OBM Integrative and Complementary Medicine is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc. It covers all evidence-based scientific studies on integrative, alternative and complementary approaches to improving health and wellness.

Topics contain but are not limited to:

  • Acupuncture
  • Acupressure
  • Acupotomy
  • Bioelectromagnetics applications
  • Pharmacological and biological treatments including their efficacy and safety
  • Diet, nutrition and lifestyle changes
  • Herbal medicine
  • Homeopathy
  • Manual healing methods (e.g., massage, physical therapy)
  • Kinesiology
  • Mind/body interventions
  • Preventive medicine
  • Research in integrative medicine
  • Education in integrative medicine
  • Related policies

The journal publishes a variety of article types: Original Research, Review, Communication, Opinion, Comment, Conference Report, Technical Note, Book Review, etc.

There is no restriction on paper length, provided that the text is concise and comprehensive. Authors should present their results in as much detail as possible, as reviewers are encouraged to emphasize scientific rigor and reproducibility.

Publication Speed (median values for papers published in 2024): Submission to First Decision: 6.8 weeks; Submission to Acceptance: 14.3 weeks; Acceptance to Publication: 6 days (1-2 days of FREE language polishing included)

Open Access Review

A Review on the Ethnomedicinal Plants Used in Zimbabwe for the Treatment and Management of Skin Conditions: Perspectives on Pharmacological and Toxicological Evaluation

Elliot Nyagumbo 1,2 ORCID logo, Trust Nyirenda 3, Cephas Mawere 4, Alfred M. Mutaramutswa 1, Godwins Ngorima 1, Donald T. Kapanga 1, Fabian Maunganidze 3, Marvellous Matsheza 5, William Pote 5, Lucy Mabaya 1, Michael Bhebhe 2,*

  1. National Pathology Research and Diagnostic Centre, Midlands State University, Gweru, Zimbabwe

  2. Department of Biochemistry, Midlands State University, Gweru, Zimbabwe

  3. Department of Physiology, Midlands State University, Gweru, Zimbabwe

  4. Department of Biotechnology, Harare Institute of Technology, Harare, Zimbabwe

  5. Department of Biomedical Sciences, Great Zimbabwe University, Masvingo, Zimbabwe

Correspondence: Michael Bhebhe

Academic Editor: James D. Adams

Special Issue: Evidence-Based Application of Natural Products in the Prevention and Treatment of Diseases

Received: March 05, 2025 | Accepted: July 23, 2025 | Published: August 19, 2025

OBM Integrative and Complementary Medicine 2025, Volume 10, Issue 3, doi:10.21926/obm.icm.2503036

Recommended citation: Nyagumbo E, Nyirenda T, Mawere C, Mutaramutswa AM, Ngorima G, Kapanga DT, Maunganidze F, Matsheza M, Pote W, Mabaya L, Bhebhe M. A Review on the Ethnomedicinal Plants Used in Zimbabwe for the Treatment and Management of Skin Conditions: Perspectives on Pharmacological and Toxicological Evaluation. OBM Integrative and Complementary Medicine 2025; 10(3): 036; doi:10.21926/obm.icm.2503036.

© 2025 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Medicinal plants remain central to healthcare in Africa, where up to 80% of the population relies on traditional remedies to manage a range of health conditions, including skin ailments. In Zimbabwe, the country's rich biodiversity and favourable agroecological zones support a diverse range of ethnomedicinal plants. However, much of the knowledge surrounding these plants remains dispersed and scientifically under-evaluated. This review consolidates evidence from ethnobotanical surveys, peer-reviewed research, and grey literature to identify 93 plant species used in Zimbabwe for the treatment of 21 skin-related conditions, ranging from wounds and ulcers to measles and fungal infections. Most remedies are applied topically, often in powder or paste form, with leaves and roots cited as the most commonly used plant parts. Notably, plant species from the Fabaceae, Asteraceae, and Verbenaceae families featured most frequently. Pharmacological assessments revealed that many of these plants exhibit antimicrobial, anti-inflammatory, and antioxidant properties. Yet, safety data is uneven; 40.9% of species had confirmed low toxicity, 8.6% were associated with high toxicological risks, and the remainder lacked sufficient toxicological profiling. By compiling and critically analyzing this knowledge, the study bridges gaps between traditional practice and biomedical research. It highlights species with potential for further pharmacological validation and underscores the role of indigenous knowledge in informing future dermatological drug discovery. Ultimately, this work contributes to the broader literature on integrative medicine by mapping out a culturally and scientifically relevant repository of medicinal plants used in Zimbabwe.

Graphical abstract

Click to view original image

Keywords

Ethnobotanical; ethnomedicine; pharmacological; toxicology; traditional plants; skin; wounds; Zimbabwe

1. Introduction

Traditional medicine is described by the World Organization as the sum of the knowledge, skills, and practices based on the theories, beliefs, and experiences indigenous to different cultures, whether explicable or not, used in the maintenance of health and the prevention, diagnosis, improvement, or treatment of physical and mental illness [1]. Traditional medicines form an integral part of the cultural heritage in Africa, with approximately 80% of the population on the continent relying on these remedies for their healthcare needs [2]. Modern technology applied to traditional knowledge has played a significant role in deriving at least 40% of all modern medicines directly or indirectly from medicinal plants [3]. Throughout history, traditional medicines have consistently held a crucial position in global healthcare, persistently utilised for addressing a wide range of conditions and ailments.

The skin, which consists of multiple layers such as the epidermis, dermis, and hypodermis, is the body's largest organ. As an organ, it provides protection and acts as a physical barrier against infections [4]. Skin diseases affect about 1.8 billion people in the world [1]. Skin diseases are common in Africa, and the prevalence varies by region and population. Some common skin diseases in Africa include scabies, eczema, acne, and fungal infections [5]. Skin conditions such as cancers, eczema, herpes infection, fungal infection, anti-ageing concerns, itching, insect bites, pemphigus vulgaris, trauma, psoriasis, athlete's foot infections, rashes, skin pigmentation, acne, and primary and minor wound infections are the most prevalent skin disorders [6]. Skin diseases affect individuals of all age groups, but children are particularly vulnerable due to the thinness and delicacy of their skin [7]. The utilisation of natural plant-based medicines for treating skin disorders is prevalent in Zimbabwe and worldwide [2]. This approach is favoured due to several advantages, such as minimal side effects, cost-effectiveness, and longstanding acceptance of its use. Plants contain phytochemical substances that are employed in the treatment of skin disorders.

Phytochemicals consist of primary and secondary metabolites. Secondary metabolites have long been recognised for their bioactivity against various diseases. Several secondary metabolites, such as mangiferin, lutein, curcumin, resveratrol, embelin, naringenin, quercetin, lycopene, gingerol, and apigenin, are utilised for the treatment of skin disorders [4]. Herbal remedies like tea tree oil, Aloe vera, and green tea extract have been used to reduce inflammation and control acne breakouts. Numerous studies have examined medicinal plant extracts for bioactive properties like antioxidants, anti-inflammatory effects, and antimicrobial activities in skin conditions, to validate their efficacy [8]. Traditional healers in Zimbabwe use a plethora of plant species for dermatological issues. A survey in south-central Zimbabwe identified 93 medicinal plant species from 41 families used to treat 18 different categories of diseases, including skin infections. Trees and shrubs are predominantly used, with leaves being the most common plant part applied. Common skin conditions treated include abscesses, acne, burns, boils, ringworm, rashes, shingles, sores, wounds, and warts [9].

The Fabaceae family is notably significant, with about 101 species used traditionally in Zimbabwe for various medicinal purposes, including skin-related issues. For example, species like Indigofera and Senna are used for conditions like wounds and sores [9]. Plants like Hypoxis hemerocallidea and Helichrysum paronychioides have high cultural importance in treating skin diseases [10]. These plants are often used in the form of roots, whole plants, or leaves, prepared through concoctions, macerations, or decoctions.

Aloe excelsa, a plant that is indigenous to Zimbabwe and some parts of Southern Africa, is very effective against skin diseases like wounds. Aloe ecelsa is known for its anti-inflammatory, skin protection, anti-bacterial, anti-viral, antiseptic, and wound healing properties [11]. Although traditional medicine is gaining greater acceptance in Zimbabwe, the valuable indigenous knowledge concerning traditional remedies remains inadequately documented. Proper documentation of the conventional use of medicinal plants in Zimbabwe for treating skin diseases is crucial. Indigenous Knowledge Systems (IKS) documentation and scientific validation play a vital role in safeguarding indigenous knowledge and cultural traditions. IKS offers alternative, complementary, and more accessible treatment options. The ethnopharmacological approach contributes to biodiversity preservation while serving as a foundation for discovering innovative pharmaceuticals and therapeutic strategies based on medicinal plant knowledge.

2. Materials and Methods

2.1 Protocol Development

The Joanna Briggs Institute (JBI) scoping review methodology [12] guided the study design. It followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews (PRISMA-ScR) [13,14]. Here, three members of the research team (CM, AM, and EN) reviewed literature from databases, registers, and websites, as well as books (see Figure 1). The current policy prohibits the registration of scoping reviews on PROSPERO, which is the International Prospective Register of Systematic Reviews. Therefore, this review was not eligible for registration. However, the protocol is accessible from the authors upon request.

Click to view original image

Figure 1 Flowchart for the review of literature [14].

2.2 Identifying the Research Question

The study was guided by the research question: What is the pharmacology and toxicology of medicinal plants traditionally used in Zimbabwe for the management of skin conditions?

2.3 Information Sources

Electronic databases such as Science Direct, PubMed, and Google Scholar were mined for literature on medicinal plants used for the treatment of skin conditions in Zimbabwe. The systematic literature search was done using key terms as shown in the Table S1. Bibliographies of cited studies were also searched, and Google, websites, and organisations such as university libraries were used for unpublished data or grey literature. Three co-authors then screened titles and abstracts and checked full-text articles for eligibility. The ethnobotanical survey was done in the form of interviews and questionnaires with the traditional herbal practitioners.

2.4 Study Selection

Records identified through the search strategy were downloaded as BibTeX files are imported into Zotero. This was followed by the removal of duplicate articles. A CSV file with article data was then exported for records screening. The titles and abstracts of the downloaded articles were thus independently screened by three researchers (CM, AMM, EN) using pre-defined inclusion and exclusion criteria. Where all three researchers agreed, studies were included, while consensus was used to settle disparities. Moreover, the full-text review was completed by the three researchers (CM, AM, EN). In case of uncertainty regarding the eligibility of an article, consensus was also used as a resolution among the three researchers.

2.5 Inclusion Criteria

Peer-reviewed journal articles included in this study covered herbal remedies used for skin conditions with a focus on Zimbabwe. Studies published in English addressing phytochemistry, pharmacology, and toxicology of medicinal plants were included. Articles published between the 1st of January 1990, and the 17th of December 2021, were included in this scoping review. Each paper was to be original research or a protocol.

2.6 Exclusion Criteria

This scoping review excluded studies on veterinary herbal remedies, research involving non-Zimbabwean populations, and records not related to wound healing or skin conditions. Additionally, studies with unavailable full texts, those focusing on unrelated diseases, and unpublished works such as theses and dissertations were omitted. Scoping, systematic, and narrative reviews, as well as studies with insufficient or inconclusive data for analysis, were also excluded.

2.7 Data Charting Process

Relevant variables were extracted using a developed data-charting form. The charted variables were the literature characteristics (family, plant name, growth habit, ethnomedicinal use(s), and parts used as well as distribution), pharmacological activities, and toxicological profiles of each plant.

2.8 Data Analysis

The PRISMA-ScR checklist was used to report the results of the scoping review. A data extraction framework developed by the researchers (EN, CM) was then employed for a deductive analysis. Details generated from the study were the forms and routes of administration of the traditional medicines for the management of skin diseases. A descriptive analysis highlighting the plant families and plant parts used for making herbal medicines, the forms in which the traditional remedies are produced, the family of plants used for the traditional remedy, and the skin conditions the conventional treatments are made to prevent or cure was conducted. A compilation of the names of the plant families studied was also done, and the findings were presented in tables, graphs, and charts.

2.9 Synthesis

The quality or methodology of bias adopted by the included articles was not evaluated in this review [15]. This is so because scoping reviews are not intended to produce a critically appraised and synthesised answer to a specific question. Instead, they merely aim at giving an overview or map of all the evidence. Accordingly, it was not necessary to assess the methodological limitations or bias of evidence included in this scoping review [16].

3. Results and Discussion

3.1 Literature Search

Our search identified 1335 records, which were downloaded and imported into Zotero. About 8 Duplicates and 48 ineligible studies were then removed before screening of records. A comma-separated values (CSV) file was generated for analysis after exporting library data from Zotero. Of the remaining 1279 records, screened titles (n = 683) and abstracts (n = 346) were excluded because they were not specific or relevant to skin conditions or Zimbabwe. Of the 250 reports that were sought for retrieval, 46 could not be retrieved as they were not open source. The remaining 204 studies had 95 review papers, which were then excluded, leaving 109 reports to be included in this scoping review. The authors also considered grey literature from organisations, websites, Google search, and retrieved referenced citations (Total 329 reports) for data on the pharmacology and toxicology profile of medicinal plants used in Zimbabwe for the management of skin conditions. Thus, a total of 438 articles were included in this scoping review. Figure 1 shows the flow diagram of the records screening process.

3.2 Ethnobotanical Surveys of Medicinal Plants Used in Zimbabwe for Treating and Managing Skin Conditions

Ethnobotanical surveys play a crucial role as preliminary screening tools in the exploration of the pharmacological potential of plants. We have identified that Zimbabwe utilises a minimum of 93 plant families for the treatment of skin diseases. This indicates that Zimbabwe has a vast repository of medicinal plants for skin disorders. Bitew [17] conducted a review that identified similar plant species used for treating wounds and skin conditions, including plants found in Ethiopia: Hypoestes forskalei, Solanum incanum, and Ziziphus mucronata. Several other plants have been highlighted by Mabona and Van Vuuren [18] in a review on medicinal plants used for the treatment of skin diseases in Southern Africa.

The different plant families that are mainstream in the treatment and management of skin infections in Zimbabwe are revealed in Figure 2. The Fabaceae, Asteraceae, and Verbenaceae families contain the majority of the medicinal plants that have been used traditionally in Zimbabwe to treat and manage skin conditions. This could be attributed to the greater prevalence and higher number of plant species belonging to those families within the flora of Zimbabwe. From the collected data, the Fabaceae family stands out as the most diverse and abundant plant family in the country, hosting a significant number of prevalent species. Fabaceae is a family of plants that are commonly found in the country as trees or shrubs, including common species such as Bobgunnia madagascariensis and Pterocarpus angolensis.

Click to view original image

Figure 2 Families of medicinal plant species used to treat and manage skin conditions in Zimbabwe.

3.3 Plant Parts Used of Medicinal Plants Used in Zimbabwe for Treating and Managing Skin Conditions

Plant roots are the most commonly used part for the treatment of skin diseases. Although the utilisation of roots for medicinal purposes is the least environmentally sustainable, it remains one of the most preferred sources of medicine. Plant leaves closely follow, maybe as a result of their easy availability, accessibility, convenience in their preparation, and efficacy of their phytoconstituents. However, the primary plant parts utilised for skin treatment are the roots, leaves, and bark. However, with some herbs, a complete plant, fruit, bark, stem, or any other part of the plant is used for the treatment of skin diseases [19]. Based on the data presented in Figure 3, it is evident that there are multiple plant part options available for the treatment of specific skin diseases.

Click to view original image

Figure 3 Plant parts used for medicinal preparations used for the management of skin conditions in Zimbabwe.

3.4 Mode of Preparation and Administration of Medicinal Plants Traditionally Used in Zimbabwe for Treating and Managing Skin Conditions

It is reported that southern Africans have traditionally relied on several plants for wound healing [20,21]. Roots, stems, leaves, and bark are the most commonly used parts of plants for preparing infusions and concoctions. As an example, Boophone disticha, a hallucinogenic plant widely distributed in South Africa and Zimbabwe, was reported to be effective in treating wounds after circumcision in males [21]. There have been records of Ximenia caffra being used to treat wounds. X. caffra leaves were reportedly used topically for wound treatment [22].

Different methods of herbal preparations and applications are shown in Table 1 and Figure 4. The various formulations utilized by local individuals for preparing medicinal plants are powder and soot. Fresh or dried plant materials are used. The most common method of treatment is the use of herbal powder, which is prepared by grinding dried leaves, roots, or bark. The herbal powder is applied topically to the wound or ulcer. Herbal infusions are essentially herbal teas consumed for their antibiotic or analgesic properties. Decoctions are prepared by heating a specific quantity of herbs with water and consumed orally or used to bathe. Decoctions are typically utilized for tougher plant materials, including roots, bark, and seeds. Based on the gathered data, some herbs are also used in the form of soup or ash to treat skin conditions. The resulting liquid can be employed to cleanse wounds, serve as an antiseptic, or be applied to skin rashes. This method is straightforward to prepare, making it one of the most commonly used formulations. Plant soup and soot are the least frequently used for managing skin diseases.

Table 1 Medicinal plants used in Zimbabwe for the treatment and management of skin conditions.

Click to view original image

Figure 4 Mode of preparation of plants used for the treatment and management of skin conditions in Zimbabwe.

Ointments are created by combining finely ground herbal powder with petroleum jelly, oil, or wax. Ointments and plant poultices represent 8% of herbal preparations. Typically, a heated mass of plant material is used as a dressing, applied as either a cold or hot compress for treating superficial skin conditions such as urticaria, skin irritation, or sunburn. In these cases, antipruritic agents, skin-calming substances, or protective barriers like pastes may be utilized to manage the symptoms. Understanding the permeability of compounds in medicinal plants is crucial, as these compounds are expected to exert effects on the skin and should be suitable for their intended therapeutic purposes [18]. Various dressings can include mixtures such as salt, porridge, water (from infusions and decoctions), soup, castor oil, snuff, goat's blood, and other plants like Vigna unguicalata, Dicoma anomala, and Ricinus communis seeds. Preparation methods involve boiling, grinding, burning, cooking, crushing, heating, squeezing, and warming.

External Treatment: This involves rubbing herbal preparations on the body, washing with infusions, squeezing them into lesions, applying them to wounds, and using fibers as bandages or fomentations. Internal Treatment: These herbal remedies can also be taken orally. For instance, plants like Abutilon angulatum, Boophone disticha, Combretum platypetalum, and Leonotis ocymifolia are known to treat burns. Most documented medicinal plants are used topically, a method that is often preferred due to its lower risk of absorption and toxicity [46]. Studies have shown that applying Aloe vera gel twice daily for eight weeks can significantly reduce acne lesions and improve acne severity [47]. However, only a limited number of Zimbabwean medicinal plants have been scientifically researched for treating skin conditions.

This review primarily focuses on medicinal plants, without delving into their use as topical agents for cosmetic purposes, skin-lightening effects, or other traditional applications. These plants are known for their ability to improve skin tone and provide anti-aging, antibacterial, and anti-free radical benefits for aged, photoaged, stressed, and tired skin, offering both protective and restorative effects. While the emphasis here is on medicinal plants, further research into their cultural applications as topical agents should not be overlooked, as they play a significant role in traditional skincare practices.

3.5 Common Skin Conditions Treated Traditionally by Medicinal Plants in Zimbabwe

Traditionally, herbal remedies have been employed in Africa to treat and manage a diverse array of illnesses. Onayade et al. [48] described the use of plants in first aid, wound cleansing, wound washing, and pus extraction, as well as for infected and festering wounds. In addition, plants treat boils, abscesses, cuts, skin lesions, wounds, snake bites, insect bites, bruises, pains, ulcers, fractures, trauma, sprains, aches, suppurations, inflammations, scabies, rabies, and dress wounds. Some clinical practices used in wound management can be compared to those in African traditional medicine and Chinese traditional medicine, such as bone setting, fracture management, uvulectomy, abdominal surgery, trephination, and circumcision [49].

From our investigations, Table 1 presents a compilation of plants that have been and are currently being used in Zimbabwe for the treatment of different types of skin conditions. It has been reported that most plants used for dermatological purposes are related to wound healing as well [18]. This review found that 21 skin conditions in Zimbabwe are being treated using plant-based medicine (Figure 5). Zimbabwean traditional medical practitioners have utilized these herbal remedies for first aid, wound cleansing, pus extraction, and treatment of infected and festering wounds. They are also employed in the management of boils, abscesses, cuts, skin lesions, snake and dog bites, insect stings, bruises, pain relief, burn soothing, ulcers, fractures, sprains, aches, suppurations, inflammations, scabies, rabies, and wound dressing. Among the various skin conditions, wounds were found to be the most frequently treated using herbal remedies, followed by measles and tropical ulcers. On the other hand, skin cancers and tumors were observed to be the least treated diseases with herbs. Aloe vera has been observed to expedite the healing process in cases of chronic leg ulcers, surgically induced wounds, and frostbite. The data indicate that one specific plant is used in the treatment of various skin disorders. For example, burns can be treated using Abutilon angulatum, Boophone disticha, Combretum platypetalum, and Leonotis ocymifolia, among other plants.

Click to view original image

Figure 5 Common skin conditions treated traditionally by medicinal plants in Zimbabwe.

3.6 Pharmacological Properties of Medicinal Plants Used in Zimbabwe for Treating and Managing Skin Conditions

The plants in Table 2 exhibit diverse activities and mechanisms of action, contributing to their effectiveness in treating and managing skin conditions. They possess cicatrizant, antiseptic, antifungal, antiviral, antibacterial, antioxidant, antipyretic, anesthetic, analgesic, hemostatic, antimicrobial, anti-inflammatory, growth-promoting, and collagen synthesis/fibroblast formation-enhancing properties [48]. Some plants aid in wound closure, with or without scar formation, while others stimulate glycosaminoglycan synthesis.

Table 2 Pharmacological and toxicological assessment in terms of LD50 (Lethal Dose 50%) and LC50 (Lethal Concentration 50%) of medicinal plants used in Zimbabwe for treating and managing skin conditions.

The mechanism through which these plants speed up the process of various skin conditions varies from plant to plant. The presented data represent a crucial source of traditional knowledge, especially regarding the use of herbal remedies in Africa for treating and managing skin conditions. Traditional knowledge in rural Zimbabwe identifies readily available plants with a historical record of treating skin disorders. However, scientific research on these plants remains limited. Herbal extracts from these plants likely contain diverse molecules with various functions relevant to managing and treating skin conditions. These functions include signaling, lubrication, proliferation support, wound contraction, and provision of cofactors, antioxidants, radical scavenging properties, and essential nutrients [451]. Furthermore, studies suggest the presence of key pharmacological properties like antimicrobial activity, radical scavenging, and antioxidant effects [134], which may help manage and treat skin conditions.

Pharmacological interaction studies of plants that are used in combination with other plants to treat skin infections lack thorough scientific research. Various cultures have recognized the therapeutic benefits of synergistic interactions, and traditional African healing systems often employ combination therapy based on this principle, with a belief that it enhances efficacy. However, ethnopharmacological information regarding these combinations usually lacks adequate scientific validation [18]. Emerging preclinical evidence by Hamza et al. [162] has shown the promising therapeutic potential of Combretum molle, a plant whose extracts exhibit significant potential in wound healing and ulcer repair, particularly in diabetic foot models and deep wound ulcers. In vivo wound healing progression documented through serial photographic captures demonstrates the extract's promising ability for skin regeneration, restoring healthy appearance, combating damaging effects, and stimulating antioxidant enzyme regeneration, highlighting its potential for therapeutic applications [162].

The pharmacological properties of medicinal plants used in Zimbabwe for skin conditions reveal a wide range of benefits, including antimicrobial, anti-inflammatory, antioxidant, and wound healing effects. Many species demonstrate multiple activities, enhancing their therapeutic potential. For example, Psorospernum febrifugum is recognized for its anti-psoriatic and anti-acne effects. At the same time, Heteromorpha arborescens is noted for its anti-scabies properties, and several other plants are reported to promote wound healing. Additionally, plants such as Citrullus lanatus, Sansevieria hyacinthoides, Schotia brachypetala, Ximenia caffra, and Zea mays are known for their anti-aging and skin regeneration benefits. This diversity reflects the rich biodiversity inherent in traditional medicinal practices across Zimbabwe, underscoring the vital role these plants play in health promotion and the effective management of skin conditions. This overview emphasizes their significant contribution to local healthcare.

3.7 Toxicological Evaluation of Medicinal Plants Used in Zimbabwe for Treating and Managing Skin Conditions

Basing our understanding of their potential therapeutic effects on traditional knowledge passed down through generations regarding medicinal plants has informed our understanding of their potential therapeutic effects. However, relying solely on this knowledge to assume the safety and absence of toxicity in these plants is not scientifically verified. Recent scientific studies have revealed the toxic, mutagenic, and carcinogenic properties of various plant species traditionally used for medicinal purposes. Despite their applications in treating skin conditions, these medicinal plants have also been documented to induce adverse effects, including allergic reactions, phytodermatitis, and an increased risk of photosensitization. A review of the evidence bases for botanicals in dermatology conducted by Reuter et al. [452] identified several traditionally used medicinal plants with documented reports of associated toxic effects, with the toxicity evaluations depicted in Table 3.

Table 3 Toxicological evaluation of medicinal plants used by local people in Zimbabwe to treat and manage skin conditions.

A scientific evaluation of the toxicological profiles of 93 Zimbabwean plants traditionally used for treating skin conditions was conducted. The analysis revealed that 58 plants (approximately 62.4%) have documented toxicological studies, while the remaining 35 plants (37.6%) lack such studies.

According to the assessment, medicinal extracts from the plants with published studies were evaluated for their effects on liver cells, genotoxicity, sub-acute toxicity, cytotoxic activity on human monocyte cells, and anticancer properties. To assess the toxic effects of the plants, laboratory tests were conducted using the Ames test (an in vitro test of bacterial and mammalian cells), the micronucleus test (chromosomes of white blood cells), and the comet test (DNA damage). Additionally, other toxicological effects of the plants were studied using the brine shrimp lethality test (BSLT) and the rodent acute toxicity test (RTA). As a result, Munodawafa et al. [75] suggested that these tests provide accurate, cost-effective, and simple assessments of herbal extract safety.

It is determined by the rodent acute toxicity test, also known as the Lethality Dose (LD50) test. LD50 is the dose of a substance (usually in mg/kg of body weight) that causes death in 50% of test animals (typically rats or mice) within a specified time (often 24 hours to 14 days). It is used to assess acute toxicity (short-term poisoning potential) of chemicals, drugs, or plant extracts. Malebo et al. [453] established toxicity classifications based on dosage (mg/kg body weight). Substance is considered highly toxic when its dosage is <50 mg/kg body weight, toxic at 50-300 mg/kg, moderately toxic at 300-1000 mg/kg, mildly toxic: 1000-2000 mg/kg, non-toxic: 2000-5000 mg/kg body weight. LC50 (Lethal Concentration 50%) is the concentration of a substance (usually in µg/mL or ppm) in air, water, or another medium that kills 50% of test organisms for example fish, insects, or cell cultures over a set exposure period. Bussmann et al. [317] and Erhabor et al. [318] indicated that substances with LC50 values less than or equal to 249 µg/mL are highly toxic, those between 250 and 499 µg/mL are moderately toxic, those between 500 and 999 µg/mL are weak or low, and those above 1000 µg/mL are considered safe.

3.8 Phytochemical Evaluation of Medicinal Plants Used in Zimbabwe for Treating and Managing Skin Conditions

Phytochemicals exhibit a wide range of biological activities that contribute to skin health, including free radical scavenging, inhibition of radical chain reactions, metal chelation, and modulation of oxidative enzymes. Additionally, they serve as cofactors for antioxidant enzymes, enhancing the skin’s defense mechanisms against oxidative stress. Studies have demonstrated that plant extracts can stimulate endogenous antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX), which play a crucial role in mitigating reactive oxygen species (ROS)-induced damage [454]. Plant-derived antioxidants, including polyphenols, tocopherols, carotenoids, and ascorbic acid, effectively neutralize free radicals, thereby promoting skin health [455]. Furthermore, research by Mabona and Van Vuuren [18] highlights the traditional use of Southern African medicinal plants in treating skin disorders, attributing their efficacy to bioactive compounds such as flavonoids, alkaloids, saponins, and phenolics, which possess anti-inflammatory, antimicrobial, and wound-healing properties. The therapeutic potential of herbal medicines in dermatology is well-documented, with numerous studies underscoring their effectiveness in managing skin conditions.

Vitale et al. [456] and Shubayr [457] emphasize that phytochemicals like flavonoids, alkaloids, and saponins enhance collagen synthesis, cell proliferation, and angiogenesis, accelerating wound repair. Among the most studied botanicals, Aloe spp. Contain over 75 bioactive compounds, including vitamins (A, C, E, B12), enzymes (amylase, catalase, peroxidase), minerals (zinc, copper, selenium), polysaccharides (glucomannans, polymannans), and polyphenols (anthraquinones), all of which contribute to skin hydration, regeneration, and anti-inflammatory effects [456,458]. Notably, Aloe polysaccharides show significant promise in tissue engineering due to their ability to promote cell proliferation and angiogenesis, making them valuable for regenerative medicine [459].

Psidium guajava is rich in photoprotective phenolics like flavonoids and tannins, without containing photosensitizing coumarins, making them suitable for sun protection [460]. Similarly, Zingiber officinale contains anthocyanins, which exhibit potent antioxidant and analgesic effects by inhibiting cyclooxygenases (COXs), lipoxygenases (LOXs), and nitric oxide synthase, thereby reducing skin inflammation and pain [461].

The root extract of Kigelia africana promotes wound healing through its glycosides (antimicrobial), phenolics/tannins (coagulation, antioxidant), and saponins (tissue repair). These compounds synergistically enhance inflammation control, cellular proliferation, collagen formation, and microbial defense, accelerating wound closure while preventing infection and oxidative damage, demonstrating their therapeutic potential in wound care [462,463].

Venkataravana et al. [464] study identified key phytocompounds in Citrullus lanatus (flavonoids, terpenes, lycopene, resveratrol, lignans, tannins, and indoles) via GC-MS analysis. These compounds exhibit antioxidant, anti-inflammatory, and antimicrobial properties that support wound healing by enhancing angiogenesis, cell proliferation, and tissue remodeling, while also benefiting skin regeneration and managing inflammatory skin conditions [464].

Despite the promising pharmacological properties of these phytochemicals, further research is necessary to fully explore their mechanisms of action and validate their efficacy and safety in treating skin conditions. Given the complexity of plant-derived compounds, a thorough correlation between their phytochemistry and dermatological effects is essential. Therefore, consultation with healthcare professionals before using herbal remedies is strongly advised to ensure safe and practical application.

4. Conclusions

This review provides the first comprehensive synthesis of medicinal plants traditionally used in Zimbabwe for the treatment and management of skin conditions, drawing on both ethnobotanical data and pharmacological evidence. By identifying 93 plant species across diverse botanical families, the study reveals a rich, yet underexplored, pharmacopoeia rooted in indigenous knowledge systems. The findings underscore the significant role of traditional medicine in primary healthcare and emphasize the need for formal scientific validation of these plant-based therapies. In particular, the pharmacological profiles of many species demonstrating antimicrobial, anti-inflammatory, antioxidant, and wound-healing properties highlight their potential for developing affordable, culturally accepted treatments for common and chronic skin conditions. At the same time, toxicological assessments signal the importance of rigorous safety evaluation before wider clinical use. By consolidating scattered data and identifying priority species for further research, this study contributes meaningfully to the global discourse on ethnopharmacology and integrative dermatology. It also supports broader calls to preserve and promote indigenous health knowledge while guiding future pharmaceutical discovery efforts in Africa and beyond.

Acknowledgments

We acknowledge that the Midlands State University National Pathology Research and Diagnostic Centre provided technical support for this work.

Author Contributions

Elliot Nyagumbo, Trust Nyirenda, Cephas Mawere, Marvellous Matsheza: Conceptualization, writing – original draft, formal analysis, writing – review and editing. Elliot Nyagumbo, Cephas Mawere, Alfred M. Mutaramutswa, Donald T. Kapanga: Software, writing – review and editing. Elliot Nyagumbo, Cephas Mawere, Godwins Ngorima, Alfred M. Mutaramutswa: Methodology, writing – review and editing. Fabian Maunganidze, William Pote, Michael Bhebhe, Lucy Mabaya: Writing – original draft, writing – review and editing. All authors have read and approved the published version of the manuscript.

Funding

No funding.

Competing Interests

The authors have declared that no competing interests exist.

Data Availability Statement

The data supporting this systematic review are from previously reported studies and datasets, which have been cited. The processed data are available from the corresponding author upon request.

Additional Materials

The following additional materials are uploaded at the page of this paper.

  1. Table S1: Detailed search strategy for three database searches.

References

  1. World Health Organization. WHO’s first global meeting on skin NTDs calls for greater efforts to address their burden [Internet]. Geneva: World Health Organization; 2023. Available from: https://www.who.int/news/item/31-03-2023-who-first-global-meeting-on-skin-ntds-calls-for-greater-efforts-to-address-their-burden.
  2. Booth Z. Traditional medicines should be used in healthcare [Internet]. Johannesburg, South Africa: University of the Witwatersrand; 2023 [cited date 2024 January 9]. Available from: https://www.wits.ac.za/news/latest-news/opinion/2023/2023-08/traditional-medicines-should-be-used-in-healthcare.html.
  3. World Health Organization. Traditional medicine [Internet]. Geneva: World Health Organization; 2023. Available from: https://www.who.int/news-room/questions-and-answers/item/traditional-medicine.
  4. Mohd Zaid NA, Sekar M, Bonam SR, Gan SH, Lum PT, Begum MY, et al. Promising natural products in new drug design, development, and therapy for skin disorders: An overview of scientific evidence and understanding their mechanism of action. Drug Des Dev Ther. 2023; 16: 23-66. [CrossRef] [Google scholar] [PubMed]
  5. Namutebi F. P34 Changing patterns of skin diseases in patients attending a tertiary private hospital in central Uganda. Br J Dermatol. 2023; 188: ljad113-062. [CrossRef] [Google scholar]
  6. Ahuja A, Gupta J, Gupta R. Miracles of herbal phytomedicines in treatment of skin disorders: Natural healthcare perspective. Infect Disord Drug Targets. 2021; 21: 328-338. [CrossRef] [Google scholar] [PubMed]
  7. Mengist Dessie A, Fenta Feleke S, Getaye Workie S, Getinet Abebe T, Mossu Chanie Y, Kassa Yalew A. Prevalence of skin disease and its associated factors among primary schoolchildren: A cross-sectional study from a northern Ethiopian town. Clin Cosmet Investig Dermatol. 2022; 15: 791-801. [CrossRef] [Google scholar] [PubMed]
  8. Ghassemi M, Sadeghzadeh‑Bazargan A, Behrangi E, Roohaninasab M, Goodarzi A, Nobari NN. A review on the role of medicinal plants in treatment of acne. Prensa Med Argent. 2021; 107: 295. doi: 10.47275/0032-745X-295. [Google scholar]
  9. Maroyi A. Medicinal uses of the Fabaceae family in Zimbabwe: A review. Plants. 2023; 12: 1255. [CrossRef] [Google scholar] [PubMed]
  10. Asong JA, Ndhlovu PT, Khosana NS, Aremu AO, Otang-Mbeng W. Medicinal plants used for skin-related diseases among the Batswanas in Ngaka Modiri Molema District Municipality, South Africa. S Afr J Bot. 2019; 126: 11-20. [CrossRef] [Google scholar]
  11. Coopoosamy RM. Isolation of volatile compounds of Aloe excelsa (Berger). Afr J Biotechnol. 2010; 9: 7289-7294. [Google scholar]
  12. Peters MD, Marnie C, Tricco AC, Pollock D, Munn Z, Alexander L, et al. Updated methodological guidance for the conduct of scoping reviews. JBI Evid Synth. 2020; 18: 2119-2126. [CrossRef] [Google scholar] [PubMed]
  13. Tricco AC, Lillie E, Zarin W, O'Brien KK, Colquhoun H, Levac D, et al. PRISMA extension for scoping reviews (PRISMA-ScR): Checklist and explanation. Ann Intern Med. 2018; 169: 467-473. [CrossRef] [Google scholar] [PubMed]
  14. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 2021; 372: n71. [CrossRef] [Google scholar] [PubMed]
  15. Tricco AC, Lillie E, Zarin W, O’brien K, Colquhoun H, Kastner M, et al. A scoping review on the conduct and reporting of scoping reviews. BMC Med Res Methodol. 2016; 16: 15. [CrossRef] [Google scholar] [PubMed]
  16. Peters MD, Godfrey CM, Khalil H, McInerney P, Parker D, Soares CB. Guidance for conducting systematic scoping reviews. JBI Evid Implement. 2015; 13: 141-146. [CrossRef] [Google scholar] [PubMed]
  17. Bitew H, Gebregergs H, Tuem KB, Yeshak MY. Ethiopian medicinal plants traditionally used for wound treatment: A systematic review. Ethiop J Health Dev. 2019; 33: 102-127. [Google scholar]
  18. Mabona U, Van Vuuren SF. Southern African medicinal plants used to treat skin diseases. S Afr J Bot. 2013; 87: 175-193. [CrossRef] [Google scholar]
  19. Bhat JA, Kumar M, Bussmann RW. Ecological status and traditional knowledge of medicinal plants in Kedarnath Wildlife Sanctuary of Garhwal Himalaya, India. J Ethnobiol Ethnomed. 2013; 9: 1. [CrossRef] [Google scholar] [PubMed]
  20. Chingwaru C, Bagar T, Maroyi A, Kapewangolo PT, Chingwaru W. Wound healing potential of selected Southern African medicinal plants: A review. J Herb Med. 2019; 17: 100263. [CrossRef] [Google scholar]
  21. Van Wyk B, Gericke N. People’s Plants. A Guide to Useful Plants of Southern Africa. 3rd ed. Pretoria, South Africa: Briza Publications; 2007. [Google scholar]
  22. Maroyi A. Ximenia caffra Sond. (Ximeniaceae) in sub-Saharan Africa: A synthesis and review of its medicinal potential. J Ethnopharmacol. 2016; 184: 81-100. [CrossRef] [Google scholar] [PubMed]
  23. Gelfand M. The traditional medical practitioner in Zimbabwe: His principles of practice and pharmacopoeia. Gweru, Zimbabwe: Mambo Press; 1985. [Google scholar]
  24. Matowa PR, Gundidza M, Gwanzura L, Nhachi CF. A survey of ethnomedicinal plants used to treat cancer by traditional medicine practitioners in Zimbabwe. BMC Complement Med Ther. 2020; 20: 278. [CrossRef] [Google scholar] [PubMed]
  25. Maroyi A. Ethnobotanical study of two threatened medicinal plants in Zimbabwe. Int J Biodivers Sci Manag. 2008; 4: 148-153. [CrossRef] [Google scholar]
  26. Chimponda T, Mukanganyama S. Antimycobacterial activities of selected medicinal plants from Zimbabwe against Mycobacterium aurum and Corynebacterium glutamicum. Trop Biomed. 2010; 27: 595-610. [Google scholar]
  27. Mangoyi R, Chitemerere T, Chimponda T, Chirisa E, Mukanganyama S. Multiple anti-infective properties of selected plant species from Zimbabwe. In: Novel plant bioresources: Applications in food, medicine and cosmetics. John Wiley & Sons, Inc.; 2014. pp. 179-190. [CrossRef] [Google scholar]
  28. Shumba EM. Traditional medicinal plant practice in southern Africa: A situational analysis in Zambia and Zimbabwe. Harare, Zimbabwe: WWF-World Wide Fund for Nature; 2009. [Google scholar]
  29. Maroyi A. An ethnobotanical survey of medicinal plants used by the people in Nhema communal area, Zimbabwe. J Ethnopharmacol. 2011; 136: 347-354. [CrossRef] [Google scholar] [PubMed]
  30. Watt JM, Breyer-Brandwijk MG. The medicinal and poisonous plants of Southern and Eastern Africa. Edinburgh, Scotland: E. and S. Livingstone Ltd.; 1962. [Google scholar]
  31. Chinemana F, Drummond RB, Mavi S, De Zoysa I. Indigenous plant remedies in Zimbabwe. J Ethnopharmacol. 1985; 14: 159-172. [CrossRef] [Google scholar] [PubMed]
  32. Chavunduka GL. Traditional healers and the Shona patient. Gwelo, Zimbabwe: Mambo Press; 1976. [Google scholar]
  33. Rodgers CB, Verotta L. Chemistry and biological properties of the African Combretaceae. In: Chemistry, biological and pharmacological properties of African medicinal plants. Harare, Zimbabwe: UZ Publications; 1996. pp. 121-142. [Google scholar]
  34. Viol DI. Screening of traditional medicinal plants from Zimbabwe for photochemistry, antioxidant, antimicrobial, antiviral and toxicological activities. Harare, Zimbabwe: University of Zimbabwe; 2009. [Google scholar]
  35. Viol DI, Chagonda LS, Moyo SR, Mericli AH. Toxicity and antiviral activities of some medicinal plants used by traditional medical practitioners in Zimbabwe. Am J Plant Sci. 2016; 7: 1538. [CrossRef] [Google scholar]
  36. Marekerah L. A survey on the biological activities of selected plants used to manage diarrhoea and cancer in Vumba, Zimbabwe. Gweru, Zimbabwe: Midlands State University; 2015. [Google scholar]
  37. Maroyi AL. Dicoma anomala sond.: A review of its botany, ethnomedicine, phytochemistry and pharmacology. Asian J Pharm Clin Res. 2018; 11: 70-77. [CrossRef] [Google scholar]
  38. Maroyi A. Medicinal uses, biological and chemical properties of wild grape (Lannea edulis): An indigenous fruit plant of tropical Africa. Asian J Pharm Clin Res. 2019; 12: 16-20. [CrossRef] [Google scholar]
  39. Maroyi A. Traditional use of medicinal plants in south-central Zimbabwe: Review and perspectives. J Ethnobiol Ethnomed. 2013; 9: 31. [CrossRef] [Google scholar] [PubMed]
  40. Wild H, Gelfand M. Some native herbal remedies at present in use in Mashonaland. Cent Afr J Med. 1959; 5: 292-305. [Google scholar]
  41. Kambizi L, Afolayan AJ. An ethnobotanical study of plants used for the treatment of sexually transmitted diseases (njovhera) in Guruve District, Zimbabwe. J Ethnopharmacol. 2001; 77: 5-9. [CrossRef] [Google scholar] [PubMed]
  42. Nyenya RT, Stedje B. Ethnobotanical studies in the genus Sansevieria Thunb. (Asparagaceae) in Zimbabwe. Ethnobot Res Appl. 2011; 9: 421-443. [CrossRef] [Google scholar]
  43. Maroyi A. Sansevieria hyacinthoides (L.) Druce: A review of its botany, medicinal uses, phytochemistry, and biological activities. Asian J Pharm Clin Res. 2019; 12: 21-26. [CrossRef] [Google scholar]
  44. Maroyi A. Exotic plants in indigenous pharmacopoeia of south-central Zimbabwe: Traditional knowledge of herbal medicines. Res J Bot. 2017; 12: 46-52. [CrossRef] [Google scholar]
  45. Mongalo NI, McGaw LJ, Segapelo TV, Finnie JF, Van Staden J. Ethnobotany, phytochemistry, toxicology and pharmacological properties of Terminalia sericea Burch. ex DC. (Combretaceae)–A review. J Ethnopharmacol. 2016; 194: 789-802. [CrossRef] [Google scholar] [PubMed]
  46. Lipsky BA, Hoey C. Topical antimicrobial therapy for treating chronic wounds. Clin Infect Dis. 2009; 49: 1541-1549. [CrossRef] [Google scholar] [PubMed]
  47. Hajheydari Z, Saeedi M, Morteza-Semnani K, Soltani A. Effect of Aloe vera topical gel combined with tretinoin in treatment of mild and moderate acne vulgaris: A randomized, double-blind, prospective trial. J Dermatol Treat. 2014; 25: 123-129. [CrossRef] [Google scholar] [PubMed]
  48. Onayade OA, Onayade AA, Sofowora A. Wound healing with plants: The African perspective. In: Chemistry, biological and pharmacological properties of African medicinal plants. Harare, Zimbabwe: University of Zimbabwe Publications; 1996. pp. 77-120. [Google scholar]
  49. Sofowora A. Medicinal Plants and Traditional Medicine in Africa. Chichester, UK: John Wiley and Sons; 1982. [Google scholar]
  50. Gradé JT, Arble BL, Weladji RB, Van Damme P. Anthelmintic efficacy and dose determination of Albizia anthelmintica against gastrointestinal nematodes in naturally infected Ugandan sheep. Vet Parasitol. 2008; 157: 267-274. [CrossRef] [Google scholar] [PubMed]
  51. Muthee JK, Gakuya DW, Mbaria JM, Mulei CM. Phytochemical screening and cytotoxicity of selected plants used as anthelmintics in Loitoktok Sub-County, Kenya. J Phytopharmacol. 2016; 5: 15-19. [CrossRef] [Google scholar]
  52. Wale K, Kwape TE, Sethibe L, Gaobotse G, Loeto D, Sethebe B. Antibacterial and antioxidant potential of Albizia anthelmintica as a medicinal plant on pathogenic veterinary isolates. J Med Plants Res. 2018; 12: 456-462. [CrossRef] [Google scholar]
  53. Sobeh M, Rezq S, Sabry OM, Abdelfattah MA, El Raey MA, El-Kashak WA, et al. Albizia anthelmintica: HPLC-MS/MS profiling and in vivo anti-inflammatory, pain killing and antipyretic activities of its leaf extract. Biomed Pharmacother. 2019; 115: 108882. [CrossRef] [Google scholar] [PubMed]
  54. He Y, Wang Q, Ye Y, Liu Z, Sun H. The ethnopharmacology, phytochemistry, pharmacology and toxicology of genus Albizia: A review. J Ethnopharmacol. 2020; 257: 112677. [CrossRef] [Google scholar] [PubMed]
  55. Nabil M, El Raey MA, Abdo W, Abdelfattah MA, El-Shazly AM, Sobeh M, et al. Gastro-protective effects of Albizia anthelmintica leaf extract on indomethacin-induced gastric ulcer in wistar rats: In silico and in vivo Studies. Antioxidants. 2021; 10: 176. [CrossRef] [Google scholar] [PubMed]
  56. Somova LI, Shode FO, Moodley K, Govender Y. Cardiovascular and diuretic activity of kaurene derivatives of Xylopia aethiopica and Alepidea amatymbica. J Ethnopharmacol. 2001; 77: 165-174. [CrossRef] [Google scholar] [PubMed]
  57. Clarkson C, Maharaj VJ, Crouch NR, Grace OM, Pillay P, Matsabisa MG, et al. In vitro antiplasmodial activity of medicinal plants native to or naturalised in South Africa. J Ethnopharmacol. 2004; 92: 177-191. [CrossRef] [Google scholar] [PubMed]
  58. Mulaudzi RB, Ndhlala AR, Finnie JF, Van Staden J. Antimicrobial, anti-inflammatory and genotoxicity activity of Alepidea amatymbica and Alepidea natalensis (Apiaceae). S Afr J Bot. 2009; 75: 584-587. [CrossRef] [Google scholar]
  59. Louvel S, Moodley N, Seibert I, Steenkamp P, Nthambeleni R, Vidal V, et al. Identification of compounds from the plant species Alepidea amatymbica active against HIV. S Afr J Bot. 2013; 86: 9-14. [CrossRef] [Google scholar]
  60. Wintola OA, Afolayan AJ. Alepidea amatymbica Eckl. & Zeyh.: A review of its traditional uses, phytochemistry, pharmacology, and toxicology. Evid Based Complementary Altern Med. 2014; 2014: 284517. [CrossRef] [Google scholar] [PubMed]
  61. Mangoale RM, Afolayan AJ. Comparative phytochemical constituents and antioxidant activity of wild and cultivated Alepidea amatymbica Eckl & Zeyh. BioMed Res Int. 2020; 2020: 5808624. [CrossRef] [Google scholar] [PubMed]
  62. Steenkamp V, Fernandes AC, Jansen van Rensburg CE. Antibacterial activity of Venda medicinal plants. Fitoterapia. 2007; 78: 561-564. [CrossRef] [Google scholar] [PubMed]
  63. Mukherjee PK, Nema NK, Maity N, Mukherjee K, Harwansh RK. Phytochemical and therapeutic profile of Aloe vera. J Nat Remedies. 2013; 14: 1-26. [Google scholar]
  64. Hamidi MR, Jovanova B, Panovska TK. Toxicological evaluation of the plant products using Brine Shrimp (Artemia salina L.) model. Maced Pharm Bull. 2014; 60: 9-18. [CrossRef] [Google scholar]
  65. Al-Snafi AE. The pharmacological importance of Aloe vera-A review. Int J Phytopharm Res. 2015; 6: 28-33. [Google scholar]
  66. Taukoorah U, Mahomoodally MF. Crude Aloe vera gel shows antioxidant propensities and inhibits pancreatic lipase and glucose movement in vitro. Adv Pharmacol Pharm Sci. 2016; 2016: 3720850. [CrossRef] [Google scholar] [PubMed]
  67. Guo X, Mei N. Aloe vera: A review of toxicity and adverse clinical effects. J Environ Sci Health C. 2016; 34: 77-96. [CrossRef] [Google scholar] [PubMed]
  68. Haesler E. Evidence summary: Wound management low resource communities-Aloe vera for wound healing. Wound Pract Res. 2017; 25: 115-117. [Google scholar]
  69. Namunana S, Lutoti S, Nyamaizi G, Agaba G, Apun I, Ssebunnya C, et al. Formulation, development and validation of a wound healing herbal ointment from extracts of Bidens pilosa and Aloe barbadensis. J Pharm Pharmacol Res. 2018; 2: 32-38. [CrossRef] [Google scholar]
  70. Hęś M, Dziedzic K, Górecka D, Jędrusek-Golińska A, Gujska E. Aloe vera (L.) Webb.: Natural sources of antioxidants–A review. Plant Foods Hum Nutr. 2019; 74: 255-265. [CrossRef] [Google scholar] [PubMed]
  71. Añibarro-Ortega M, Pinela J, Barros L, Ćirić A, Silva SP, Coelho E, et al. Compositional features and bioactive properties of Aloe vera leaf (fillet, mucilage, and rind) and flower. Antioxidants. 2019; 8: 444. [CrossRef] [Google scholar] [PubMed]
  72. Sánchez M, González-Burgos E, Iglesias I, Gómez-Serranillos MP. Pharmacological update properties of Aloe vera and its major active constituents. Molecules. 2020; 25: 1324. [CrossRef] [Google scholar] [PubMed]
  73. Maroyi A. Annona stenophylla Engl. & Diels: Review of its botany, medicinal uses and biological activities. J Pharm Sci Res. 2019; 11: 3385-3390. [Google scholar]
  74. Okello OO, David N, Oloro J. Anti-fertility activity of aqueous root bark extracts of Asparagus africanus Lam and Annona senegalensis Pers combination on female Sprague Dawley rats. Afr J Pharm Pharmaco. 2019; 13: 12-16. [CrossRef] [Google scholar]
  75. Munodawafa T, Moyo S, Chipurura B, Chagonda L. Brine shrimp lethality bioassay of some selected Zimbabwean traditional medicinal plants. Int J Phytopharmacol. 2016; 7: 229-232. [Google scholar]
  76. Hassan HS, Ahmadu AA, Hassan AS. Analgesic and anti-inflammatory activities of Asparagus africanus root extract. Afr J Tradit Complement Altern Med. 2008; 5: 27-31. [CrossRef] [Google scholar] [PubMed]
  77. Okolie OD. An evaluation of the anti-diabetic properties Asparagus Africanus Lam. root extracts. Bloemfontein, South Africa: Central University of Technology, Free State; 2014. [Google scholar]
  78. Kebede S, Afework M, Debella A, Ergete W, Makonnen E. Toxicological study of the butanol fractionated root extract of Asparagus africanus Lam., on some blood parameter and histopathology of liver and kidney in mice. BMC Res Notes. 2016; 9: 49. [CrossRef] [Google scholar] [PubMed]
  79. El-Ishaq A, Alshawsh MA, Chik ZB. Evaluating the oestrogenic activities of aqueous root extract of Asparagus africanus Lam in female Sprague-Dawley rats and its phytochemical screening using Gas Chromatography-Mass Spectrometry (GC/MS). PeerJ. 2019; 7: e7254. [CrossRef] [Google scholar] [PubMed]
  80. El-Ishaq A, Alshawsh MA, Mun KS, Chik Z. Biochemical and toxicological effects of methanolic extract of Asparagus africanus Lam in Sprague-Dawley rats. PeerJ. 2020; 8: e9138. [CrossRef] [Google scholar] [PubMed]
  81. Elisabeth T, Esther N, Elisabeth NB. Sedative and hypnotic effects of the roots of Asparagus africanus (Asparagaceae) decoction on white mice (Mus musculus Swiss). GSC Biol Pharm Sci. 2020; 11: 254-260. [CrossRef] [Google scholar]
  82. Pereira RL, Ibrahim T, Lucchetti L, da Silva AJ, de Moraes VL. Immunosuppressive and anti-inflammatory effects of methanolic extract and the polyacetylene isolated from Bidens pilosa L. Immunopharmacology. 1999; 43: 31-37. [CrossRef] [Google scholar] [PubMed]
  83. Chiang LC, Chang JS, Chen CC, Ng LT, Lin CC. Anti-Herpes simplex virus activity of Bidens pilosa and Houttuynia cordata. Am J Chin Med. 2003; 31: 355-362. [CrossRef] [Google scholar] [PubMed]
  84. Abajo C, Boffill MÁ, del Campo J, Méndez MA, González Y, Mitjans M, et al. In vitro study of the antioxidant and immunomodulatory activity of aqueous infusion of Bidens pilosa. J Ethnopharmacol. 2004; 93: 319-323. [CrossRef] [Google scholar] [PubMed]
  85. Deba F, Xuan TD, Yasuda M, Tawata S. Chemical composition and antioxidant, antibacterial and antifungal activities of the essential oils from Bidens pilosa Linn. var. Radiata. Food Control. 2008; 19: 346-352. [CrossRef] [Google scholar]
  86. Hassan KA, Deogratius O, Nyafuono JF, Francis O, Engeu OP. Wound healing potential of the ethanolic extracts of Bidens pilosa and Ocimum suave. Afr J Pharm Pharmacol. 2011; 5: 132-136. [CrossRef] [Google scholar]
  87. Bartolome AP, Villaseñor IM, Yang WC. Bidens pilosa L. (Asteraceae): Botanical properties, traditional uses, phytochemistry, and pharmacology. Evid Based Complementary Altern Med. 2013; 2013: 340215. [CrossRef] [Google scholar] [PubMed]
  88. Yang WC. Botanical, pharmacological, phytochemical, and toxicological aspects of the antidiabetic plant Bidens pilosa L. Evid Based Complementary Altern Med. 2014; 2014: 698617. [CrossRef] [Google scholar] [PubMed]
  89. Xuan TD, Khanh TD. Chemistry and pharmacology of Bidens pilosa: An overview. J Pharm Investig. 2016; 46: 91-132. [CrossRef] [Google scholar] [PubMed]
  90. Borel C, Hostettmann K. Molluscicidal saponins from Swartzia madagascariensis Desvaux. Helv Chim Acta. 1987; 70: 570-576. [CrossRef] [Google scholar]
  91. Ouattara Y, Sanon S, Traoré Y, Mahiou V, Azas N, Sawadogo L. Antimalarial activity of Swartzia madagascariensis desv. (Leguminosae), Combretum glutinosum guill. & perr. (Combretaceae) and Tinospora bakis miers. (Menispermaceae), Burkina Faso medicinal plants. Afr J Tradit Complement Altern Med. 2006; 3: 75-81. [CrossRef] [Google scholar]
  92. Adeyemi MH, Amupitan JO. Chemical and biological evaluation of the stem bark of Bobgunnia madagascariensis (Desv.) JH Kirkbr. & Wiersema). J Basic Appl Chem. 2011; 1: 46-51. [Google scholar]
  93. Sani MH, Ibrahim G, Danmalam UH, Muhammad ZA, Kachallah M. Phytochemical study and antibacterial properties of the leaf extracts of Swartzia madagascariensis Desv (Fabaceae). Br Microbiol Res J. 2016; 11: BMRJ.22372. [CrossRef] [Google scholar]
  94. Chingwaru C, Bagar T, Chingwaru W. Aqueous extracts of Flacourtia indica, Swartzia madagascariensis and Ximenia caffra are strong antibacterial agents against Shigella spp., Salmonella typhi and Escherichia coli O157. S Afr J Bot. 2020; 128: 119-127. [CrossRef] [Google scholar]
  95. Constance C, Bagar T, Walter C. Aqueous Extracts of Pericopsis angolensis and Swartzia madagascariensis with high antimicrobial activities against Escherichia coli O157, Shigella spp. and Salmonella enterica subsp. enterica (Serovar typhi). Afr J Biotechnol. 2019; 18: 831-844. [CrossRef] [Google scholar]
  96. Amang AP, Baponwa O, Mezui C, Siwe GT, Vandi LV, Njuh A, et al. Effects of aqueous extract of root barks of Swartzia madagascariensis (Caesalpiniaceae) on acute kidney failure induced with gentamicin in Wistar rats. J Med Plants Stud. 2020; 8: 183-197. [Google scholar]
  97. Heyman HM, Hussein AA, Meyer JJ, Lall N. Antibacterial activity of South African medicinal plants against methicillin resistant Staphylococcus aureus. Pharm Biol. 2009; 47: 67-71. [CrossRef] [Google scholar]
  98. Gadaga LL. Investigation of the toxicological and pharmacological activity of a hydroethanolic extract of Boophone disticha bulb. Harare, Zimbabwe: University of Zimbabwe; 2012. [Google scholar]
  99. Gadaga LL, Tagwireyi D, Dzangare J, Nhachi CF. Acute oral toxicity and neurobehavioural toxicological effects of hydroethanolic extract of Boophone disticha in rats. Hum Exp Toxicol. 2011; 30: 972-980. [CrossRef] [Google scholar] [PubMed]
  100. Cheesman L. Micropropagation and pharmacological evaluation of Boophone disticha. Pietermaritzburg, South Africa: University of KwaZulu-Natal; 2013. [Google scholar]
  101. Nair JJ, Van Staden J. Traditional usage, phytochemistry and pharmacology of the South African medicinal plant Boophone disticha (L.f.) Herb. (Amaryllidaceae). J Ethnopharmacol. 2014; 151: 12-26. [CrossRef] [Google scholar] [PubMed]
  102. Ganga T, Gadaga LL, Tagwireyi D. Neurobehavioural toxicity study of a hydro-ethanolic extract of boophone disticha in sprague dawley rats. In: Drug discovery from herbs: Approaches and applications. Daya Publishing House; 2017. pp. 277-290. [Google scholar]
  103. Pote W, Musarira S, Chuma D, Gadaga LL, Mwandiringana E, Tagwireyi D. Effects of a hydroethanolic extract of Boophone disticha bulb on anxiety-related behaviour in naive BALB/c mice. J Ethnopharmacol. 2018; 214: 218-224. [CrossRef] [Google scholar] [PubMed]
  104. Pote W, Tagwireyi D, Chinyanga HM, Musara C, Nyandoro G, Chifamba J, et al. Cardiovascular effects of Boophone disticha aqueous ethanolic extract on early maternally separated BALB/C mice. J Ethnopharmacol. 2013; 148: 379-385. [CrossRef] [Google scholar] [PubMed]
  105. Mathisen E, Diallo D, Andersen ØM, Malterud KE. Antioxidants from the bark of Burkea africana, an African medicinal plant. Phytother Res. 2002; 16: 148-153. [CrossRef] [Google scholar] [PubMed]
  106. Yaro AH, Malami S, Yau J, Sagoe CN, Anuka JA. Some behavioural studies on methanol root bark extract of Burkea africana (fabaceae) in mice. Bayero J Pure Appl Sci. 2015; 8: 216-219. [CrossRef] [Google scholar]
  107. Dzoyem JP, Eloff JN. Anti-inflammatory, anticholinesterase and antioxidant activity of leaf extracts of twelve plants used traditionally to alleviate pain and inflammation in South Africa. J Ethnopharmacol. 2015; 160: 194-201. [CrossRef] [Google scholar] [PubMed]
  108. Moura I, Duvane JA, Ribeiro N, Ribeiro-Barros I. Woody species from the Mozambican Miombo woodlands: A review on their ethnomedicinal uses and pharmacological potential. J Med Plants Res. 2018; 12: 15-31. [CrossRef] [Google scholar]
  109. Namadina MM, Aliyu BS, Haruna H, Sunusi U, Kamal RM, Balarabe S, et al. Pharmacognostic and acute toxicity study of Burkea africana root. J Appl Sci Environ Manag. 2020; 24: 565-573. [CrossRef] [Google scholar]
  110. Ezenyi IC, Okpoko CK, Ufondu CA, Okhale SE, Adzu B. Antiplasmodial, antinociceptive and antipyretic potential of the stem bark extract of Burkea africana and identification of its antiplasmodial-active fraction. J Tradit Complement Med. 2021; 11: 311-317. [CrossRef] [Google scholar] [PubMed]
  111. Sanwal R, Chaudhary AK. Wound healing and antimicrobial potential of Carissa spinarum Linn. in albino mice. J Ethnopharmacol. 2011; 135: 792-796. [CrossRef] [Google scholar] [PubMed]
  112. Koffuor GA, Sam GH, Dadzeasah PE, Owiafe EO, Gyapong AA. Erythropoietic effect of the ethanolic root bark extract of Carissa edulis in phenylhydrazine-induced anemic Sprague-Dawley rats. Res J Pharmacol. 2012; 6: 20-24. [CrossRef] [Google scholar]
  113. Al-Youssef HM, Hassan WH. Phytochemical and pharmacological aspects of Carissa Edulis Vahl: A review. Int J Curr Res Chem Pharm Sci. 2014; 1: 12-24. [Google scholar]
  114. Ibrahim H, Williams FE, Salawu KM, Usman AM. Phytochemical screening and acute toxicity studies of crude ethanolic extract and flavonoid fraction of Carissa edulis leaves. Biokemistri. 2015; 27: 39-43. [Google scholar]
  115. Woode E, Ansah C, Ainooson GK, Abotsi WM, Mensah AY, Duweijua M. Anti-inflammatory and antioxidant properties of the root extract of Carissa edulis (Forsk.) Vahl (Apocynaceae). J Sci Technol. 2007; 27: 5-15. [CrossRef] [Google scholar]
  116. Osseni R, Akoha S, Adjagba M, Azonbakin S, Lagnika L, Awede B, et al. In vivo toxicological assessment of the aqueous extracts of the leaves of Carissa edulis (Apocynaceae) in Wistar rats. Eur J Med Plants. 2016; 15: EJMP.26083. [CrossRef] [Google scholar]
  117. Ya’u J, Malami S, Abdullahi SB, Yaro AH. Analgesic and anti-inflammatory activities of the residual aqueous fraction of Carissa edulis root bark (Vahl) in experimental animals. Afr J Pharmacol Ther. 2017; 6: 173-177. [Google scholar]
  118. Kaunda JS, Zhang YJ. The genus Carissa: An ethnopharmacological, phytochemical and pharmacological review. Nat Prod Bioprospecting. 2017; 7: 181-199. [CrossRef] [Google scholar] [PubMed]
  119. Lincone OL, Mungai MP, Chege BM, Wangechi MA, George SC, Kayaja WD. Myorelaxant effect of aqueous root bark extract of Carissa edulis (Forssk.) Vahl. is mediated through activation of nitric oxide synthase. Afr J Pharm Pharmacol. 2019; 13: 124-133. [CrossRef] [Google scholar]
  120. Kalk P. A comparison of the catecholamine releasing effect of the khat alkaloids (-)-cathinone and (+)-norpseudoephedrine. Drug Alcohol Depend. 1983; 11: 395-401. [CrossRef] [Google scholar] [PubMed]
  121. Kalix P. Catha edulis, a plant that has amphetamine effects. Pharm World Sci. 1996; 18: 69-73. [CrossRef] [Google scholar] [PubMed]
  122. Kalix P, Braenden OJ. Pharmacological aspects of the chewing of khat leaves. Pharmacol Rev. 1985; 37: 149-164. [CrossRef] [Google scholar]
  123. Al-Meshal IA, Tariq M, Parmar NS, Ageel AM. Anti-inflammatory activity of the flavonoid fraction of khat (Catha edulis Forsk). Agents Actions. 1986; 17: 379-380. [CrossRef] [Google scholar] [PubMed]
  124. Connor J, Makonnen E, Rostom A. Comparison of analgesic effects of khat (Catha edulis Forsk) extract, D-amphetamine and ibuprofen in mice. J Pharm Pharmacol. 2000; 52: 107-110. [CrossRef] [Google scholar] [PubMed]
  125. Kassie F, Darroudi F, Kundi M, Schulte-Hermann R, Knasmüller S. Khat (Catha edulis) consumption causes genotoxic effects in humans. Int J Cancer. 2001; 92: 329-332. [CrossRef] [Google scholar] [PubMed]
  126. Al-Hebshi N, Skaug N. Khat (Catha edulis)—An updated review. Addict Biol. 2005; 10: 299-307. [CrossRef] [Google scholar] [PubMed]
  127. Al-hebshi N, Al-haroni M, Skaug N. In vitro antimicrobial and resistance-modifying activities of aqueous crude khat extracts against oral microorganisms. Arch Oral Biol. 2006; 51: 183-188. [CrossRef] [Google scholar] [PubMed]
  128. Dudai N, Fischer R, Segev D, Chaimovitsh D, Rosenzweig N, Shimoni M. Antioxidative activity of khat (Catha edulis Forsk.). In: II International Symposium on Natural Preservatives in Food, Feed, and Cosmetics. Korbeek-Lo, Belgium: International Society for Horticultural Science; 2006. pp. 85-92. [CrossRef] [Google scholar]
  129. Wabe NT. Chemistry, pharmacology, and toxicology of khat (Catha edulis forsk): A review. Addict Health. 2011; 3: 137. [Google scholar] [PubMed]
  130. Siddiqui R, Warsame AA, Khan NA. Antibacterial and anti-acanthamoebic properties of Catha edulis (Khat). J Bacteriol Parasitol. 2012; 3: 1. [CrossRef] [Google scholar]
  131. Ketema T, Yohannes M, Alemayehu E, Ambelu A. Evaluation of immunomodulatory activities of methanolic extract of khat (Catha edulis, Forsk) and cathinone in Swiss albino mice. BMC Immunol. 2015; 16: 9. [CrossRef] [Google scholar] [PubMed]
  132. Abou-Elhamd AS, Ageely H, Abu-Elmagd M, Zayed AE. Catha edulis forsk mediates embryotoxic effects in rats: An experimental study. Int J Morphol. 2018; 36: 1087-1094. [CrossRef] [Google scholar]
  133. Bedada W, de Andrés F, Engidawork E, Hussein J, LLerena A, Aklillu E. Effects of Khat (Catha edulis) use on catalytic activities of major drug-metabolizing cytochrome P450 enzymes and implication of pharmacogenetic variations. Sci Rep. 2018; 8: 12726. [CrossRef] [Google scholar] [PubMed]
  134. Marume A, Matope G, Katsande S, Khoza S, Mutingwende I, Mduluza T, et al. Wound healing properties of selected plants used in ethnoveterinary medicine. Front Pharmacol. 2017; 8: 544. [CrossRef] [Google scholar] [PubMed]
  135. Mohanty A, Sahu P, Das C. Wound healing activities of methanolic extract of Cissus quadrangularis on albino rat. Int J Drug Formul Res. 2010; 1: 176-184. [Google scholar]
  136. Sen MK, Dash BK. A review on phytochemical and pharmacological aspects of Cissus quadrangularis L. Int J Green Pharm. 2012; 6: 169-173. [CrossRef] [Google scholar]
  137. Buddhadev S, Buddhadev S. A review update on plant Cissus quadrangularis L. Punarnav. 2014; 2: 1-10. [Google scholar]
  138. Chan YS, Cheah YH, Chong PZ, Khor HL, Teh WS, Khoo KS, et al. Antifungal and cytotoxic activities of selected medicinal plants from Malaysia. Pak J Pharm Sci. 2018; 31: 119-127. [Google scholar]
  139. Kavitha A, Babu AN, Nadendla RR. Acute toxicity study of Cissus quadrangularis in Swiss Albino mice. Panacea J Pharm Pharm Sci. 2018; 7: 748-756. [Google scholar]
  140. Sundaran J. A short review on pharmacological activity of Cissus quadrangularis. Bioinformation. 2020; 16: 579. [CrossRef] [Google scholar] [PubMed]
  141. Ado A, Dabo NT. Cytotoxicity of Citrullus lanatus (Thunb) Mansf. on Artemia salina (brine shrimp test). Niger J Chem Res. 2013; 18: 1-5. [Google scholar]
  142. Poduri A, Rateri DL, Saha SK, Saha S, Daugherty A. Citrullus lanatus ‘sentinel’(watermelon) extract reduces atherosclerosis in LDL receptor-deficient mice. J Nutr Biochem. 2013; 24: 882-886. [CrossRef] [Google scholar] [PubMed]
  143. Varghese S, Narmadha R, Gomathi D, Kalaiselvi M, Devaki K. Evaluation of hypoglycemic effect of ethanolic seed extracts of Citrullus lanatus. J Phytopharmacol. 2013; 2: 31-40. [CrossRef] [Google scholar]
  144. Deshmukh CD, Jain A, Tambe MS. Phytochemical and pharmacological profile of Citrullus lanatus (THUNB). Biolife. 2015; 3: 483-488. [CrossRef] [Google scholar]
  145. Swapnil S, Singh G, Dave V, Choudhary N, Yadav S. Healing potential of Citrullus lanatus in acetic acid induced ulcerated rats. J Pharm Sci Emerg Drugs. 2015; 3: 1. [CrossRef] [Google scholar]
  146. Komane B, Vermaak I, Kamatou G, Summers B, Viljoen A. The topical efficacy and safety of Citrullus lanatus seed oil: A short-term clinical assessment. S Afr J Bot. 2017; 112: 466-473. [CrossRef] [Google scholar]
  147. Siddiqui WA, Shahzad M, Shabbir A, Ahmad A. Evaluation of anti-urolithiatic and diuretic activities of watermelon (Citrullus lanatus) using in vivo and in vitro experiments. Biomed Pharmacother. 2018; 97: 1212-1221. [CrossRef] [Google scholar] [PubMed]
  148. Ogbeifun HE, Peters DE, Monanu M. Effect of aqueous extract of Citrullus Lanatus (water melon) seeds on alloxan induced-diabetic wistar rats. Asian J Res Bioch. 2020; 6: 30-44. [CrossRef] [Google scholar]
  149. Gundidza M, Chinyanganya F, Chagonda L, De Pooter HL, Mavi S. Phytoconstituents and antimicrobial activity of the leaf essential oil of Clausena anisata (Willd.) JD Hook ex. Benth. Flavour Fragr J. 1994; 9: 299-303. [CrossRef] [Google scholar]
  150. Arbab IA, Abdul AB, Aspollah M, Abdullah R, Abdelwahab SI, Ibrahim MY, et al. A review of traditional uses, phytochemical and pharmacological aspects of selected members of Clausena genus (Rutaceae). J Med Plants Res. 2012; 6: 5107-5118. [CrossRef] [Google scholar]
  151. Irungu BN, Mbabu MJ, Kiboi DM, Moindi E, Kinyua J, Mwirichia RK. In vivo antimalarial and acute toxicity properties of hexane and chloroform extracts from Clausena anisata (Willd.) Benth. Afr J Pharmacol Ther. 2012; 1: 24-29. [Google scholar]
  152. Agyepong N, Agyare C, Ossei PP, Boakye YD. Antioxidant and in vivo wound healing activities of Clausena anisata. Eur J Med Plants. 2015; 10: EJMP.19792. [CrossRef] [Google scholar]
  153. Ishola IO, Olayemi SO, Oreagba IA, Ifeanyi CI, Popoola TD. Antinociceptive and anti-arthritic properties of hydroethanolic leaf extract of Clausena anisata (Willd.) Hook. f. ex Benth (Rutaceae) in Rodents: Possible mechanism of actions. Niger J Physiol Sci. 2015; 30: 39-49. [Google scholar]
  154. Yakoob AT, Tajuddin NB, Hussain MI, Mathew S, Govindaraju A, Qadri I. Antioxidant and hypoglycemic activities of Clausena anisata (Willd.) Hook. f. ex Benth root mediated synthesized silver nanoparticles. Pharmacogn J. 2016; 8: 579-586. [CrossRef] [Google scholar]
  155. De Canha MN, Kishore N, Kumar V, Meyer D, Nehar S, Singh B, et al. The potential of Clausena anisata (Willd.) Hook. f. ex Benth against Propionibacterium acnes. S Afr J Bot. 2018; 119: 410-419. [CrossRef] [Google scholar]
  156. Fouad MA, Wanas AS, Khalil HE. Phytochemical and biological studies of Clerodendrum Glabraum leaves. Int J Pharmacogn Phytochem. 2013; 28: 1164-1168. [Google scholar]
  157. Ndlovu G, Fouche G, Tselanyane M, Cordier W, Steenkamp V. In vitro determination of the anti-aging potential of four southern African medicinal plants. BMC Complement Altern Med. 2013; 13: 304. [CrossRef] [Google scholar] [PubMed]
  158. Chiramba O, Mukanganyama S. Cytotoxic effects of Combretum platypetalum Welw. ex MA Lawson subsp. oatesii (Rolfe) Exell (Combretaceae) leaf extracts on Jurkat T-cells and reversal of effects by reduced glutathione. J Biol Act Prod Nat. 2016; 6: 250-265. [CrossRef] [Google scholar]
  159. Magwenzi R, Nyakunu C, Mukanganyama S. The Effect of selected Combretum species from Zimbabwe on the growth and drug efflux systems of Mycobacterium aurum and Mycobacterium smegmatis. J Microb Biochem Technol. 2014; 3: S3. [CrossRef] [Google scholar]
  160. Chirisa E, Mukanganyama S. Evaluation of in vitro anti-inflammatory and antioxidant activity of selected Zimbabwean plant extracts. J Herbs Spices Med Plants. 2016; 22: 157-172. [CrossRef] [Google scholar]
  161. Wende M, Sithole S, Chi GF, Stevens MY, Mukanganyama S. The effects of combining cancer drugs with compounds isolated from Combretum zeyheri Sond. and Combretum platypetalum Welw. Ex MA Lawson (Combretaceae) on the viability of Jurkat T cells and HL-60 cells. BioMed Res Int. 2021; 2021: 6049728. [CrossRef] [Google scholar] [PubMed]
  162. Hamza RZ, Al-Motaani SE, Al-Talhi T. Retracted: Therapeutic and Ameliorative Effects of Active Compounds of Combretum molle in the Treatment and Relief from Wounds in a Diabetes Mellitus Experimental Model. Coatings. 2021; 11: 324. [CrossRef] [Google scholar]
  163. Asres K, Bucar F. Anti-HIV activity against immunodeficiency virus type 1 (HIV-I) and type II (HIV-II) of compounds isolated from the stem bark of Combretum molle. Ethiop Med J. 2005; 43: 15-20. [Google scholar]
  164. Asres K, Mazumder A, Bucar F. Antibacterial and antifungal activities of extracts of Combretum molle. Ethiop Med J. 2006; 44: 269-277. [Google scholar]
  165. Ojewole JA. Analgesic and antiinflammatory effects of mollic acid glucoside, a 1α-hydroxycycloartenoid saponin extractive from Combretum molle R. Br. ex G. Don (Combretaceae) leaf. Phytother Res. 2008; 22: 30-35. [CrossRef] [Google scholar] [PubMed]
  166. Ojewole JA. Cardiovascular effects of mollic acid glucoside, a 1α-hydroxycycloartenoid saponin extractive from Combretum molle R Br ex G Don (Combretaceae) leaf: Cardiovascular topic. Cardiovasc J Afr. 2008; 19: 128-134. [CrossRef] [Google scholar] [PubMed]
  167. Ojewole JA, Adewole SO. Hypoglycaemic effect of mollic acid glucoside, a 1α-hydroxycycloartenoid saponin extractive from Combretum molle R. Br. ex G. Don (Combretaceae) leaf, in rodents. J Nat Med. 2009; 63: 117-123. [CrossRef] [Google scholar] [PubMed]
  168. Masoko P, Picard J, Howard RL, Mampuru LJ, Eloff JN. In vivo antifungal effect of Combretum and Terminalia species extracts on cutaneous wound healing in immunosuppressed rats. Pharm Biol. 2010; 48: 621-632. [CrossRef] [Google scholar] [PubMed]
  169. Yeo D, Bouagnon R, Djyh BN, Tuo C, N’guessan JD. Acute and subacute toxic study of aqueous leaf extract of Combretum molle. Trop J Pharm Res. 2012; 11: 217-223. [CrossRef] [Google scholar]
  170. Suleiman MM, Simon MK, Ajanusi OJ, Idris AL, Abubakar MS. In vitro anthelmintic activity of the stem-bark of Combretum molle R. Br. x. G. Don (Combretaceae) against Haemonchus contortus. J Med Plants Res. 2013; 7: 952-956. [Google scholar]
  171. Koevi KK, Millogo V, Fokou JB, Sarr A, Ouedraogo GA, Bassene E. Phytochemical analysis and antioxidant activities of Combretum molle and Pericopsis laxiflora. Int J Biol Chem Sci. 2015; 9: 2423-2431. [CrossRef] [Google scholar]
  172. Anato M, Ketema T. Anti-plasmodial activities of Combretum molle (Combretaceae) [Zwoo] seed extract in Swiss albino mice. BMC Res Notes. 2018; 11: 312. [CrossRef] [Google scholar] [PubMed]
  173. Grace OM, Prendergast HD, Jäger AK, Van Staden J, Van Wyk AE. Bark medicines used in traditional healthcare in KwaZulu-Natal, South Africa: An inventory. S Afr J Bot. 2003; 69: 301-363. [CrossRef] [Google scholar]
  174. Okokon JE, Ofodum KC, Ajibesin KK, Danladi B, Gamaniel KS. Pharmacological screening and evaluation of antiplasmodial activity of Croton zambesicus against Plasmodium berghei berghei infection in mice. Indian J Pharmacol. 2005; 37: 243-246. [CrossRef] [Google scholar]
  175. Okokon JE, Umoh UF, Udobang JA, Etim EI. Antiulcerogenic activity of ethanolic leaf extract of Croton zambesicus Muell. Arg. Afr J Biomed Res. 2011; 14: 43-47. [Google scholar]
  176. Salatino A, Salatino ML, Negri G. Traditional uses, chemistry and pharmacology of Croton species (Euphorbiaceae). J Braz Chem Soc. 2007; 18: 11-33. [CrossRef] [Google scholar]
  177. Okokon JE, Dar A, Choudhary MI. Immunomodulatory, cytotoxic and antileishmanial activity of phytoconstituents of Croton zambesicus. Phytopharmacol J. 2013; 4: 31-40. [Google scholar]
  178. Mfotie Njoya E, Eloff JN, McGaw LJ. Croton gratissimus leaf extracts inhibit cancer cell growth by inducing caspase 3/7 activation with additional anti-inflammatory and antioxidant activities. BMC Complement Altern Med. 2018; 18: 305. [CrossRef] [Google scholar] [PubMed]
  179. Mahmoud AB, Danton O, Kaiser M, Khalid S, Hamburger M, Mäser P. HPLC-based activity profiling for antiprotozoal compounds in Croton gratissimus and Cuscuta hyalina. Front Pharmacol. 2020; 11: 1246. [CrossRef] [Google scholar] [PubMed]
  180. Kiondo F, Feyissa T, Ndakidemi PA, Seth M. In vitro propagation of Dalbergia melanoxylon Guill. & Perr.: A multipurpose tree. Am J Res Commun. 2014; 2: 181-194. [Google scholar]
  181. Matata DZ, Ngassapa OD, Machumi F, Moshi MJ. Screening of plants used as traditional anticancer remedies in mkuranga and same districts, Tanzania, using brine shrimp toxicity bioassay. Evid Based Complementary Altern Med. 2018; 2018: 3034612. [CrossRef] [Google scholar] [PubMed]
  182. Najeeb TM, Issa TO, Mohamed YS, Ahmed RH, Makhawi AM, Khider TO. Phytochemical screening, antioxidant and antimicrobial activities of Dalberegia melanoxylon tree. World Appl Sci J. 2018; 36: 826-833. [Google scholar]
  183. Moshi MJ, Cosam JC, Mbwambo ZH, Kapingu M, Nkunya MH. Testing beyond ethnomedical claims: Brine shrimp lethality of some Tanzanian plants. Pharm Biol. 2004; 42: 547-551. [CrossRef] [Google scholar]
  184. Kazembe T, Munyarari E, Charumbira I. Use of traditional herbal medicines to cure malaria. Bull Environ Pharmacol Life Sci. 2012; 1: 63-85. [Google scholar]
  185. Saha S, Shilpi JA, Mondal H, Hossain F, Anisuzzman M, Hasan MM, et al. Ethnomedicinal, phytochemical, and pharmacological profile of the genus Dalbergia L. (Fabaceae). Phytopharmacology. 2013; 4: 291-346. [Google scholar]
  186. Dzoyem JP, McGaw LJ, Eloff JN. In vitro antibacterial, antioxidant and cytotoxic activity of acetone leaf extracts of nine under-investigated Fabaceae tree species leads to potentially useful extracts in animal health and productivity. BMC Complement Altern Med. 2014; 14: 147. [CrossRef] [Google scholar] [PubMed]
  187. Sreenivasa S, Vinay K, Mohan NR. Phytochemical analysis, antibacterial and antioxidant activity of leaf extract of Datura stramonium. Int J Sci Res. 2012; 1: 83-86. [Google scholar]
  188. Gaire BP, Subedi L. A review on the pharmacological and toxicological aspects of Datura stramonium L. J Integr Med. 2013; 11: 73-79. [CrossRef] [Google scholar] [PubMed]
  189. Sayyed A, Shah M. Phytochemistry, pharmacological and traditional uses of Datura stramonium L. Rev J Pharmacogn Phytochem. 2014; 2: 123-125. [Google scholar]
  190. Pallavi Sharma PS, Richa Bhardwaj RB, Ankita Yadav AY, Sharma RA. Study of antioxidant activity of Datura stramonium Linn. Res J Phytochem. 2014; 8: 112-118. [CrossRef] [Google scholar]
  191. Ananth A, Rajan S. In-vitro antioxidant activity of Datura stramonium L. leaves. Adv Appl Sci Res. 2015; 6: 147-151. [Google scholar]
  192. Iqbal S, Sivaraj C, Gunasekaran K. Antioxidant and anticancer activities of methanol extract of seeds of Datura stramonium l. Free Radic Antioxid. 2017; 7: 184-189. [CrossRef] [Google scholar]
  193. Shekhar P, Joshi A, Malviya S, Kharia A. Wound healing activity of the hydro-alcoholic extract of Datura stramonium leaves in wistar albino rats. J Drug Deliv Ther. 2017; 7: 214-215. [Google scholar]
  194. Cherie Melaku B, Amare GG. Evaluation of antidiabetic and antioxidant potential of hydromethanolic seed extract of Datura stramonium Linn (Solanaceae). J Exp Pharmacol. 2020; 12: 181-189. [CrossRef] [Google scholar] [PubMed]
  195. More G, Tshikalange TE, Lall N, Botha F, Meyer JJ. Antimicrobial activity of medicinal plants against oral microorganisms. J Ethnopharmacol. 2008; 119: 473-477. [CrossRef] [Google scholar] [PubMed]
  196. Madiga MC, Cockeran R, Mokgotho MP, Anderson R, Mampuru LJ. Dichloromethane extract of Dicerocaryum senecioides leaves exhibits remarkable anti-inflammatory activity in human T-lymphocytes. Nat Prod Res. 2009; 23: 998-1006. [CrossRef] [Google scholar] [PubMed]
  197. Chokoe PK, Mampuru LJ, Mokgotho MP. The anti-proliferative, antioxidative and anti-inflammatory properties of the D2 fraction and HPLC semi-purified sub-fractions of Dicerocaryum senecioides. Polokwane, South Africa: University of Limpopo; 2011. [Google scholar]
  198. Mampuru LJ, Chokoe PK, Madiga MC, Theron A, Anderson R, Mokgotho MP. Antioxidant and anti-proliferative capacity of a dichloromethane extract of Dicerocaryum senecioides leaves. In: Phytochemicals as nutraceuticals-global approaches to their role in nutrition and health. Rijeka, Croatia: IntechOpen; 2012. pp. 97-114. [Google scholar]
  199. Malemela KM, Mbazima VG, Riedel van Heerden S. Investigation of the probable anti-cancer effects of the crude methanol extract of Dicerocaryum senecioides, (Klotzch) J. Abels, leaves on cervical HeLa cancer cell. Polokwane, South Africa: University of Limpopo; 2018. [Google scholar]
  200. Rambwawasvika H, Dzomba P, Gwatidzo L. Hair growth promoting effect of Dicerocaryum senecioides phytochemicals. Int J Med Chem. 2019; 2019: 7105834. [CrossRef] [Google scholar] [PubMed]
  201. Aworet-Samseny RR, Souza A, Kpahé F, Konaté K, Datté JY. Dichrostachys cinerea (L.) wight et Arn (Mimosaceae) hydro-alcoholic extract action on the contractility of tracheal smooth muscle isolated from guinea-pig. BMC Complement Altern Med. 2011; 11: 23. [CrossRef] [Google scholar] [PubMed]
  202. Viol DI, Chagonda LS, Moyo SR, Mericli AH. Antibacterial, antifungal and phytochemical activities of some medicinal plants from Zimbabwe. J Biol Act Prod Nat. 2013; 3: 273-284. [CrossRef] [Google scholar]
  203. Irie-N’guessan AG, Kouakou SL, Koua KB, Effo KE, Djadji AT, Diarrassouba NG, et al. Anticonvulsant and analgesic assessment of Dichrostachys cinerea root bark, an Ivorian anti-asthmatic herbal, in mice. J Pharmacol Clin Res. 2018; 6: 1-6. [CrossRef] [Google scholar]
  204. Genevieve N, Amenan GI, Yesse ZN, Anderson DR, Landry K, Landry KS, et al. Anti-infective assessment of Dichrostachys cinerea root bark, an Ivorian anti-asthmatic herbal. Adv Pharmacol Pharm. 2018; 6: 72-76. [CrossRef] [Google scholar]
  205. Steenkamp V, Mathivha E, Gouws MC, Van Rensburg CE. Studies on antibacterial, antioxidant and fibroblast growth stimulation of wound healing remedies from South Africa. J Ethnopharmacol. 2004; 95: 353-357. [CrossRef] [Google scholar] [PubMed]
  206. Becker JV, Van der Merwe MM, van Brummelen AC, Pillay P, Crampton BG, Mmutlane EM, et al. In vitro anti-plasmodial activity of Dicoma anomala subsp. gerrardii (Asteraceae): Identification of its main active constituent, structure-activity relationship studies and gene expression profiling. Malar J. 2011; 10: 295. [CrossRef] [Google scholar] [PubMed]
  207. Balogun FO, Ashafa AO. Aqueous root extract of Dicoma anomala Sond ameliorates isoproterenol–induced myocardial infarction in Wistar rats. Trop J Pharm Res. 2016; 15: 1651-1657. [CrossRef] [Google scholar]
  208. Balogun FO, Ashafa AO. Aqueous root extracts of Dicoma anomala (Sond.) extenuates postprandial hyperglycaemia in vitro and its modulation on the activities of carbohydrate-metabolizing enzymes in streptozotocin-induced diabetic Wistar rats. S Afr J Bot. 2017; 112: 102-111. [CrossRef] [Google scholar]
  209. Shava C, Mutsaka P, Moyo B, Sithole S, Chitemerere T, Mukanganyama S. Antibacterial and anticancer properties of Dolichos kilimandscharicus (Fabaceae). J Biol Act Prod Nat. 2016; 6: 112-135. [CrossRef] [Google scholar]
  210. Munissi J. Cytotoxic and antimicrobial activities of the constituents of ten plant species from Tanzania. Tanzan J Sci. 2019; 45: 44-52. [Google scholar]
  211. Sithole S, Mushonga P, Nhamo LN, Fru Chi G, Mukanganyama S. Phytochemical Fingerprinting and Activity of Extracts from the Leaves of Dolichos kilimandscharicus (Fabaceae) on Jurkat-T Cells. BioMed Res Int. 2020; 2020: 1263702. [CrossRef] [Google scholar] [PubMed]
  212. Donkor S, Larbie C, Komlaga G, Emikpe BO. Phytochemical, antimicrobial, and antioxidant profiles of Duranta erecta L. parts. Biochem Res Int. 2019; 2019: 8731595. [CrossRef] [Google scholar] [PubMed]
  213. Donkor S, Larbie C, Komlaga G, Emikpe BO. Toxicity of the hydroethanolic leaves extract of Duranta erecta L. in rat models. J Pharm Res Int. 2020; 32: 9-18. [CrossRef] [Google scholar]
  214. Ugwu C, Anosike CA. Phytochemical screening and antioxidant potentials of methanol extract of Duranta erecta leaves. Asian J Res Biochem. 2020; 7: 16-26. [CrossRef] [Google scholar]
  215. McFeeters H, Gilbert MJ, Thompson RM, Setzer WN, Cruz-Vera LR, McFeeters RL. Inhibition of essential bacterial peptidyl-tRNA hydrolase activity by tropical plant extracts. Nat Prod Commun. 2012; 7: 1934578X1200700836. [CrossRef] [Google scholar]
  216. Bunalema L, Kirimuhuzya C, Tabuti JR, Waako P, Magadula JJ, Otieno N, et al. The efficacy of the crude root bark extracts of Erythrina abyssinica on rifampicin resistant Mycobacterium tuberculosis. Afr Health Sci. 2011; 11: 587-593. [Google scholar]
  217. Chitopoa W, Muchachaa I, Mangoyi R. Evaluation of the antimicrobial activity of Erythrina abyssinica leaf extract. J Microb Biochem Technol. 2019; 11: 43-46. [Google scholar]
  218. Nkeh-Chungag BN, Tiya S, Mbafor JT, Ndebia EJ, Rusike S, Iputo JE. Effects of the methanol extract of Erythrina abyssinica on hot flashes in ovariectomized rats. Afr J Biotechnol. 2013; 12: 598-601. [Google scholar]
  219. Nasimolo J, Kiama SG, Gathumbi PK, Makanya AN, Kagira JM. Erythrina abyssinica prevents meningoencephalitis in chronic Trypanosoma brucei brucei mouse model. Metab Brain Dis. 2014; 29: 509-519. [CrossRef] [Google scholar] [PubMed]
  220. Macharia FK, Mwangi PW, Yenesew A, Bukachi F, Nyaga NM, Wafula DK. Hepatoprotective effects of erythrina abyssinica lam ex dc against non alcoholic fatty liver disease in sprague dawley rats. BioRxiv. 2019. doi: 10.1101/577007. [CrossRef] [Google scholar]
  221. Magama S, Pretorius JC, Zietsman PC, van Wyk BE. Antimicrobial properties of extracts from Euclea crispa subsp. crispa (Ebenaceae) towards human pathogens. S Afr J Bot. 2003; 69: 193-198. [CrossRef] [Google scholar]
  222. Pretorius JC, Magama S, Zietsman PC. Purification and identification of antibacterial compounds from Euclea crispa subsp. crispa (Ebenaceae) leaves. S Afr J Bot. 2003; 69: 579-586. [CrossRef] [Google scholar]
  223. Maroyi AL. Euclea crispa: Review of its botany, ethnomedicinal uses, and pharmacological properties. Asian J Pharmaceut Clin Res. 2018; 11: 5-9. [CrossRef] [Google scholar]
  224. Mencherini T, Picerno P, Del Gaudio P, Festa M, Capasso A, Aquino R. Saponins and polyphenols from Fadogia ancylantha (Makoni tea). J Nat Prod. 2010; 73: 247-251. [CrossRef] [Google scholar] [PubMed]
  225. Nyirenda KK, Saka JD, Naidoo D, Maharaj VJ, Muller CJ. Antidiabetic, anti-oxidant and antimicrobial activities of Fadogia ancylantha extracts from Malawi. J Ethnopharmacol. 2012; 143: 372-376. [CrossRef] [Google scholar] [PubMed]
  226. Feng ZL, Wu SP, Li WH, Guo TT, Liu QC. Concise synthesis and antidiabetic effect of three natural triterpenoid saponins isolated from Fadogia ancylantha (Makoni tea). Helv Chim Acta. 2015; 98: 1254-1266. [CrossRef] [Google scholar]
  227. Bhebhe M, Chipurura B, Muchuweti M. Determination and comparison of phenolic compound content and antioxidant activity of selected local Zimbabwean herbal teas with exotic Aspalathus linearis. S Afr J Bot. 2015; 100: 213-218. [CrossRef] [Google scholar]
  228. Tiya SY, Sewani-Rusike CR, Taderera T. Hepatoprotective effects of Fadogia ancylantha (Makoni Tea) on ethanol-induced liver damage in Wistar rats. J Biol Act Prod Nat. 2019; 9: 352-363. [CrossRef] [Google scholar]
  229. Eldeen IM, Elgorashi EE, Van Staden J. Antibacterial, anti-inflammatory, anti-cholinesterase and mutagenic effects of extracts obtained from some trees used in South African traditional medicine. J Ethnopharmacol. 2005; 102: 457-464. [CrossRef] [Google scholar] [PubMed]
  230. Kunle OO, Shittu A, Nasipuri RN, Kunle OF, Wambebe C, Akah PA. Gastrointestinal activity of Ficus sur. Fitoterapia. 1999; 70: 542-547. [CrossRef] [Google scholar]
  231. Muregi FW, Ishih A, Miyase T, Suzuki T, Kino H, Amano T, et al. Antimalarial activity of methanolic extracts from plants used in Kenyan ethnomedicine and their interactions with chloroquine (CQ) against a CQ-tolerant rodent parasite, in mice. J Ethnopharmacol. 2007; 111: 190-195. [CrossRef] [Google scholar] [PubMed]
  232. Ramde-Tiendrebeogo A, Tibiri A, Hilou A, Lompo M, Millogo-Kone H, Nacoulma OG, et al. Antioxidative and antibacterial activities of phenolic compounds from Ficus sur Forssk. and Ficus sycomorus L. (Moraceae): Potential for sickle cell disease treatment in Burkina Faso. Int J Biol Chem Sci. 2012; 6: 328-336. [CrossRef] [Google scholar]
  233. Ishola IO, Olayemi SO, Yemitan OK, Ekpemandudiri NK. Mechanisms of anticonvulsant and sedative actions of the ethanolic stem-bark extract of Ficus sur Forssk (Moraceae) in rodents. Pak J Biol Sci. 2013; 16: 1287-1294. [CrossRef] [Google scholar] [PubMed]
  234. Solomon-Wisdom GO, Shittu GA, Agboola YA. Antimicrobial and phytochemical screening activities of Ficus sur (Forssk). N Y Sci J. 2011; 4: 15-18. [Google scholar]
  235. Akomas SC, Ijioma SN, Emelike CU. In vivo and In vitro spasmolytic Effect of Ficus sur Forssk Ethanol Leaf Extract on the Gastrointestinal Tract. Br Biotechnol J. 2014; 4: 1182-1190. [CrossRef] [Google scholar]
  236. Olayemi IK, Samuel OM, Ukubuiwe AC, Ande AT, Adeniyi KA, Shittu KO. Larvicidal activities of leaf extracts of Adansonia digitata L.(Malvales: Malvaceae) and Ficus sur Forssk (Rosales: Moraceae) against Culex quinquefasciatus mosquito (Diptera: Culicidae). J Mosq Res. 2017; 7: 115-124. [CrossRef] [Google scholar]
  237. Adebayo MA, Enitan SS, Owonikoko WM, Igogo E, Ajeigbe KO. Haematinic properties of methanolic stem bark and fruit extracts of Ficus sur in rats pre-exposed to phenylhydrazine-induced haemolytic anaemia. Afr J Biomed Res. 2017; 20: 85-92. [Google scholar]
  238. John-Africa LB, YIsimi C, Abbas M, Daburi A. Promotion of wound healing in mice using a formulation of Ficus sur Forssk aqueous leaf extract. Acad J. 2018; 1: B60E616. [Google scholar]
  239. Akoto CO, Acheampong A, Kwame O, Boateng BA. An Assessment of the anti-inflammatory, antimicrobial, and antioxidant activities of Ficus sur stem-bark. Org Med Chem Int J. 2020; 9: 116-124. [CrossRef] [Google scholar]
  240. Ayele M, Makonnen E, Ayele AG, Tolcha Y. Evaluation of the diuretic activity of the aqueous and 80% methanol extracts of Ficus sur Forssk (Moraceae) leaves in saline-loaded rats. J Exp Pharmacol. 2020; 12: 619-627. [CrossRef] [Google scholar] [PubMed]
  241. Yakubu OE, Ojogbane E, Abu MS, Shaibu CO, Ayegba WE. Haematinic effects of ethanol extract of Ficus sur leaves on diethylnitrosamine-induced toxicity in Wistar rats. J Pharmacol Toxicol. 2020; 15: 16-21. [CrossRef] [Google scholar]
  242. Tatematsu H, Mori M, Yang TH, Chang JJ, Lee TT, Lee KH. Cytotoxic principles of Securinega virosa: Virosecurinine and viroallosecurinine and related derivatives. J Pharm Sci. 1991; 80: 325-357. [CrossRef] [Google scholar] [PubMed]
  243. Moshi MJ, Kapingu MC, Uiso FC, Mbwambo ZH, Mahunnah RL. Some pharmacological properties of an aqueous extract of Securinega virosa roots. Pharm Biol. 2000; 38: 214-221. [CrossRef] [Google scholar] [PubMed]
  244. Magaji MG, Anuka JA, Abdu-Aguye I, Yaro AH, Hussaini IM. Behavioural effects of the methanolic root bark extract of Securinega virosa in rodents. Afr J Tradit Complement Altern Med. 2008; 5: 147-153. [CrossRef] [Google scholar] [PubMed]
  245. Magaji MG, Anuka JA, Abdu-Aguye I, Yaro AH, Hussaini IM. Preliminary studies on anti-inflammatory and analgesic activities of Securinega virosa (Euphorbiaceae) in experimental animal models. J Med Plants Res. 2008; 2: 39-44. [Google scholar]
  246. Magaji MG, Yaro AH, Mohammed A, Zezi AU, Tanko Y, Bala TY. Preliminary antidiarrhoeal activity of methanolic extracts of Securinega virosa (Euphorbiaceae). Afr J Biotechnol. 2007; 6: 2752-2757. [CrossRef] [Google scholar]
  247. Tanko Y, Okasha MA, Magaji GM, Yerima M, Yaro AH, Saleh MI, et al. Anti-diabetic properties of Securinega virosa (Euphorbiaceae) leaf extract. Afr J Biotechnol. 2008; 7: 22-24. [Google scholar]
  248. Sanogo R, Vassallo A, Malafronte N, Imparato S, Russo A, Piaz FD. New phenolic glycosides from Securinega virosa and their antioxidant activity. Nat Prod Commun. 2009; 4. doi: 10.1177/1934578X0900401207. [CrossRef] [Google scholar]
  249. Yerima M, Magaji MG, Yaro AH, Tanko Y, Mohammed MM. Analgesic and antiinflammatory activities of the methanolic leaves extract of Securinega virosa (Euphorbiaceae). Niger J Pharm Sci. 2009; 8: 47-53. [Google scholar]
  250. Ezeonwumelu JO, Omar AN, Ajayi AM, Okoruwa AG, Tanayen JK, Kiplagat DM, et al. Phytochemical screening, acute toxicity, anti-inflammatory and antipyretic studies of aqueous extract of the root of Flueggea virosa (Roxb. ex Willd.) in rats. Int J Pharm Biomed Sci. 2012; 3: 128-135. [Google scholar]
  251. Garba MM, Hamza YA, Muhammad MA, Akpojo AJ, Ibrahim AA, Marte HI. Neuropharmacological studies on ethyl acetate fraction of Securinega virosa root bark extract. Afr J Pharm Pharmacol. 2013; 7: 275-279. [CrossRef] [Google scholar]
  252. Abere TA, Egharevba CO, Chukwurah IO. Pharmacognostic evaluation and antisickling activity of the leaves of Securinega virosa Roxb. ex Willd. (Euphorbiaceae). Afr J Biotechnol. 2014; 13: 4040-4045. [CrossRef] [Google scholar]
  253. Sempombe J, Mugoyela V, Mihale MJ, Zacharia A, Ipagala P, Kilulya KF. Preliminary in vivo antitrypanosomal activity and cytotoxicity of Entada abyssinica, Securinega virosa and Ehretia amoena. East Cent Afr J Pharm Sci. 2014; 17: 37-43. [Google scholar]
  254. Garba MM, Jamilu Y, Muhammad MA, Akpojo AJ, Ibrahim AA, Marte HI. Securinega virosa (Euphorbiaceae) root bark extract inhibits glioblastoma multiforme cell survival in vitro. Afr J Pharm Pharmacol. 2015; 9: 684-693. [CrossRef] [Google scholar]
  255. Zhang H, Zhang CR, Han YS, Wainberg MA, Yue JM. New Securinega alkaloids with anti-HIV activity from Flueggea virosa. RSC Adv. 2015; 5: 107045-107053. [CrossRef] [Google scholar]
  256. Shehu A, Magaji MG, Sanni B, Abdu-Aguye SN. Antidepressant activity of methanol root bark extract of Securinega virosa (Ex Willd.) Baill in albino mice. Bayero J Pure Appl Sci. 2017; 10: 277-282. [CrossRef] [Google scholar]
  257. Ahmad A, Abubakar AH, Garba MM, Jamiu I, Yusuf PO. Antivenin studies of leaf extracts of S. virosa from Zaria, Nothern Nigeria. Innoriginal Int J Sci. 2018; 5: 29-32. [Google scholar]
  258. Ajaib M, Wahla SQ, Wahla UG, Perveen KM, Shah S. Phytochemical screening and anthelmintic activity of Flueggea virosa. J Chem Soc Pak. 2018; 40: 702. [Google scholar]
  259. Renu SN, Archana R, Jaswinder K, Santosh K, Fatima N. Taxonomy, phytochemistry, pharmacology and traditional uses of Flueggea virosa (Roxb. ex Willd.) Royle: A review. Int J Life Sci. 2018; 6: 579-585. [Google scholar] [PubMed]
  260. Misonge OJ, Kamindu GN, Sabina W, Muita G. An ethnobotanical survey of plants used for the treatment and management of cancer in Embu County, Kenya. J Med Plants. 2019; 7: 39-46. [Google scholar]
  261. Salawu MO, Yekeen A, Nafiu MO, Oloyede HO. Anti-ulcerogenic potential of aqueous extract of securinega virosa leaf in indomethacin-induced ulcerated rats. Not Sci Biol. 2019; 11: 196-204. [CrossRef] [Google scholar]
  262. El Kheir YM, El Tohami MS. Investigation of molluscicidal activity of certain Sudanese plants used in folk-medicine. II. Molluscicidal activity of the different morphological organs of Gnidia kraussiana Meisn" abu gotna" family Thymelaeaceae. J Trop Med Hyg. 1979; 82: 242-247. [Google scholar]
  263. Ndhlala AR, Ncube B, Okem A, Mulaudzi RB, Van Staden J. Toxicology of some important medicinal plants in southern Africa. Food Chem Toxicol. 2013; 62: 609-621. [CrossRef] [Google scholar] [PubMed]
  264. Yanadaiah J. Assessment of antidiabetic activity of ethanol extract of grewia flavescens juss leaves against alloxan induced diabetes in rats. J Glob Trends Pharm Sci. 2013; 4: 1086-1090. [Google scholar]
  265. Sana SS, Badineni VR, Arla SK, Boya VK. Eco-friendly synthesis of silver nanoparticles using leaf extract of Grewia flaviscences and study of their antimicrobial activity. Mater Lett. 2015; 145: 347-350. [CrossRef] [Google scholar]
  266. Pramodini GN, Chowdary KA. Study of acute toxicity and anti-inflammatory activity of ethanolic extract of Grewia flavescens juss whole plant using rats. Eur J Pharm Med Res. 2017; 4: 595-599. [Google scholar]
  267. Pramodini GN, Alam P, Rafiq M, Khasim SM. Evaluation of mosquito larvicidal activity of ethanolic extract of Grewia flavescens Juss. whole plant (Family: Tiliaceae) against Culex quinquefasciatus. J Drug Deliv Ther. 2019; 9: 252-255. [CrossRef] [Google scholar]
  268. Tahir AE, Ibrahim AM, Satti GM, Theander TG, Kharazmi A, Khalid SA. The potential antileishmanial activity of some Sudanese medicinal plants. Phytother Res. 1998; 12: 576-579. [CrossRef] [Google scholar]
  269. Lindsey KL, Budesinsky M, Kohout L, Van Staden J. Antibacterial activity of maytenonic acid isolated from the root-bark of Maytenus senegalensis. S Afr J Bot. 2006; 72: 473-477. [CrossRef] [Google scholar]
  270. Da Silva G, Taniça M, Rocha J, Serrano R, Gomes ET, Sepodes B, et al. In vivo anti-inflammatory effect and toxicological screening of Maytenus heterophylla and Maytenus senegalensis extracts. Hum Exp Toxicol. 2011; 30: 693-700. [CrossRef] [Google scholar] [PubMed]
  271. Makgatho ME, Nxumalo W, Raphoko LA. Anti-mycobacterial, oxidative, proliferative and-inflammatory activities of dichloromethane leaf extracts of Gymnosporia senegalensis (Lam.) Loes. S Afr J Bot. 2018; 114: 217-222. [CrossRef] [Google scholar]
  272. Kpoyizoun PK, Metowogo K, Missebukpo A, Eklu-Gadegbeku K, Aklikokou KA, Gbeassor M. Effect of Maytenus senegalensis roots on OVA-induced airway inflammation in a mouse asthma model. Afr J Pharm Pharmacol. 2019; 13: 49-56. [CrossRef] [Google scholar]
  273. Kpoyizoun PK, Metowogo K, Kantati YT, Missebukpo A, Dare T, Lawson-Evi P, et al. Antiinflammatory and antioxidant evaluation of Maytenus senegalensis hydroalcoholic roots extract fractions in allergic asthma. J Phytopharmacol. 2020; 9: 252-257. [CrossRef] [Google scholar]
  274. Bradacs G, Maes L, Heilmann J. In vitro cytotoxic, antiprotozoal and antimicrobial activities of medicinal plants from Vanuatu. Phytother Res. 2010; 24: 800-809. [CrossRef] [Google scholar] [PubMed]
  275. Lundgaard NH, Prior RM, Light ME, Stafford GI, Van Staden J, Jäger AK. COX-1 inhibition of Heteromorpha arborescens. S Afr J Bot. 2008; 74: 335-337. [CrossRef] [Google scholar]
  276. Katerere D, Parry O. Pharmacological actions of Heteromorpha trifoliata (" dombwe") on rat isolated muscle preparations. Cent Afr J Med. 2000; 46: 9-13. [CrossRef] [Google scholar]
  277. Abifarin TO, Otunola GA, Afolayan AJ. Assessment of the phytochemical, antioxidant and antibacterial activities of Heteromorpha arborescens (Spreng.) Cham & Schltdl. leaf extracts. F1000Research. 2020; 9: 1079. [CrossRef] [Google scholar] [PubMed]
  278. Osman HM. Anti-diarrhoeal Activity of Hydnora abyssinica aqueous root extract in rats. Khartoum, Sudan: University of Khartoum; 2010. [Google scholar]
  279. Yagi S, Drouart N, Bourgaud F, Henry M, Chapleur Y, Laurain-Mattar D. Antioxidant and antiglycation properties of Hydnora johannis roots. S Afr J Bot. 2013; 84: 124-127. [CrossRef] [Google scholar]
  280. Yagi S, Yagi AI, Gadir EA, Henry M, Chapleur Y, Laurain-Mattar D. Toxicity of Hydnora johannis Becca. dried roots and ethanol extract in rats. J Ethnopharmacol. 2011; 137: 796-801. [CrossRef] [Google scholar] [PubMed]
  281. Koko WS, Mesaik MA, Ranjitt R, Galal M, Choudhary MI. Immunosuppressive phenolic compounds from Hydnora abyssinica A. Braun. BMC Complement Altern Med. 2015; 15: 400. [CrossRef] [Google scholar] [PubMed]
  282. Onyancha JM, Cherongis CN, Nzivo JM. Phytochemical screening and evaluation of antioxidant activity of methanolic extract of Kenyan Hydnora abyssinica A. Braun (Hydnoraceae). J Innov Pharm Biolog Sci. 2015; 2: 1-6. [Google scholar]
  283. Onyancha JM, Gikonyo NK, Wachira SW, Mwitari PG, Gicheru MM. Anticancer activities and safety evaluation of selected Kenyan plant extracts against breast cancer cell lines. J Pharmacogn Phytother. 2018; 10: 21-26. [CrossRef] [Google scholar]
  284. Al-Fatimi M, Ali NA, Kilian N, Franke K, Arnold N, Kuhnt C, et al. Ethnobotany, chemical constituents and biological activities of the flowers of Hydnora abyssinica A. Br. (Hydnoraceae). Pharmazie. 2016; 71: 222-226. [Google scholar]
  285. Nghinaunye T. Phytochemical, antimicrobial and cytotoxicity evaluation of rhizome extracts of Hydnora Abyssinica from Acacia Nigrences host. Windhoek, Namibia: University of Namibia; 2019. [Google scholar]
  286. Abdallah HM, Abdel-Naim AB, Ashour OM, Shehata IA, Abdel-Sattar EA. Anti-inflammatory activity of selected plants from Saudi Arabia. Z Naturforsch C. 2014; 69: 1-9. [CrossRef] [Google scholar]
  287. D’Ambola M, Bosco A, Ariano A, Rinaldi L, Bader A, Amadesi A, et al. In vitro anthelminthic efficacy of Hypoestes forskaolii (Vahl) R. Br (Acanthaceae) extracts on gastrointestinal nematodes of sheep. Vet Sci. 2018; 5: 89. [CrossRef] [Google scholar] [PubMed]
  288. Sillo AJ, Makirita WE, Swai H, Chacha M. Larvicidal activity of Hypoestes forskaolii (Vahl) R. Br root extracts against Anopheles gambiae Giless. s, Aedes aegypti L, and Culex quinquefasciatus Say. J Expl Pharmacol. 2019; 11: 23-27. [CrossRef] [Google scholar] [PubMed]
  289. Misganaw D, Amare GG, Mengistu G. Chemo suppressive and curative potential of Hypoestes forskalei against Plasmodium berghei: Evidence for in vivo antimalarial activity. J Exp Pharmacol. 2020; 12: 313-323. [CrossRef] [Google scholar] [PubMed]
  290. Costa SS, Muzitano MF, Camargo LM, Coutinho MA. Therapeutic potential of Kalanchoe species: Flavonoids and other secondary metabolites. Nat Prod Commun. 2008; 3. doi: 10.1177/1934578X0800301236. [CrossRef] [Google scholar]
  291. Akah PA. Antidiarrheal activity of Kigelia africana in experimental animals. J Herbs Spices Med Plants. 1996; 4: 31-38. [CrossRef] [Google scholar]
  292. Bello I, Shehu MW, Musa M, Asmawi MZ, Mahmud R. Kigelia africana (Lam.) Benth. (Sausage tree): Phytochemistry and pharmacological review of a quintessential African traditional medicinal plant. J Ethnopharmacol. 2016; 189: 253-276. [CrossRef] [Google scholar] [PubMed]
  293. Recio MC, Giner RM, Manez S, Rios JL, Marston A, Hostettmann K. Screening of tropical medicinal plants for antiinflammatory activity. Phytother Res. 1995; 9: 571-574. [CrossRef] [Google scholar]
  294. Maroyi A. Kirkia acuminata Oliv.: A review of its ethnobotany and pharmacology. Afr J Tradit Complement Altern Med. 2017; 14: 217-226. [CrossRef] [Google scholar] [PubMed]
  295. Maroyi A. Ethnomedicinal uses, phytochemistry and pharmacological properties of the genus, Kirkia. Trop J Pharm Res. 2016; 15: 2507-2516. [CrossRef] [Google scholar]
  296. Chakuma N, Chipurura B, Muchuweti M, Chitindingu K, Bhebhe M, Chagonda L. Total phenolic content, free radical scavenging and antioxidant potential of Lannea discolor (Sond.) Engl bark and root extracts. J Biol Act Prod Nat. 2015; 5: 71-77. [CrossRef] [Google scholar]
  297. Kabongo‐Kayoka PN, Eloff JN, Obi CL, McGaw LJ. Antimycobacterial activity and low cytotoxicity of leaf extracts of some African Anacardiaceae tree species. Phytother Res. 2016; 30: 2001-2011. [CrossRef] [Google scholar] [PubMed]
  298. Maroyi A. Lannea discolor: Its botany, ethnomedicinal uses, phytochemistry, and pharmacological properties. Asian J Pharm Clin Res. 2018; 11: 49. [CrossRef] [Google scholar]
  299. Sigidi MT, Traoré AN, Boukandou MM, Tshisikhawe MP, Ntuli SS, Potgieter N. Anti-HIV, pro-inflammatory and cytotoxicity properties of selected Venda plants. Indian J Tradit Knowl. 2017; 16: 545-552. [Google scholar]
  300. Banda M, Nyirenda J, Muzandu K, Sijumbila G, Mudenda S. Antihyperglycemic and antihyperlipidemic effects of aqueous extracts of Lannea edulis in alloxan-induced diabetic rats. Front Pharmacol. 2018; 9: 1099. [CrossRef] [Google scholar] [PubMed]
  301. Pour BM, Sasidharan S. In vivo toxicity study of Lantana camara. Asian Pac J Trop Biomed. 2011; 1: 230-232. [CrossRef] [Google scholar] [PubMed]
  302. Patel J, Qureshi S, Kumar GS, Kumar DB, Kumar KA. Phytochemicals and pharmacological activities of Lantana Camara Linn. Res J Pharmacol Pharmacodyn. 2010; 2: 418-422. [Google scholar]
  303. Ved A, Arsi T, Prakash O, Gupta A. A review on phytochemistry and pharmacological activity of Lantana camara Linn. Int J Pharm Sci Res. 2018; 9: 37-43. [Google scholar]
  304. Shah M, Alharby HF, Hakeem KR. Lantana camara: A comprehensive review on phytochemistry, ethnopharmacology and essential oil composition. Lett Appl NanoBioSci. 2020; 9: 1199-1207. [CrossRef] [Google scholar]
  305. McGaw LJ, Eloff JN, Meyer JJ. Screening of 16 poisonous plants for antibacterial, anthelmintic and cytotoxic activity in vitro. S Afr J Bot. 2005; 71: 302-306. [CrossRef] [Google scholar]
  306. Makhafola MA, Middleton L, Olivier MT, Olaokun OO. Cytotoxic and antibacterial activity of selected medicinal plants used in South African traditional medicine. Asian J Chem. 2019; 31: 2623-2627. [CrossRef] [Google scholar]
  307. Habtemariam S, Gray AI, Waterman PG. Diterpenes from the leaves of Leonotis ocymifolia var. raineriana. J Nat Prod. 1994; 57: 1570-1574. [CrossRef] [Google scholar]
  308. Vagionas K, Graikou K, Chinou IB, Runyoro D, Ngassapa O. Chemical analysis and antimicrobial activity of essential oils from the aromatic plants Artemisia afra jacq. and Leonotis ocymifolia (Burm. F.) iwarsson var. raineriana (vision1) Iwarsson growing in Tanzania. J Essent Oil Res. 2007; 19: 396-400. [CrossRef] [Google scholar]
  309. Eguale T, Tadesse D, Giday M. In vitro anthelmintic activity of crude extracts of five medicinal plants against egg-hatching and larval development of Haemonchus contortus. J Ethnopharmacol. 2011; 137: 108-113. [CrossRef] [Google scholar] [PubMed]
  310. Alemu A, Tamiru W, Nedi T, Shibeshi W. Analgesic and Anti-Inflammatory Effects of 80% Methanol Extract of Leonotis ocymifolia (Burm. f.) Iwarsson Leaves in Rodent Models. Evid Based Complementary Altern Med. 2018; 2018: 1614793. [CrossRef] [Google scholar] [PubMed]
  311. Teklu T, Engidawork E, Nedi T, Teklehaymanot T, Gebremeskel L. Evaluation of the antimalarial activity of the hydroalcoholic extract of leaf of leonotis ocymifolia (Burm. f.) Iwarsson (Lamiaceae) against plasmodium berghei in mice. Evid Based Complementary Altern Med. 2020; 2020: 5384804. [CrossRef] [Google scholar] [PubMed]
  312. Endris A, Asfaw N, Bisrat D. Chemical composition, antimicrobial and antioxidant activities of the essential oil of Lippia javanica leaves from Ethiopia. J Essent Oil Res. 2016; 28: 221-226. [CrossRef] [Google scholar]
  313. Osunsanmi FO, Zharare GE, Opoku AR. Phytochemical constituents and antioxidant potential of crude extracts from Lippia Javanica (Burm. f.) Spreng Leaves. Pharmacogn J. 2019; 11: 803-807. [CrossRef] [Google scholar]
  314. Wijaya S, Jin KT, Nee TK, Wiart C. In vitro 5-LOX inhibitory and antioxidant activities of extracts and compounds from the aerial parts of Lopholaena coriifolia (Sond.) E. Phillips & CA Sm. J Complement Integr Med. 2012; 9. doi: 10.1515/1553-3840.1615. [CrossRef] [Google scholar] [PubMed]
  315. Kenéz Á, Lestár Z, Lenkey B, Antus S. Synthesis and structure-activity relationship study of monotesone-A, an antifungal component of Monotes engleri. Nat Prod Res. 2008; 22: 383-392. [CrossRef] [Google scholar] [PubMed]
  316. Viljoen AM, Klepser ME, Ernst EJ, Keele D, Roling E, Van Vuuren S, et al. The composition and antimicrobial activity of the essential oil of the resurrection plant Myrothamnus flabellifolius. S Afr J Bot. 2002; 68: 100-105. [CrossRef] [Google scholar]
  317. Bussmann RW, Malca G, Glenn A, Sharon D, Nilsen B, Parris B, et al. Toxicity of medicinal plants used in traditional medicine in Northern Peru. J Ethnopharmacol. 2011; 137: 121-140. [CrossRef] [Google scholar] [PubMed]
  318. Erhabor JO, Komakech R, Kang Y, Tang M, Matsabisa MG. Ethnopharmacological importance and medical applications of Myrothamnus flabellifolius Welw. (Myrothamnaceae)-A review. J Ethnopharmacol. 2020; 252: 112576. [CrossRef] [Google scholar] [PubMed]
  319. Kwape TE, Majinda RR, Chaturvedi P. Antioxidant and antidiabetic potential of Myrothamnus flabellifolius found in Botswana. Cogent Biol. 2016; 2: 1275403. [CrossRef] [Google scholar]
  320. Iqbal Z, Lateef M, Jabbar A, Ghayur MN, Gilani AH. In vitro and in vivo anthelmintic activity of Nicotiana tabacum L. leaves against gastrointestinal nematodes of sheep. Phytother Res. 2006; 20: 46-48. [CrossRef] [Google scholar] [PubMed]
  321. Shang SZ, Zhao W, Tang JG, Xu XM, Sun HD, Pu JX, et al. Antiviral sesquiterpenes from leaves of Nicotiana tabacum. Fitoterapia. 2016; 108: 1-4. [CrossRef] [Google scholar] [PubMed]
  322. Ameya G, Manilal A, Merdekios B. In vitro antibacterial activity and phytochemical analysis of Nicotiana tabacum L. extracted in different organic solvents. Open Microbiol J. 2017; 11: 352. [CrossRef] [Google scholar] [PubMed]
  323. Batoro J, Ekowati G. An ethnobotanical tobacco (Nicotiana tabacum L.) in Indonesia. A review. Adv Life Sci. 2017; 7: 26-29. [Google scholar]
  324. Khaleel RI. Bio-toxicity study of some selected plant by Artemia salina (Leach) test. Plant Arch. 2019; 19: 2847-2850. [Google scholar]
  325. Popova V, Tumbarski Y, Ivanova T, Hadjikinova R, Stoyanova A. Tobacco resinoid (Nicotiana tabacum L.) as an active ingredient of cosmetic gels. J Appl Pharm Sci. 2019; 9: 111-118. [CrossRef] [Google scholar]
  326. Ajani RS, Balogun OT. Nicotiana tabacum (Tobacco) promotes wound healing in diabetic rats. J Complement Altern Med Res. 2020; 9: 31-45. [CrossRef] [Google scholar]
  327. Sulaiman FA, Nafiu MO, Yusuf BO, Muritala HF, Adeyemi SB, Omar SA, et al. The GC-MS fingerprints of Nicotiana tabacum L. extract and propensity for renal impairment and modulation of serum triglycerides in Wistar rats. J Pharm Pharmacogn Res. 2020; 8: 191-200. [CrossRef] [Google scholar]
  328. Naidoo Y, Sadashiva CT, Kasim N, Nicholas A, Naidoo G. Chemical composition and antimicrobial activity of the essential oil of Ocimum obovatum E. Mey. Ex Benth. (Lamiaceae). J Essent Oil Bear Plants. 2014; 17: 142-147. [CrossRef] [Google scholar]
  329. Naidoo Y, Sadashiva CT, Naidoo G, Raghu K. Antibacterial, antioxidant and phytochemical properties of the ethanolic extract of Ocimum obovatum E. Mey. ex Benth. Indian J Tradit Knowl. 2016; 15: 57-61. [Google scholar]
  330. Ogbonnia SO, Mbaka GO, Anyika EN, Ladiju O, Igbokwe HN, Emordi JE, et al. Evaluation of anti-diabetics and cardiovascular effects of Parinari curatellifolia seed extract and Anthoclista vogelli root extract individually and combined on postprandial and alloxan-induced diabetic albino rats. J Adv Med Med Res. 2011; 4: 146-162. [CrossRef] [Google scholar]
  331. Olaleye MT, Amobonye AE, Komolafe K, Akinmoladun AC. Protective effects of Parinari curatellifolia flavonoids against acetaminophen-induced hepatic necrosis in rats. Saudi J Biol Sci. 2014; 21: 486-492. [CrossRef] [Google scholar] [PubMed]
  332. Gweshelo D, Muswe R, Mukanganyama S. In vivo and in vitro inhibition of rat liver glutathione transferases activity by extracts from Combretum zeyheri (Combretaceae) and Parinari curatellifolia (Chrysobalanaceae). BMC Complement Altern Med. 2016; 16: 238. [CrossRef] [Google scholar] [PubMed]
  333. Mbunde MV, Innocent E, Mabiki F, Andersson PG. Ethnobotanical survey and toxicity evaluation of medicinal plants used for fungal remedy in the Southern Highlands of Tanzania. J Intercult Ethnopharmacol. 2016; 6: 84. [CrossRef] [Google scholar] [PubMed]
  334. Osafo N, Boakye YD, Agyare C, Obeng S, Foli JE, Minkah PA. African plants with antiproliferative properties. In: Natural Products and Cancer Drug Discovery. IntechOpen; 2017. pp. 1-23. [CrossRef] [Google scholar]
  335. Halilu EM, October N, Ugwah-Oguejiofor CJ, Jega AY, Nefai MS. Anti-snake venom and analgesic activities of extracts and betulinic and oleanolic acids isolated from Parinari curatellifolia. J Med Plants Econ Dev. 2020; 4: 1-8. Available from: https://hdl.handle.net/10520/ejc-jomped-v4-n1-a3. [CrossRef]
  336. Josiah SS, Oyeleye SI, Crown OO, Olaleye MT. Ameliorative effect of Parinari curatellifolia seed extracts on sodium nitroprusside–induced cardiovascular toxicity in rats. Comp Clin Pathol. 2020; 29: 239-246. [CrossRef] [Google scholar]
  337. Peni IJ, Elinge CM, Yusuf H, Itodo AU, Agaie BM, Mbongo AN, et al. Phytochemical screening and antibacterial activity of Parinari curatellifolia stem extract. J Med Plants Res. 2010; 4: 2099-2102. [Google scholar]
  338. McGaw LJ, Van der Merwe D, Eloff JN. In vitro anthelmintic, antibacterial and cytotoxic effects of extracts from plants used in South African ethnoveterinary medicine. Vet J. 2007; 173: 366-372. [CrossRef] [Google scholar] [PubMed]
  339. Sewani-Rusike CR. Antifertility effects of Pouzolzia mixta in female wistar rats. Afr J Tradit Complement Altern Med. 2013; 10: 526-532. [CrossRef] [Google scholar]
  340. Samie A, Tambani T, Harshfield E, Green E, Ramalivhana JN, Bessong PO. Antifungal activities of selected Venda medicinal plants against Candida albicans, Candida krusei and Cryptococcus neoformans isolated from South African AIDS patients. Afr J Biotechnol. 2010; 9: 2965-2976. [Google scholar]
  341. Motlhanka DM. Polyphenolic content and antioxidant analysis of Pseudolachnostylis maprouneifolia Pax Var dikindtii used as livestock feed by farmers from Eastern Botswana. J Pharm Sci Innov. 2012; 1: 51-57. [Google scholar]
  342. Lawal F, Bapela MJ, Adebayo SA, Nkadimeng SM, Yusuf AA, Malterud KE, et al. Anti-inflammatory potential of South African medicinal plants used for the treatment of sexually transmitted infections. S Afr J Bot. 2019; 125: 62-71. [CrossRef] [Google scholar]
  343. Ojewole JA. Hypoglycemic and hypotensive effects of Psidium guajava Linn. (Myrtaceae) leaf aqueous extract. Methods Findings Exp Clin Pharmacol. 2005; 27: 689-695. [CrossRef] [Google scholar] [PubMed]
  344. Ojewole JAO. Antiinflammatory and Analgesic Effects of Psidium guajava Linn. (Myrtaceae) Leaf Aqueous Extract in Rats and Mice. Methods Find Exp Clin Pharmacol. 2006; 28: 441-446. [CrossRef] [Google scholar] [PubMed]
  345. Akinola O, OB O, Dosumu OO. Ethanol extract of the leaves of Psidium guajava Linn enhances sperm output in healthy Wistar rats. Afr J Med Med Sci. 2007; 36: 137-140. [Google scholar]
  346. Chen HY, Yen GC. Antioxidant activity and free radical-scavenging capacity of extracts from guava (Psidium guajava L.) leaves. Food Chem. 2007; 101: 686-694. [CrossRef] [Google scholar]
  347. Choi SY, Hwang JH, Park SY, Jin YJ, Ko HC, Moon SW, et al. Fermented guava leaf extract inhibits LPS-induced COX-2 and iNOS expression in Mouse macrophage cells by inhibition of transcription factor NF-κB. Phytother Res. 2008; 22: 1030-1034. [CrossRef] [Google scholar] [PubMed]
  348. Mittal P, Gupta V, Kaur G, Garg AK, Singh A. Phytochemistry and pharmacological activities of Psidium guajava. Int J Pharm Sci Res. 2010; 1: 9-19. [Google scholar]
  349. Jang M, Jeong SW, Cho SK, Ahn KS, Lee JH, Yang DC, et al. Anti-inflammatory effects of an ethanolic extract of guava (Psidium guajava L.) leaves in vitro and in vivo. J Med Food. 2014; 17: 678-685. [CrossRef] [Google scholar] [PubMed]
  350. Fernandes KP, Bussadori SK, Marques MM, Wadt NS, Bach E, Martins MD. Healing and cytotoxic effects of Psidium guajava (Myrtaceae) leaf extracts. Braz J Oral Sci. 2010; 9: 449-454. [Google scholar]
  351. Kafle A, Mohapatra SS, Reddy I, Chapagain M. A review on medicinal properties of Psidium guajava. J Med Plants Stud. 2018; 6: 44-47. [Google scholar]
  352. Daswani PG, Gholkar MS, Birdi TJ. Psidium guajava: A single plant for multiple health problems of rural Indian population. Pharmacogn Rev. 2017; 11: 167. [CrossRef] [Google scholar] [PubMed]
  353. Delorino SB. Histologic property of Psidium guajava Linn Guava leaf extract in wound. Eur J Mol Clin Med. 2020; 7: 1385-1391. [Google scholar]
  354. Delorino SB, Ogalesco ML, Rebadulla KR, Rongcales MT, Salubre JI, Talacay MK, et al. Wound healing efficacy of guava leaf extract. J Pharm Res Int. 2020; 32: 27-35. [CrossRef] [Google scholar]
  355. Epifano F, Fiorito S, Genovese S. Phytochemistry and pharmacognosy of the genus Acronychia. Phytochemistry. 2013; 95: 12-18. [CrossRef] [Google scholar] [PubMed]
  356. Elufioye TO, Bamgbose MO, Alabi SO. Evaluation of antioxidant and antiacne activity of Psorospermum febrifugum (Spach) and Psorospermum corymbiferum (Hochr.). Br J Pharm Res. 2016; 12: BJPR.27984. [CrossRef] [Google scholar]
  357. Agbogba F, Senou M, Tchogou AP, Lokonon JE, Sacramento TI, Medoatinsa E, et al. Ethyl acetate fraction of Psorospermum febrifugum Spach aqueous extract did not exhibit acute or sub-chronic toxicity. experimental study on Wistar rats. Int J Biosci. 2019; 14: 475-482. [CrossRef] [Google scholar]
  358. Félicienne A, Isabelle ST, Pascal TA, Espérance M, Rose KE, Eugène A, et al. The aqueous extract of the root bark of Psorospermum febrifugum Spach effectively corrects anaemia. Experimental study on Wistar rats. J Appl Biosci. 2019; 139: 14137-14146. [CrossRef] [Google scholar]
  359. Asogwa FC, Asogwa CJ, Okoye CO. Anti-inflammatory properties of Psoropermum febrifugum stem bark ethanol extract. IDOSR J Appl Sci. 2017; 2: 95-105. [Google scholar]
  360. Asogwa FC, Ibezim A, Ntie-Kang F, Asogwa CJ, Okoye CO. Anti-psoriatic and immunomodulatory evaluation of psorospermum febrifugum spach and its phytochemicals. Sci Afr. 2020; 7: e00229. [CrossRef] [Google scholar]
  361. Zininga T, Anokwuru CP, Sigidi MT, Tshisikhawe MP, Ramaite II, Traoré AN, et al. Extracts obtained from Pterocarpus angolensis DC and Ziziphus mucronata exhibit antiplasmodial activity and inhibit heat shock protein 70 (Hsp70) function. Molecules. 2017; 22: 1224. [CrossRef] [Google scholar] [PubMed]
  362. Chipinga JV. Efficacy of Pterocarpus angolensis crude extracts against Candida krusei, Staphylococcus aureus, Streptococcus agalactiae and Escherichia coli. Malawi Med J. 2018; 30: 219-224. [CrossRef] [Google scholar] [PubMed]
  363. Okeleye BI, Mkwetshana NT, Ndip RN. Evaluation of the antibacterial and antifungal potential of Peltophorum africanum: Toxicological effect on human chang liver cell line. Sci World J. 2013; 2013: 878735. [CrossRef] [Google scholar] [PubMed]
  364. Mazimba O. Pharmacology and phytochemistry studies in Peltophorum africanum. Bull Fac Pharm Cairo Univ. 2014; 52: 145-153. [CrossRef] [Google scholar]
  365. Sigidi MT, Anokwuru CP, Zininga T, Tshisikhawe MP, Shonhai A, Ramaite ID, et al. Comparative in vitro cytotoxic, anti-inflammatory and anti-microbiological activities of two indigenous Venda medicinal plants. Transl Med Commun. 2016; 1: 9. [CrossRef] [Google scholar]
  366. Santos ES, Luís Â, Gonçalves J, Rosado T, Pereira L, Gallardo E, et al. Julbernardia paniculata and Pterocarpus angolensis: From ethnobotanical surveys to phytochemical characterization and bioactivities evaluation. Molecules. 2020; 25: 1828. [CrossRef] [Google scholar] [PubMed]
  367. Burgess EP, Koha EM, Hutchins RF, Douglas L. Toxicity of leaves from the castor oil plant, Ricinus communis L. (Euphorbiaceae), to adult grass grub, Costelytra zealandica (White) (Coleoptera: Scarabaeidae). N Z J Exp Agric. 1988; 16: 63-66. [CrossRef] [Google scholar]
  368. Almeida RN, Navarro DS, Barbosa-Filho JM. Plants with central analgesic activity. Phytomedicine. 2001; 8: 310-322. [CrossRef] [Google scholar] [PubMed]
  369. Ilavarasan R, Mallika M, Venkataraman S. Toxicological assessment of Ricinus communis Linn root extracts. Toxicol Mech Methods. 2011; 21: 246-250. [CrossRef] [Google scholar] [PubMed]
  370. Prasad MK, Rachhadiya RM, Shete RV. Pharmacological investigation on the wound healing effects of castor oil in rats. Int J Univ Pharm Life Sci. 2011; 1: 21-28. [Google scholar]
  371. Bhakta S, Das SK. In praise of the medicinal plant Ricinus communis L.: A review. Glob J Res Med Plants Indig Med. 2015; 4: 95. [Google scholar]
  372. Ahmad N, Mishra A, Ahsan F, Mahmood T, Hasan N, Khan Z. Ricinus communis: Pharmacological actions and marketed medicinal products. World J Pharm Life Sci. 2016; 2: 179-188. [Google scholar]
  373. Kumar M. A review on phytochemical constituents and pharmacological activities of Ricinus communis L. Plant. Int J Pharmacogn Phytochem Res. 2017; 9: 466-472. [CrossRef] [Google scholar]
  374. Tkachenko H, Buyun L, Osadowski Z, Maryniuk M. The antibacterial activity of certain Sansevieria Thunb. species against Escherichia coli. Agrobiodiversity Improv Nutr Health Life Qual. 2017. Available from: https://agrobiodiversity.uniag.sk/scientificpapers/article/view/119. [CrossRef]
  375. McGaw LJ, Jäger AK, Van Staden J, Eloff JN. Variation in antibacterial activity of Schotia species. S Afr J Bot. 2002; 68: 41-46. [CrossRef] [Google scholar]
  376. McGaw LJ, Bagla VP, Mokoka TA, Elgorashi EE, Eloff JN. South African ethnoveterinary plant extracts with antimicrobial and antiviral potential. Afr J Tradit Complement Altern Med. 2009; 6: 472. [Google scholar]
  377. Mathabe MC, Nikolova RV, Lall N, Nyazema NZ. Antibacterial activities of medicinal plants used for the treatment of diarrhoea in Limpopo Province, South Africa. J Ethnopharmacol. 2006; 105: 286-293. [CrossRef] [Google scholar] [PubMed]
  378. Adewusi EA, Moodley N, Steenkamp V. Antioxidant and acetylcholinesterase inhibitory activity of selected southern African medicinal plants. S Afr J Bot. 2011; 77: 638-644. [CrossRef] [Google scholar]
  379. Du K, Marston A, van Vuuren SF, van Zyl RL, Coleman C, Zietsman PC, et al. Flavonolacyl glucosides from the aril of Schotia brachypetala Sond. and their antioxidant, antibacterial and antimalarial activities. Phytochem Lett. 2014; 10: cxxiii-cxxviii. [CrossRef] [Google scholar]
  380. Hassaan Y, Handoussa H, El-Khatib AH, Linscheid MW, El Sayed N, Ayoub N. Evaluation of plant phenolic metabolites as a source of Alzheimer’s drug leads. BioMed Res Int. 2014; 2014: 843263. [CrossRef] [Google scholar] [PubMed]
  381. Sobeh M, ElHawary E, Peixoto H, Labib RM, Handoussa H, Swilam N, et al. Identification of phenolic secondary metabolites from Schotia brachypetala Sond. (Fabaceae) and demonstration of their antioxidant activities in Caenorhabditis elegans. PeerJ. 2016; 4: e2404. [CrossRef] [Google scholar] [PubMed]
  382. Ojewole JA. Analgesic, anti-inflammatory and hypoglycaemic effects of Rhus chirindensis (Baker F.) [Anacardiaceae] stem-bark aqueous extract in mice and rats. J Ethnopharmacol. 2007; 113: 338-345. [CrossRef] [Google scholar] [PubMed]
  383. Moosa ZM, Mabandla M. The effect of Searsia chirindensis stem-bark extract on renal and liver function in a rat model of neurotoxicity. Biomed Pharmacother. 2017; 86: 368-372. [CrossRef] [Google scholar] [PubMed]
  384. Gundidza M, Gweru N, Mmbengwa V, Ramalivhana NJ, Magwa Z, Samie A. Phytoconstituents and biological activities of essential Oil from Rhus lancea L. F. Afr J Biotechnol. 2008; 7: 2787-2789. [Google scholar]
  385. Mulaudzi RB, Ndhlala AR, Kulkarni MG, Van Staden J. Pharmacological properties and protein binding capacity of phenolic extracts of some Venda medicinal plants used against cough and fever. J Ethnopharmacol. 2012; 143: 185-193. [CrossRef] [Google scholar] [PubMed]
  386. Madzinga M, Kritzinger Q. Searsia lancea. In: Underexplored Medicinal Plants from Sub-Saharan Africa. San Diego, CA: Academic Press; 2020. pp. 253-259. [CrossRef] [Google scholar]
  387. Songca SP, Sebothoma C, Samuel BB, Eloff JN. A biflavonoid and a carotenoid from Rhus leptodictya: Isolation, characterization and antibacterial properties. Afr J Biochem Res. 2012; 6: 172-178. [Google scholar]
  388. Ahmed AS, McGaw LJ, Moodley N, Naidoo V, Eloff JN. Cytotoxic, antimicrobial, antioxidant, antilipoxygenase activities and phenolic composition of Ozoroa and Searsia species (Anacardiaceae) used in South African traditional medicine for treating diarrhoea. S Afr J Bot. 2014; 95: 9-18. [CrossRef] [Google scholar]
  389. Matamela T, Green IR, Mtunzi FM. A novel biflavonoid from Rhus leptodictya. Nat Prod Commun. 2016; 11. doi: 10.1177/1934578X1601100922. [CrossRef] [Google scholar]
  390. Mtunzi FM, Ejidike IP, Matamela T, Dikio E, Klink MJ. Phytochemical profiling, antioxidant and antibacterial activities of leaf extracts from Rhus leptodictya. Int J Pharmacogn Phytochem Res. 2017; 9: 1090-1099. [CrossRef] [Google scholar]
  391. Okoli CO, Akah PA, Ezugworie U. Anti-inflammatory activity of extracts of root bark of Securidaca longipedunculata Fres (Polygalaceae). Afr J Tradit Complement Altern Med. 2006; 3: 54-63. [CrossRef] [Google scholar]
  392. Muanda FN, Dicko A, Soulimani R. Assessment of polyphenolic compounds, in vitro antioxidant and anti-inflammation properties of Securidaca longepedunculata root barks. C R Biol. 2010; 333: 663-669. [CrossRef] [Google scholar] [PubMed]
  393. Sanusi J, Jibia AB, Runka JY, Liadi S, Abubakar AA, Zurmi RU. Antimicrobial activity of aqueous and ethanol extracts of violet plant (Securidaca longipedunculata Fres) on tested pathogenic bacteria. Int J Pharm Sci Res. 2015; 6: 3276-3284. [Google scholar]
  394. Nguta JM. In Vivo Antimalarial Activity, Toxicity, and Phytochemical Composition of Total Extracts from Securidaca longepedunculata Fresen. (Polygalaceae). Biomed Biotechnol Res J. 2019; 3: 196-201. [CrossRef] [Google scholar]
  395. Mwonjoria J, Ngeranwa J, Kariuki H, Githinji C, Sagini M, Wambugu S. Ethno medicinal, phytochemical and pharmacological aspects of Solanum incanum (lin.). Int J Pharmacol Toxicol. 2014; 2: 17-20. [CrossRef] [Google scholar]
  396. Dakone D, Guadie A. A review on ethnomedicinal use, nutritional value, phytochemistry and pharmacological characteristics of Solanum incanum L. An important medicinal plant. Int J Sci Technol Res. 2016; 5: 350-354. [Google scholar]
  397. Doyo D. Evaluation of wound healing and anti-inflammatory activity of 80% methanolic extract of Solanum Incanum Linnaeus (Solanaceae) leaves in mice. Addis Ababa, Ethiopia: Addis Ababa University; 2016. [Google scholar]
  398. Masondo NA, Stafford GI, Aremu AO, Makunga NP. Acetylcholinesterase inhibitors from southern African plants: An overview of ethnobotanical, pharmacological potential and phytochemical research including and beyond Alzheimer's disease treatment. S Afr J Bot. 2019; 120: 39-64. [CrossRef] [Google scholar]
  399. Qureshi Z, Khan T, Shah AJ, Wahid F. Solanum incanum extract enhances wound healing and tissue regeneration in burn mice model. Bangladesh J Pharmacol. 2019; 14: 101-106. [CrossRef] [Google scholar]
  400. Ngezahayo J, Ribeiro SO, Fontaine V, Hari L, Stévigny C, Duez P. In vitro study of five herbs used against microbial infections in Burundi. Phytother Res. 2017; 31: 1571-1578. [CrossRef] [Google scholar] [PubMed]
  401. Samie A, Obi CL, Bessong PO, Namrita L. Activity profiles of fourteen selected medicinal plants from Rural Venda communities in South Africa against fifteen clinical bacterial species. Afr J Biotechnol. 2005; 4: 101-106. [Google scholar]
  402. Van Vuuren SF, Naidoo D. An antimicrobial investigation of plants used traditionally in southern Africa to treat sexually transmitted infections. J Ethnopharmacol. 2010; 130: 552-558. [CrossRef] [Google scholar] [PubMed]
  403. Deliwe M, Amabeoku GJ. Evaluation of the antidiarrhoeal and antidiabetic activities of the leaf aqueous extract of Syzygium cordatum Hoscht. ex C. Krauss (Mytraceae) in rodents. Int J Pharmacol. 2013; 9: 125-133. [CrossRef] [Google scholar]
  404. Cordier W, Gulumian M, Cromarty AD, Steenkamp V. Attenuation of oxidative stress in U937 cells by polyphenolic-rich bark fractions of Burkea africana and Syzygium cordatum. BMC Complement Altern Med. 2013; 13: 116. [CrossRef] [Google scholar] [PubMed]
  405. Mzindle NB. Anti-inflammatory, anti-oxidant and wound-healing properties of selected South Africa medicinal plants. Durban, South Africa: Durban University of Technology; 2017. [Google scholar]
  406. Maroyi A. Alternative medicines for HIV/AIDS in resource-poor settings: Insight from traditional medicines use in sub-Saharan Africa. Trop J Pharm Res. 2014; 13: 1527-1536. [CrossRef] [Google scholar]
  407. Maroyi A. Syzygium cordatum hochst. Ex krauss: An overview of its ethnobotany, phytochemistry and pharmacological properties. Molecules. 2018; 23: 1084. [CrossRef] [Google scholar] [PubMed]
  408. Martijena ID, Garcia DA, Marin RH, Perillo MA, Zygadlo JP. Anxiogenic-like and antidepressant-like effects of the essential oil from Tagetes minuta. Fitoterapia. 1998; 69: 155-160. [Google scholar]
  409. Marin RH, Garcia DA, Martijena ID, Zygadlo JA, Arce A, Perillo MA. Anxiogenic-like effects of Tagetes minuta L essential oil on T-maze and tonic immobility behaviour in domestic chicks. Fundam Clin Pharmacol. 1998; 12: 426-432. [CrossRef] [Google scholar] [PubMed]
  410. Lacroix D, Prado S, Kamoga D, Kasenene J, Namukobe J, Krief S,et al. Antiplasmodial and cytotoxic activities of medicinal plants traditionally used in the village of Kiohima, Uganda. J Ethnopharmacol. 2011; 133: 850-855. [CrossRef] [Google scholar] [PubMed]
  411. Ali NA, Sharopov FS, Al-Kaf AG, Hill GM, Arnold N, Al-Sokari SS, et al. Composition of essential oil from Tagetes minuta and its cytotoxic, antioxidant and antimicrobial activities. Nat Prod Commun. 2014; 9. doi: 10.1177/1934578X1400900233. [CrossRef] [Google scholar]
  412. Shahzadi I, Shah MM. Acylated flavonol glycosides from Tagetes minuta with antibacterial activity. Front Pharmacol. 2015; 6: 195. [CrossRef] [Google scholar] [PubMed]
  413. Wanzala W, Wagacha JM, Dossaji SF, Gakuubi MM. Bioactive properties of Tagetes minuta L. (Asteraceae) essential oils: A review. Am J Essent Oils Nat Prod. 2016; 4: 27-36. [Google scholar]
  414. Dos Santos DC, Schneider LR, da Silva Barboza A, Campos AD, Lund RG. Systematic review and technological overview of the antimicrobial activity of Tagetes minuta and future perspectives. J Ethnopharmacol. 2017; 208: 8-15. [CrossRef] [Google scholar] [PubMed]
  415. Karimian P, Kavoosi G, Amirghofran Z. Anti–oxidative and anti–inflammatory effects of Tagetes minuta essential oil in activated macrophages. Asian Pac J Trop Biomed. 2014; 4: 219-227. [CrossRef] [Google scholar] [PubMed]
  416. Bessong PO, Obi CL, Igumbor E, Andreola ML, Litvak S. In vitro activity of three selected South African medicinal plants against human immunodeficiency virus type 1 reverse transcriptase. Afr J Biotechnol. 2004; 3: 555-559. [CrossRef] [Google scholar]
  417. Moshi MJ, Mbwambo ZH. Some pharmacological properties of extracts of Terminalia sericea roots. J Ethnopharmacol. 2005; 97: 43-47. [CrossRef] [Google scholar] [PubMed]
  418. Masoko P, Eloff JN. Screening of twenty-four South African Combretum and six Terminalia species (Combretaceae) for antioxidant activities. Afr J Tradit Complement Altern Med. 2007; 4: 231-239. [CrossRef] [Google scholar] [PubMed]
  419. Nair AA, Anjum N, Tripathi YC. A review on ethnomedicinal, phytochemical, and pharmacological significance of Terminalia sericea Burch. Ex DC. J Pharm Res. 2018; 12: 420-430. [Google scholar]
  420. Maregesi SM, Nyamwisenda NT, Mwangomo D, Kidukuli A. In vitro antimicrobial activity and determination of essential metal and ash value contents of Trichodesma zeylanicum. Int J Res Pharmacol Pharmacerapeutic. 2013; 2: 417-424. [Google scholar]
  421. Ngonda F. In-vitro anti-oxidant activity and free radical scavenging potential of roots of Malawian Trichodesma zeylanicumm (Burm. f.). Asian J Biomed Pharm Sci. 2013; 3: 21. [Google scholar]
  422. Ngonda F. Evaluation of the wound healing potential of Trichodesma zeylanicum (Burm. f.) formulation in excision wounds in albino rats. Annu Res Rev Biol. 2014; 4: 828-839. [CrossRef] [Google scholar]
  423. Moyo B, Mukanganyama S. Antibacterial effects of Cissus welwitschii and Triumfetta welwitschii extracts against Escherichia coli and Bacillus cereus. Int J Bacteriol. 2015; 2015: 162028. [CrossRef] [Google scholar] [PubMed]
  424. Moyo B, Mukanganyama S. Antiproliferative activity of T. welwitschii extract on Jurkat T cells in vitro. BioMed Res Int. 2015; 2015: 817624. [CrossRef] [Google scholar] [PubMed]
  425. Mombeshora M, Mukanganyama S. Antibacterial activities, proposed mode of action and cytotoxicity of leaf extracts from Triumfetta welwitschii against Pseudomonas aeruginosa. BMC Complement Altern Med. 2019; 19: 315. [CrossRef] [Google scholar] [PubMed]
  426. Johns T, Faubert GM, Kokwaro JO, Mahunnah RL, Kimanani EK. Anti-giardial activity of gastrointestinal remedies of the Luo of East Africa. J Ethnopharmacol. 1995; 46: 17-23. [CrossRef] [Google scholar] [PubMed]
  427. Achola KJ, Mwangi JW, Munenge RW, Mwaura AM. Pharmacological activities of Vernonia glabra. Int J Pharmacogn. 1996; 34: 141-144. [CrossRef] [Google scholar]
  428. Ngonda F, Magombo Z, Mpeketul P, Mwatseteza J. Evaluation of Malawian Vernonia glabra (Steetz) Vatke leaf and Securidaca longepedunculata (Fresen) root extracts for antimicrobial activities. J Appl Pharm Sci. 2012; 2: 26-33. [Google scholar]
  429. Kitonde CK, Fidahusein DS, Lukhoba CW, Jumba MM. Antimicrobial activity and phytochemical study of Vernonia glabra (steetz) oliv. & hiern. In Kenya. Afr J Tradit Complement Altern Med. 2013; 10: 149-157. [CrossRef] [Google scholar]
  430. Mulaudzi RB, Ndhlala AR, Kulkarni MG, Finnie JF, Van Staden J. Antimicrobial properties and phenolic contents of medicinal plants used by the Venda people for conditions related to venereal diseases. J Ethnopharmacol. 2011; 135: 330-337. [CrossRef] [Google scholar] [PubMed]
  431. Munodawafa T, Chagonda LS, Moyo SR. Antimicrobial and phytochemical screening of some Zimbabwean medicinal plants. J Biol Act Prod Nat. 2013; 3: 323-330. [CrossRef] [Google scholar]
  432. Nair JJ, Mulaudzi RB, Chukwujekwu JC, Van Heerden FR, Van Staden J. Antigonococcal activity of Ximenia caffra Sond. (Olacaceae) and identification of the active principle. S Afr J Bot. 2013; 86: 111-115. [CrossRef] [Google scholar]
  433. Zhen J, Guo Y, Villani T, Carr S, Brendler T, Mumbengegwi DR, et al. Phytochemical Analysis and Anti‐Inflammatory Activity of the Extracts of the African Medicinal Plant Ximenia caffra. J Anal Methods Chem. 2015; 2015: 948262. [CrossRef] [Google scholar] [PubMed]
  434. Okokon JE, Nyong ME, Essien GE, Nyong E. Nephroprotective activity of husk extract and fractions of Zea mays against alloxan-induced oxidative stress in diabetic rats. J Basic Pharmacol Toxicol. 2017; 1: 1-10. [Google scholar]
  435. Udobang JA, Okokon JE, Obot D, Agu CE. Hepatoprotective activity of husk extract Zea mays against carbon tetrachloride induced liver injury in rats. Res J Life Sci Bioinf Pharm Chem Sci. 2019; 5: 82-94. [Google scholar]
  436. Kim DY, Won KJ, Hwang DI, Kim HB, Li Y, Lee HM. Migration-and proliferation-promoting activities in human keratinocytes of Zea mays flower absolute and its chemical composition. Chem Biodivers. 2020; 17: e2000227. [CrossRef] [Google scholar] [PubMed]
  437. Magaña Cerino JM, Peniche Pavía HA, Tiessen A, Gurrola Díaz CM. Pigmented maize (Zea mays L.) contains anthocyanins with potential therapeutic action against oxidative stress-A review. Pol J Food Nutr Sci. 2020; 70: 85-99. [CrossRef] [Google scholar]
  438. Abirami S, Priyalakshmi M, Soundariya A, Samrot AV, Saigeetha S, Emilin RR, et al. Antimicrobial activity, antiproliferative activity, amylase inhibitory activity and phytochemical analysis of ethanol extract of corn (Zea mays L.) silk. Curr Res Green Sustain Chem. 2021; 4: 100089. [CrossRef] [Google scholar]
  439. Mascolo N, Jain R, Jain SC, Capasso F. Ethnopharmacologic investigation of ginger (Zingiber officinale). J Ethnopharmacol. 1989; 27: 129-140. [CrossRef] [Google scholar] [PubMed]
  440. Ahmed K, Shaheen G, Asif HM. Zingiber officinale Roscoe (pharmacological activity). J Med Plants Res. 2011; 5: 344-348. [Google scholar] [PubMed]
  441. San Chang J, Wang KC, Yeh CF, Shieh DE, Chiang LC. Fresh ginger (Zingiber officinale) has anti-viral activity against human respiratory syncytial virus in human respiratory tract cell lines. J Ethnopharmacol. 2013; 145: 146-151. [CrossRef] [Google scholar] [PubMed]
  442. Kazerouni A, Kazerouni O, Pazyar N. Effects of ginger (Zingiber officinale) on skin conditions: A non quantitative review article. J Turk Acad Dermatol. 2013; 7: 1372r2. [CrossRef] [Google scholar]
  443. Bellik Y. Total antioxidant activity and antimicrobial potency of the essential oil and oleoresin of Zingiber officinale Roscoe. Asian Pac J Trop Dis. 2014; 4: 40-44. [CrossRef] [Google scholar]
  444. Rahmani AH, Aly SM. Active ingredients of ginger as potential candidates in the prevention and treatment of diseases via modulation of biological activities. Int J Physiol Pathophysiol Pharmacol. 2014; 6: 125. [Google scholar]
  445. Mohamed AH, Osman AA. Antibacterial and wound healing potential of ethanolic extract of Zingiber Officinale in albino rats. J Dis Med Plants. 2017; 3: 1-6. [CrossRef] [Google scholar]
  446. Teles AM, dos Santos BA, Ferreira CG, Mouchreck AN, da Silva Calabrese K, Abreu-Silva AL, et al. Ginger (Zingiber officinale) antimicrobial potential: A review. Ginger cultivation and its antimicrobial and pharmacological potentials. InTech Open; 2019. doi: 10.5772/intechopen.89780. [Google scholar]
  447. Mahboubi M. Zingiber officinale Rosc. essential oil, a review on its composition and bioactivity. Clin Phytosci. 2019; 5: 6. [CrossRef] [Google scholar]
  448. Olajuyigbe OO, Afolayan AJ. Antimicrobial potency of the ethanolic crude bark extract of Ziziphus mucronata Willd. subsp. mucronata Willd. Afr J Pharm Pharmacol. 2012; 6: 724-730. [CrossRef] [Google scholar]
  449. Nemudzivhadi V, Masoko P. Antioxidant and antibacterial properties of Ziziphus mucronata and Ricinus communis leaves extracts. Afr J Tradit Complement Altern Med. 2015; 12: 81-89. [CrossRef] [Google scholar]
  450. Ilonga SK, Kandawa-Schulz MA, Lofty HE, Lyantagaye S. Anticancer, antioxidant and antimicrobial screening of extracts from selected medicinal plants from Oshikoto, Namibia. Bioteknologi. 2018; 15: 55-69. [CrossRef] [Google scholar]
  451. Raina R, Parwez S, Verma PK, Pankaj NK. Medicinal plants and their role in wound healing. Online Vet J. 2008; 3: 21. [Google scholar]
  452. Reuter J, Merfort I, Schempp CM. Botanicals in dermatology: An evidence-based review. Am J Clin Dermatol. 2010; 11: 247-267. [CrossRef] [Google scholar] [PubMed]
  453. Malebo HM, Wiketye V, Katani SJ, Kitufe NA, Nyigo VA, Imeda CP, et al. In vivo antiplasmodial and toxicological effect of Maytenus senegalensis traditionally used in the treatment of malaria in Tanzania. Malar J. 2015; 14: 79. [CrossRef] [Google scholar] [PubMed]
  454. Michalak M. Plant extracts as skin care and therapeutic agents. Int J Mol Sci. 2023; 24: 15444. [CrossRef] [Google scholar] [PubMed]
  455. Michalak M. Plant-derived antioxidants: Significance in skin health and the ageing process. Int J Mol Sci. 2022; 23: 585. [CrossRef] [Google scholar] [PubMed]
  456. Vitale S, Colanero S, Placidi M, Di Emidio G, Tatone C, Amicarelli F, et al. Phytochemistry and biological activity of medicinal plants in wound healing: An overview of current research. Molecules. 2022; 27: 3566. [CrossRef] [Google scholar] [PubMed]
  457. Shubayr N. Phytochemicals properties of herbal extracts for ultraviolet protection and skin health: A narrative review. J Radiat Res Appl Sci. 2023; 16: 100729. [CrossRef] [Google scholar]
  458. Hekmatpou D, Mehrabi F, Rahzani K, Aminiyan A. The effect of aloe vera clinical trials on prevention and healing of skin wound: A systematic review. Iran J Med Sci. 2019; 44: 1-9. [Google scholar]
  459. Matei CE, Visan AI, Cristescu R. Aloe vera polysaccharides as therapeutic agents: Benefits versus side effects in biomedical applications. Polysaccharides. 2025; 6: 36. [CrossRef] [Google scholar]
  460. Mota MD, Costa RY, e Silva LC, Chinalia FA. Guava-fruit extract can improve the UV-protection efficiency of synthetic filters in sun cream formulations. J Photochem Photobiol B Biol. 2019; 201: 111639. [CrossRef] [Google scholar] [PubMed]
  461. Cheshfar F, Bani S, Mirghafourvand M, Hasanpour S, Javadzadeh Y. The effects of ginger (Zingiber officinale) extract ointment on pain and episiotomy wound healing in nulliparous women: A randomized clinical trial. J Caring Sci. 2023; 12: 181. [CrossRef] [Google scholar] [PubMed]
  462. Hassan SW, Mshelia PY, Abubakar MG, Adamu YA, Yakubu AS. Wound healing, antioxidants and toxicological properties of root extracts of Kigelia africana (Lam.) Benth. Int J Sci Basic Appl Res. 2015; 19: 251-268. [Google scholar] [PubMed]
  463. Karatay KB, Muftuler FZ, Law B, Aras O. Methanolic extract of Kigelia africana and wound healing: An in vitro study. J Wound Care. 2023; 32: 392-398. [CrossRef] [Google scholar] [PubMed]
  464. Venkataravana LN, Uppin J, Ramanjineyulu NC, Krishna PG, Narayana JL. Discovering bioactive phytoconstituents from Citrullus lanatus for antimicrobial and antioxidants therapeutic applications. Eur J Med Chem Rep. 2024; 12: 100229. [CrossRef] [Google scholar]
Newsletter
Download PDF Supplementary File Download Full-Text XML Download Citation
0 0

TOP