] [CrossRef]
  213. Fantoukh, O.I.; Albadry, M.A.; Parveen, A.; Hawwal, M.F.; Majrashi, T.; Ali, Z.; Khan, S.I.; Chittiboyina, A.G.; Khan, I.A. Isolation, Synthesis, and Drug Interaction Potential of Secondary Metabolites Derived from the Leaves of Miracle Tree (Moringa Oleifera) against CYP3A4 and CYP2D6 Isozymes. Phytomedicine 2019, 60, 153010. [Google Scholar] [CrossRef]
  214. Kumolosasi, E.; Cheong, M.L.C.; Ahmad Salwanizam, N.A.; Muhammad Esham, N.S.A.; Ayob, Q.A.; Ramasamy, R.; Govindan, H.; Md Redzuan, A.; Jasamai, M. Drug-Herb Interactions: Selected Antihypertensive Drugs with Moringa Oleifera Leaves Extract. Jimmy Swaggart Minist. 2022, 51, 1143–1154. [Google Scholar] [CrossRef]
Figure 1. The content of individual vitamin E isomers in M. oleifera oil mg/kg.
Figure 1. The content of individual vitamin E isomers in M. oleifera oil mg/kg.
Life 15 00881 g001
Figure 2. Relative percentage of the presence of the vitamin C identified in the different M. oleifera samples from different.
Figure 2. Relative percentage of the presence of the vitamin C identified in the different M. oleifera samples from different.
Life 15 00881 g002
Figure 3. The content of vitamins in fresh and dried leaves of M. oleifera plant mg/100 g.
Figure 3. The content of vitamins in fresh and dried leaves of M. oleifera plant mg/100 g.
Life 15 00881 g003
Figure 4. Nutrient content in fresh, dried leaves, and extract of M. oleifera [37].
Figure 4. Nutrient content in fresh, dried leaves, and extract of M. oleifera [37].
Life 15 00881 g004
Figure 5. Percentage ratio of mineral content in different morphological organs of the Moringa oleifera [48].
Figure 5. Percentage ratio of mineral content in different morphological organs of the Moringa oleifera [48].
Life 15 00881 g005
Figure 6. Percentage content of flavonoids and phenolic acids in M. oleifera leaf extract from different countries.
Figure 6. Percentage content of flavonoids and phenolic acids in M. oleifera leaf extract from different countries.
Life 15 00881 g006
Figure 7. Compounds in Water extracts of bark, leaves, and pods of M. oleifera.
Figure 7. Compounds in Water extracts of bark, leaves, and pods of M. oleifera.
Life 15 00881 g007
Figure 8. Compounds in Ethanol extracts of bark, leaves, and pods of M. oleifera.
Figure 8. Compounds in Ethanol extracts of bark, leaves, and pods of M. oleifera.
Life 15 00881 g008
Figure 9. Compounds in methanol extracts of bark, leaves, and pods of M. oleifera.
Figure 9. Compounds in methanol extracts of bark, leaves, and pods of M. oleifera.
Life 15 00881 g009
Figure 10. Total phenolic and flavonoid content of different parts of M. oleifera.
Figure 10. Total phenolic and flavonoid content of different parts of M. oleifera.
Life 15 00881 g010
Figure 11. Beneficial effects of the plant M. oleifera on the human body.
Figure 11. Beneficial effects of the plant M. oleifera on the human body.
Life 15 00881 g011
Table 1. Common names of M. oleifera in different countries and languages.
Table 1. Common names of M. oleifera in different countries and languages.
CountryLanguageCommon Name(s)
Arab countriesArabicrawag
Bangladesh/IndiaBengalisujina, sohjna, sajina
East AfricaSwahilimronge, mzunze, mlonge, mrongo
EthiopiaAmharicshiferaw
FranceFrenchacacia blanc, neverdie, moringa ailé, ben ailé, pois quenique
GermanyGermanpferderettichbaum, meerrettichbaum
Haiti/MartiniqueCreolepatois
Hong KongCantonesenuge
IndiaHindisanjna, suhujna, sondna, sohanjna, shajna, munga ara, sainjna, mungna
LaosLao‘ii h’um
Latin AmericaSpanishparaíso blanco, paraíso francés, reseda
MalaysiaMalaysajina, merunggai
MyanmarBurmesedan-da-lun, dandalonbin
NepalNepalishobhanjan, sohijan
NigeriaYorubaewe-igbale
PakistanUrdusahjnao
ThailandThaimakhonkom, ma-rum, phakihum
USA/UK/OtherEnglishmoringa tree, ben-oil tree, cabbage tree, clarifier
VietnamVietnamesechùm ngây
Table 2. Taxonomic classification of M. oleifera Lam.
Table 2. Taxonomic classification of M. oleifera Lam.
Taxonomic RankClassification
Kingdom:Plantae
Subkingdom:Tracheobionta
Superdivision:Spermatophyta
Division:Magnoliophyta
Class:Magnoliopsida
Subclass:Dilleniidae
Order:Capparales
Family:Moringaceae
Genus:Moringa
Species:Oleifera
Table 3. Vitamin content in M. oleifera Lam. leaves.
Table 3. Vitamin content in M. oleifera Lam. leaves.
Vitaminsmg/100 g
Raw LeavesDried LeavesLeaf Powder
A1.283.6316.30
B10.062.022.64
B20.0521.3020.50
C220.0015.8017.30
E448.0010.80113.00
Table 4. Mineral content in M. oleifera Lam. Leaves [47].
Table 4. Mineral content in M. oleifera Lam. Leaves [47].
Mineralsmg/100 g
Fresh LeavesDried LeavesLeaf Powder
Calcium440.002185.002003.00
Potassium259.001236.001324.00
Magnesium42.00448.00368.00
Phosphorus70.00252.00204.00
Iron0.8525.6028.20
Table 5. Distribution of major phytochemical constituents in different parts of M. oleifera.
Table 5. Distribution of major phytochemical constituents in different parts of M. oleifera.
Class CompoundsPlant PartPhytoconstituent
FlavonoidsLeavesApigenin, Apigenin-O-8-glucoside, Apigenin-7-C-glucoside, Astragalin, Daidzein, Genistein, Isoquercitrin, Isorhamnetin, Isorhamnetin 3-O-(6″-malonylglucoside), Kaempferide-3-O-(2″-O-galloyl rhamnoside), Kaempferol, Kaempferol-3-O-β-D-(6″-O-malonyl)-glucoside, Luteolin, Myricetin, Quercetin, Quercetin-3-acetylglucoside, Quercetin-O-3-glucoside, Quercetin-O-3,7-diglucoside, Rutin
FlowersRhamnetin, Isoquercitrin, Kaempferitrin
CarbamatesLeavesNiazinin A, Niazinin B, Niazimicin, Niazimimin A, Niazimimins B, Marumoside A, Marumoside B, Pterygospermin
PodsNiazicin A, Niazidin, Niazinin A, S-Methyl-N-thiocarbamate, Pterygospermin
SeedsO-n-Butyl-4-[(α-l-rhamnopyranosyloxy)benzyl]thiocarbamate, O-Ethyl-4-[(α-L-rhamnopyranosyloxy)-3-hydroxybenzyl]thiocarbamate, N-[4-(β-l-Rhamnopyranosyl)benzyl]-1-O-α-d-glucopyranosyl-thiocarboxamide
Roots1,3-Dibenzyl urea
PhenolicsLeavesSinapic acid, Gentistic acid, Syringic acid, Chlorogenic acid, Cryptochlorogenic acid, 4-O-caffeoyl quinic acid, 5-O-caffeoyl quinic acid, Epicatechin,
SeedsGallic acid, p-Coumaric acid, Ferulic acid, Caffeic acid, Protocatechuic acid, Vanillin, Ellagic acid, Catechin, Moringyne
Stems4-Hydroxymellein, p-Hydroxybenzoic acid, p-Hydroxybenzaldehyde, trans-Ferulic acid, Lasiodiplodin
Rootbarkp-Hydroxybenzaldehyde, De-O-methyllasiodiplodin
GlucosinolatesLeavesNiazirin, Niazirinin
PodsSulforaphane, Methyl-1-aminopentasulfide-5-sulfinate
SeedsNiazirin, Glucomoringin, Glucosinalbin, Glucoraphanin, Glucoiberin, Glucobarbarin
Roots4-O-(α-l-Acetylrhamnopyranosyloxy)-benzyl glucosinolate
Table 6. Antioxidant activity of different parts of M. oleifera using various solvent extracts and assays.
Table 6. Antioxidant activity of different parts of M. oleifera using various solvent extracts and assays.
Plant PartSolvent TypeAssayIC50References
LeavesPetroleum etherDPPH42.56 μg/mL[64]
Ethyl acetateDPPH5.72 μg/mL[64]
EthanolDPPH1.87 mg/mL[56]
EthanolABTS1.36 mg/mL[56]
MethanolDPPH387.00 µg/mL[61]
Crude methanolDPPH35.42 μg/L[64]
Dry leavesMethanolFRAP396.43 μmol TE/g[65]
Dry leavesMethanolORAC3197.24 μmol TE/g[65]
StemsMethanolDPPH1116.00 µg/mL[61]
RootsEthanolDPPH3.31 mg/mL[56]
RootsEthanolABTS1.24 mg/mL[56]
SeedsEthanolABTS40.35 mg/mL[56]
Crude methanolDPPH91.13 μg/mL[64]
Table 7. Summary of in vivo studies on the anti-inflammatory activity of M. oleifera extracts.
Table 7. Summary of in vivo studies on the anti-inflammatory activity of M. oleifera extracts.
Extract TypeAnimal Model/DoseObserved EffectsReferences
Aqueous leaf extract200 mg/kg (rat paw edema)Inhibition of inflammation comparable to ibuprofen (40 mg/kg) using egg albumin-induced edema model[48]
Aqueous leaf extract424 mg/kg (rat paw edema)Similar anti-inflammatory effect to ibuprofen; egg albumin-induced model[81]
95% Ethanolic leaf extract1000 mg/kg (rat paw edema)Reduced carrageenan-induced paw edema by 79% after 5 h, comparable to diclofenac[76]
Methanolic leaf extract250 and 500 mg/kg (guinea pig model)Anti-asthmatic effect; bronchodilation, ↓ WBC count and histamine in lungs (ovalbumin-sensitized)[78]
Table 10. Commercial dietary supplements based on M. oleifera Lam. and their reported applications.
Table 10. Commercial dietary supplements based on M. oleifera Lam. and their reported applications.
ProductManufacturerPlant Part/ExtractReported Purpose(s)Reference
Swanson M. oleiferaSwanson Health Products, USALeaf extractAntioxidant, supports immune function[47]
Yango M. oleiferaYango, PolandLeaf extractAnticancer and neuroprotective effects[47]
Vitama Nature M. oleiferaVitamin Nature, GermanyLeaf extractHematoprotective, antioxidant[193]
Jiva Botanicals M. oleiferaJiva Botanicals, USALeaf extractSupports metabolic function[47]
Natgrown M. oleifera leafNatgrown, USALeaf extractRegulates blood sugar, antibacterial activity[193]
M. oleifera bark extract capsulesHerbal Hills, IndiaBark extractHepatoprotective, anticancer effects[193]
Nature’s Way M. oleifera seedNature’s Way, BulgariaSeed extractAntidiabetic effects[193]
Organic M. oleifera root extractKuli Kuli, CanadaRoot extractAnti-inflammatory properties, immune stimulant[193]
M. oleifera fruit powderGrenera Nutrients, IndiaFruit extractSupports cardiovascular health[194]
Table 11. Selected cosmetic products containing M. oleifera extracts or oil and their reported applications.
Table 11. Selected cosmetic products containing M. oleifera extracts or oil and their reported applications.
Product DescriptionPlant Part UsedManufacturer CountryReported FunctionReference
Wrinkle serumSeedsItalyAnti-wrinkle, smoothing effect[198]
Natural moringa oilSeedsBulgariaHydrates, reduces wrinkles and scars[199]
Micellar waterSeedsFranceMakeup removal, hydration, soothing[200]
Moringa body yogurtSeedsUnited KingdomSkin hydration and softness[201]
Facial cleansing foamSeedsBulgariaSkin purification, pollution removal[202]
Oil body lotionSeedsIndiaMoisturizing and nourishing[203]
Facial cleansing foamLeavesUSAGentle cleansing, hydration support[204]
Anti-aging facial therapyLeavesItalyHydration, firming, and wrinkle reduction[198]
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

Panova, N.; Gerasimova, A.; Gentscheva, G.; Nikolova, S.; Makedonski, L.; Velikova, M.; Beraich, A.; Talhaoui, A.; Petkova, N.; Batovska, D.; et al. Moringa oleifera Lam.: A Nutritional Powerhouse with Multifaceted Pharmacological and Functional Applications. Life 2025, 15, 881. https://doi.org/10.3390/life15060881

AMA Style

Panova N, Gerasimova A, Gentscheva G, Nikolova S, Makedonski L, Velikova M, Beraich A, Talhaoui A, Petkova N, Batovska D, et al. Moringa oleifera Lam.: A Nutritional Powerhouse with Multifaceted Pharmacological and Functional Applications. Life. 2025; 15(6):881. https://doi.org/10.3390/life15060881

Chicago/Turabian Style

Panova, Natalina, Anelia Gerasimova, Galia Gentscheva, Stoyanka Nikolova, Lubomir Makedonski, Margarita Velikova, Abdessamad Beraich, Abdelmonaem Talhaoui, Nadezhda Petkova, Daniela Batovska, and et al. 2025. "Moringa oleifera Lam.: A Nutritional Powerhouse with Multifaceted Pharmacological and Functional Applications" Life 15, no. 6: 881. https://doi.org/10.3390/life15060881

APA Style

Panova, N., Gerasimova, A., Gentscheva, G., Nikolova, S., Makedonski, L., Velikova, M., Beraich, A., Talhaoui, A., Petkova, N., Batovska, D., & Nikolova, K. (2025). Moringa oleifera Lam.: A Nutritional Powerhouse with Multifaceted Pharmacological and Functional Applications. Life, 15(6), 881. https://doi.org/10.3390/life15060881

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