Open access peer-reviewed chapter

Traditional Plant-Based Treatments of Fungal Infections in the Republic of Suriname (South America): Phytochemical and Pharmacological Rationales

Written By

Dennis R.A. Mans

Reviewed: 17 January 2022 Published: 23 February 2022

DOI: 10.5772/intechopen.102720

Chapter metrics overview

524 Chapter Downloads

View Full Metrics

Abstract

Fungi are unicellular or multicellular thick-walled eukaryotic organisms that are not capable of photosynthesis and are placed in a biological kingdom of their own. They are ubiquitous in our environment, and include tens of thousands, perhaps even millions of species of yeasts, rusts, smuts, mildews, molds, and mushrooms. Together with bacteria, fungi are the principal decomposers of plant materials such as cellulose and lignin, fulfilling vital ecological functions in all terrestrial habitats. Some species of fungi are also of major importance in households (for instance, as foods such as edible mushrooms), medicine (for instance, as producers of antibiotics such as penicillin), and industry (for instance, for making bread, wine, and cheese). About 300 fungal species cause infections in humans, varying from relatively harmless skin complaints such as pityriasis versicolor to potentially life-threatening systemic syndromes such as candidiasis. Fortunately, a broad armamentarium of efficacious antifungal drugs has been developed, ranging from topical nystatin to parenteral amphotericin B. In addition, most, if not all traditional medical systems throughout the world have identified a large assortment of plant-based remedies for treating these infections. This also holds true for the multi-ethnic and multicultural Republic of Suriname (South America), where plant-based traditional medicines are abundantly used, either alone or in conjunction with allopathic medications. This monograph extensively addresses nine plants that are traditionally used for treating fungal infections in Suriname, and explains the phytochemical and pharmacological rationales for these applications. These sections are preceded by some general observations about the Fungal Kingdom; a few words about the characteristics of fungi, their taxonomy, and their significance to humans; information about fungal infections as well as the available forms of treatment; and some details about Suriname including health aspects, the health care structure, and the main fungal infections in the country. The monograph is concluded with an evaluation of the status of the Surinamese herbal antifungal substances and the previsions of developing them into mainstream antifungal formulations.

Keywords

  • fungal infections
  • Suriname
  • medicinal plants
  • traditional uses
  • phytochemistry
  • pharmacology
  • antifungal activity

1. Introduction

The Fungal Kingdom encompasses a diverse and broad group of organisms that includes microscopic yeasts, as well as rusts, smuts, mildews, molds, and mushrooms. These organisms are neither plants nor animals and have been placed in a group of their own because of the distinct chemical composition of their plasma membrane and cell wall, their existence as unicellular and complex multicellular organisms, their heterotrophic lifestyle involving the intake of nutrients from other organisms for their metabolism, and their ability to reproduce both asexually and sexually [1]. Fungi are eukaryotic organisms believed to have descended from an ancestor they shared with animals that lived roughly a billion years ago [2]. The earliest fungal fossils are from the Ordovician, 460–455 million years ago [3], but some scientists believe that fungi may even be 3.5 billion years old [3]. Notably, the first vascular land plants only appeared approximately 425 million years ago, and fungi may have played an essential role in the colonization of land by these early plants [3].

Since their emergence, fungi have evolved into a major taxonomic kingdom. So far, about 150,000 species of fungal organisms have been described [4]. However, studies using molecular methods have estimated that the number of fungal species on Earth may amount to 3.5–5.1 million [5]. This number is probably comparable to that of all animal species on our planet and substantially more than that of all plant species in our biodiversity [5]. Fungi thrive in almost all habitats and are only surpassed by bacteria in their ability to withstand and adapt to extreme environmental conditions [6]. Tropical regions have the highest diversity of fungi [7], and finds of novel, unusual species in these regions are regularly reported. An example is the discovery of the ascomycete Pseudotulostoma volvata in the Guiana Shield described in 2001 [8]. In addition, exploration of deserts and arctic regions has yielded an unexpectedly large fungal diversity [9, 10].

Most fungi are free-living in nature, where they function—together with bacteria—as decomposers in the energy cycle, breaking down organic matter such as leaf litter, animal excrements, wood, and dead animals, and releasing carbon, oxygen, nitrogen, and phosphorus into the soil and the atmosphere to be used by other organisms [11]. This makes fungi vital for the health of ecosystems and the entire biodiversity. Furthermore, various fungal species were at the basis of commercially successful drugs. The common indoor fungus Penicillium chrysogenum produced penicillin, the first of a series of β-lactam antibiotics that revolutionized the medical world [12]. Tolypocladium inflatum parasitizes on insects and gave cyclosporine that is commonly used as an immunosuppressant after bone marrow and organ transplants [13]. And the specific HMG-CoA reductase-inhibiting statins were first discovered as secondary metabolites in the soil-borne fungus Aspergillus terreus and became important LDL-lowering compounds that reduce the risk of arterial blockage and developing a heart attack, stroke, and diabetes mellitus [14].

Other fungi such as the baker’s or brewer's yeast Saccharomyces cerevisiae, along with various mushrooms, have been found useful as food. Saccharomyces cerevisiae converts carbohydrates into carbon dioxide and alcohols through fermentation. The products of this reaction have been used in baking and the production of alcoholic beverages for thousands of years [15]. Saccharomyces cerevisiae has also served as a model organism for studying human genetics including DNA sequences involved in disease and aging [16]. Notably, this fungal species was the first eukaryotic organism to have its genome completely sequenced [17]. Another landmark model fungal species for scientific research was the pink mold Neurospora crassa. Studies with this organism revealed for the first time that genes control the expression of enzymes and that one gene regulates one enzyme [18].

On the other hand, many fungi—such as molds on food or pathogenic spores—are less desirable. Currently, over 8000 fungal species are known to be detrimental to plants, causing, among others, smut and rust, and routinely destroying crops and plants such as beans, barley, and pine trees [4]. And around 300 fungi cause disease in humans [4]. Some, like yeast species in the genus Malassezia cause relatively harmless superficial infections such as pityriasis versicolor [19]. Others, however—like several yeast species in the genus Candida—are the causative agents of potentially fatal infections in immunocompromised individuals [20]. And certain species in the genus Aspergillus produce aflatoxins on improperly stored staple commodities such as peanuts, maize, rice, and cassava, which have been associated with liver cancer [21].

This monograph first presents some important characteristics and the taxonomy of fungi, clarifies the significance of fungi to humans, then deals with the different types of mycoses and their treatment; subsequently focuses on Suriname and elaborately addresses nine plants used for treating fungal infections in Suriname; and concludes with a few remarks on the importance of these plants for medicine.

Advertisement

2. Characteristics of fungi, their taxonomy, and their significance to humans

As mentioned above, fungi are eukaryotic, non-vascular, non-motile and heterotrophic organisms that include yeasts, rusts, smuts, mildews, molds, and mushrooms [22]. These organisms are classified under the Kingdom Fungi, next to the Kingdoms of bacteria, protists, plants, and animals [22]. Fungi are among the most widely distributed organisms on Earth [22]. Many are free-living in soil or water, performing essential ecological functions, while others form parasitic or symbiotic relationships with plants or animals [22]. A considerable number of species is also of great economic and medical importance [22]. Hereunder, a few notable characteristics of fungi are addressed, as well as some aspects of their taxonomy and their significance to humans.

2.1 Characteristics

Although, the numerous fungal species on Earth have widely diverse habitats and characteristics, they have some key aspects in common which make them sufficiently distinct from other organisms to place them in their own biological kingdom [22]. The main differences with the other kingdoms are differences in cell structure, body organization, mode of acquiring nutrition, and means of reproduction. The most important characteristics of the fungal kingdom have elaborately been reviewed in reference [22].

Firstly, fungi are eukaryotes and have their cells surrounded by an inner plasma membrane and an outer cell wall, similarly to bacterial cells and plant cells [23, 24]. However, in fungal cells, both these cellular constituents have a unique composition and distinct properties when compared to the members of the other biological kingdoms. The fungal cell wall contains adhesins and receptors which mediate the interactions of the organisms with the external environment [24]. Furthermore, it helps the organisms to maintain their structure, to protect the cells from various types of stress (particularly osmotic changes), and to guard the integrity of the cellular content [24]. Importantly, the fungal cell wall is mainly made up of glucans, chitin, and glycoproteins instead of cellulose as is the case with plant cells [24]. These ingredients are not present in human cells, providing the opportunity of selective antifungal therapies [24].

The plasma membrane of fungi contains, among others, transport proteins and proteins involved in signal transduction, cell wall and cytoskeleton synthesis, as well as various glycerophospholipids, sphingolipids, and sterols [23]. The main component of the fungal plasma membrane is the unique sterol ergosterol instead of cholesterol which is the major sterol in animal cells [23]. This distinction has therapeutically been exploited by identifying antifungal drugs that target ergosterol synthesis, perturbing the integrity of the fungal cell membrane [23].

Secondly, fungi can consist of a single cell as in the case of yeasts, or multiple cells as in the case of, for instance, mushrooms [22]. Yeasts such as the baker’s or brewer's yeast Saccharomyces cerevisiae are estimated to constitute 1% of all described fungal species [25]. They have probably evolved from multicellular ancestors [26], and some species can develop multicellular characteristics by forming strings of connected budding cells known as pseudohyphae or false hyphae [27]. Yeast species typically measure 3–4 μm in diameter, but some can reach a size of 40 μm [28].

The majority of fungal species are multicellular and is generally known as molds. A few well-known species are those in the genera Aspergillus (commonly found on compost and other decaying vegetable matter as well as stored grain [29]), Penicillium (of major importance in food spoilage and food and drug production [30]), Rhizopus (found on decaying fruit and vegetables, animal feces, and old bread [31]), and Trichophyton (the causative agents of tineas such as athlete’s foot, ringworm, jock itch, and similar infections of the nail, beard, skin and scalp [32]).

The cells of multicellular fungal species are grouped to form tubular, elongated, and filamentous structures called hyphae of 2–10 μm in diameter and up to several centimeters in length, and may contain multiple nuclei [22]. They extend by branching at their tips, and several hyphae mesh together to form the fungal body or mycelium [22]. There are in general specialized hyphal structures for nutrient uptake from living hosts such as the haustoria in most fungal phyla and the arbuscules of several mycorrhizal fungi [33]. Of note, some fungal species can exist as unicellular and multicellular forms, depending on the temperature [34]. An example of such a dimorphic fungus is Talaromyces marneffei, an important cause of opportunistic infections in individuals with HIV/AIDS-related immunodeficiency that grows as a mold at room temperature and as a yeast at human body temperature [35].

Furthermore, fungi lack chlorophyll and cannot conduct photosynthesis, acquiring food by absorbing nutrients from organic substances around them which they break down outside their body by digestive enzymes secreted from the tips of their hyphae [22]. Depending on the species, this occurs through saprophytism, parasitism, or symbiotic relationships. Saprophytic fungi feed on dead organic substances. Examples are species in the genera Aspergillus [29], Penicillium [30], and Rhizopus [31]. Parasitic fungi obtain their nutrition by living on other living organisms (plants or animals) and absorbing nutrients from their host. Examples are Taphrina which causes deformities in flowering plants such as leaf curl disease [36] and witches’ brooms [37], as well as Puccinia that causes leaf rust in, among others, wheat [38]. And symbiotic fungi such as lichens and mycorrhiza have an interdependent relationship with other species in which both are mutually benefited. Lichens are the symbiotic association between algae and fungi [39]. A mycorrhiza is a symbiotic association between a green plant and a fungus. The plant makes organic molecules such as sugars by photosynthesis and supplies them to the fungus, and the fungus provides the plant with water and minerals from the soil such as phosphorus [40].

Lastly, fungi are capable of reproducing by both asexual and sexual means. Relatively simple, single-celled fungi like Saccharomyces cerevisiae reproduce vegetatively by budding [22, 41]. Other forms of vegetative reproduction in fungi are by fission or fragmentation [22, 41]. The majority of multicellular fungal species reproduce asexually through the formation of diploid spores formed by mitosis and called conidia or zoospores or sporangiospores [22, 41]. However, in most mushrooms, puffballs, and toadstools, sexual reproduction takes place by spores (ascospores, basidiospores, and oospores) formed through meiosis [22, 41]. The haploid spores germinate into primary mycelia which form secondary mycelia when they come into contact with each other, after which the nuclei fuse and give rise to cells with the original number of chromosomes [22, 41]. On the other hand, in some fungi, the fusion of two haploid hyphae does not result in the formation of a diploid cell but first in the formation of an intermediate stage called the dikaryophase, which is followed by the formation of diploid cells [22, 41].

Sexual reproduction is an important source of genetic variability, allowing fungi to adapt to new environments. Another strategy of some fungi to generate genetic diversity—often because of stress such as antifungal therapy—is the formation of polyploid/aneuploid nuclei. Dimorphic fungi can switch between a yeast phase and a hyphal phase in response to environmental conditions. An example is the above-mentioned species Talaromyces marneffei [35], probably one of the world’s most feared fungi that can cause potentially lethal talaromycosis [42]. This is a systemic opportunistic infection in immunocompromised individuals that is characterized by fever, skin lesions, anemia, generalized lymphadenopathy, and hepatomegaly [42].

2.2 Taxonomy

The taxonomy of fungi is regularly revised as novel molecular data and DNA comparisons allow for phylogenetic analyses that replace older classifications that mainly use morphological features and reproductive characteristics. Primarily based on the characteristics of sexual reproductive structures, the Fungal Kingdom is currently subdivided into seven phyla: the Microsporidia, the Chytridiomycota, the Blastocladiomycota, the Neocallimastigomycota, the Glomeromycota, the Ascomycota, and the Basidiomycota [43]. The Microsporidia are spore-forming unicellular endobiotic fungi of 1–40 μm that parasitize on animals and humans. Roughly 1500 of the known fungal species belong to this phylum. Several species such as members of the genera Enterocytozoon and Encephalitozoon can cause microsporidiosis in immunocompromised individuals, an opportunistic infection characterized by diarrhea and wasting [44].

The Chytridiomycota, commonly known as chytrids, encompasses approximately 900 identified species that can be encountered in a wide range of aquatic and terrestrial habitats throughout the world. They produce zoospores that are capable of active movement through aqueous phases with a single flagellum. Chytrids are saprobic, feeding on decayed organic matter. Some, like Batrachochytrium dendrobatidis, parasitize on animals. Batrachochytrium dendrobatidis is the causative agent of chytridiomycosis, an infectious disease that is responsible for dramatic declines and even extinctions in amphibian populations in many countries [45].

The Blastocladiomycota are also posteriorly uniflagellated zoosporic fungi that live as saprotrophs primarily in freshwater and soil, and they parasitize on all eukaryotic groups. At this moment, less than 200 species have been described. Whereas their close relatives, the chytrids, mostly exhibit zygotic meiosis (i.e., meiosis of the zygote immediately after karyogamy, resulting in the production of several haploid cells), the blastocladiomycetes undergo sporic meiosis (i.e., meiosis resulting in the production of haploid spores that can develop directly into new, haploid, individuals). A well-known species is Paraphysoderma sedebokerense, a highly destructive pathogen of algae grown in mass cultures for biofuels and pharmaceuticals [46].

The Neocallimastigomycota are anaerobic organisms that live in the digestive system of larger herbivorous mammals [47] as well as that of humans [48] where they play an essential role in the digestion of fibre. They are also closely related to chytrids and form zoospores that are posteriorly uniflagellate or polyflagellate. However, neocallimastigomycetes lack mitochondria but contain hydrogenosomes of mitochondrial origin. This phylum consists of about 20 species that are grouped in only one family, the Neocallimastigaceae. The type species is Neocallimastix frontalis that has been studied as a means of large-scale cellulase production [49].

The Glomeromycota include only one class, the Glomeromycetes, and contain approximately 230 described terrestrial species that are widely distributed in soils worldwide. All known Glomeromycota species reproduce asexually. Many form highly branched arbuscular mycorrhizas with the thalli of bryophytes and the roots of vascular land plants [50, 51]. These mutualistic relationships are beneficial for both participants, because the fungus acquires carbohydrates formed by the plant, and the plant acquires nitrogen, phosphorous, and other minerals produced by the fungus [50, 51]. One of the most distinctive features of glomeromycetes is the arbuscules, the points of exchange between fungus and plants [50, 51].

The Ascomycota, commonly known as sac fungi or ascomycetes, is the largest phylum in the fungal kingdom, harboring over 64,000 species. Members of this phylum reproduce sexually or meiotically via the production of non-motile ascospores inside a microscopic sac-like structure called an ascus, the defining feature of this group of fungi. Because the products of meiosis—the ascospores—are retained within the ascus, ascomycetes such as Neurospora crassa have been used—as mentioned before—for elucidating the principles of genetics and heredity [18]. Many species of Ascomycota also (or exclusively) produce spores called conidia through an asexual or mitotic process [52]. The Ascomycota include fungal species that are of particular importance as sources of antibiotics and for a variety of industrial applications, but also because of their pathogenicity towards humans, animals, and/or plants (see, for instance, references [53, 54]). A few examples are fission yeasts in the genus Schizosaccharomyces, the unicellular yeast genera Saccharomyces and Candida, as well as morels, some edible mushrooms, truffles, and various filamentous ascomycetes in the genera Aspergillus, Penicillium, Fusarium, and Claviceps [53, 54].

The Basidiomycota, also known as the club fungi or basidiomycetes, include mushrooms (such as the champignon mushroom Agaricus bisporus; Figure 1), puffballs, stinkhorns, bracket fungi, other polypores, jelly fungi, boletes, chanterelles, earth stars, smuts, bunts, rusts, mirror yeasts, and the human pathogenic yeast genus Cryptococcus. With over 31,500 species described species, this group is the second largest of the Fungal Kingdom and constitutes, together with the Ascomycota, the subkingdom Dikarya (often referred to as the ‘higher fungi’). Well-known basidiomycetes are species in the genus Puccinia that cause rust in many economically important crops [55], Ustilago maydis that causes smut on maize [56], Malassezia yeasts that are mainly associated with certain skin conditions in humans [57], as well as the opportunistic human yeast Cryptococcus neoformans that usually infects the lungs or the central nervous system but sometimes also other parts of the body [58]. All Basidiomycota are filamentous fungi composed of hyphae (except for basidiomycota-yeast), and reproduce sexually via the formation of specialized club-shaped end cells called basidia that produce meiospores called basidiospores [59]. Another distinctive anatomical feature of this fungal phylum is the presence of clamp connections, hook-like structures which ensure that each hyphal cell or segment of hypha receives a set of different nuclei obtained through the mating of hyphae of differing sexual types to create genetic variation [60].

Figure 1.

The champignon mushroom Agaricus bisporus (from: https://images.app.goo.gl/Ahbf55DQRixrBZfw6).

2.3 Significance of fungi to humans

Humans have been aware of the existence of fungi—albeit indirectly in some cases—since they learned to prepare wine from grapes and bake bread. Ancient peoples were also familiar with the devastating effects of fungi on their crops but attributed these ravages to the wrath of the gods. The Romans had even designated a particular deity, Robigus, as the god of blight and wheat rust, and sacrificed a dog during the Robigalia, a festival held in his honor on April 25 to appease him and protect their grain fields from the disease [61]. Later, many devastating plant pathogenic fungal species have been identified. Examples are Glomerella cingulata that causes anthracnose (plant canker) and fruit rotting diseases on, among others, cereals, grasses, legumes, fruits, vegetables, perennial crops, and trees [62] (Figure 2); Botrytis cinerea, a necrotrophic fungus that is responsible for botrytis bunch rot or grey mould on wine grapes [63]; Gibberella pulicaris, a fungal pathogen that infects, among others, potato, strawberry, hop, and alfalfa [64]; and Podosphaera fuliginea, that causes powdery mildew on cucurbits [65].

Figure 2.

Anthracnose on grapes caused by the plant pathogenic fungus Glomerella cingulata (from: https://images.app.goo.gl/rBcDiAFTKCBwLQXH8).

Several fungal species also cause disease in humans. These are dealt with in the next section. On the other hand, various fungi have a long medicinal use and have been at the basis of many important therapeutics. For instance, ‘Ötzi the ice man’, who had lived between 3400 and 3100 BC during the Copper Age and had been found frozen in the Ötztal Alps (Austria) in 1991, carried pieces of fungus among his personal effects which he presumably had used to treat the parasites in his gastrointestinal tract [66]. Later, fungi made breakthrough contributions to medicine. A few notable examples have been given above and include the β-lactam antibiotics from Penicillium chrysogenum [12], the immune-suppressant drug cyclosporine from Tolypocladium inflatum [13], and the blood cholesterol-lowering statins from Aspergillus terreus [14].

Other drugs of fungal origin are the oral antifungal agent griseofulvin from Penicillium griseofulvum that is efficacious against fungal infections of the skin, nails, and scalp [67]; micafungin, a synthetic analogue from a lead compound produced by Coleophoma empetri for treating invasive fungal infections including candidemia, abscesses, and esophageal candidiasis [68]; and sordarin and its analogues isolated from Sordaria araneosa that target protein synthesis and are active against yeasts and yeast-like fungi [69]. Penicillium griseofulvum occurs on cereals, nuts, and stored fruits [70], Coleophoma empetri causes cranberry fruit rot disease [71], and Sordaria species are commonly found in the feces of herbivores [72]. Furthermore, the polyketide zaragozic acid also known as squalestatin produced by common soil fungi in the genus Phoma—that includes species causing heart rot and blight of beets, and dry rot of sweet potato [73]—potently inhibited mammalian squalene synthase, the rate-limiting enzyme in sterol biosynthesis, thereby blocking cholesterol production to a comparable extent as statins [74].

As well, the sympathicomimetic ergot alkaloids produced by the ergot fungus Claviceps purpurea can constrict the blood vessels around the brain and are therefore very efficacious in relieving migraine headaches [75]. However, these compounds were responsible for dreadful epidemics of poisoning in the Middle Ages, when many individuals developed ergotism after the consumption of bread made from rye or wheat contaminated with this fungal species [76]. The syndrome was popularly known as ‘St. Anthony’s fire’ because it caused intensely painful burning sensations in the limbs and extremities, along with painful seizures and spasms, diarrhea, mental effects including mania or psychosis, headaches, nausea and vomiting, and eventually gangrene, neurological diseases, and death [76]. Due to its specific uterotonic activity, Claviceps purpurea was also used to speed up child deliveries but it claimed instead many additional fatalities [76].

Several ‘domesticated’ fungi have become essential in modern households and industrial processes. As mentioned above, Saccharomyces cerevisiae is instrumental in winemaking, baking, and brewing [15]. The trichocomaceous family members Penicillium roqueforti and Penicillium camemberti are indispensable for the proper ripening of the blue-veined sheep’s milk cheese Roquefort and the cow’s milk cheese Camembert, respectively [77]. The champignon mushroom Agaricus bisporus is one of the most widely consumed mushrooms in the world [78]. The morel mushroom Morchella esculenta and several species of truffles in the genus Tuber are highly appreciated items in haute cuisine [79]. And the mycoproteins derived from the mycelia of the filamentous soil fungus Fusarium venenatum serve as the starting materials for high-protein and high-fiber foods as alternatives to meat and substitutes for fat in dairy products and cereal in breakfast cereals and snacks [80].

Furthermore, to meet the increasing worldwide demands of citric acid, malic acid, and lactic acid, these compounds are currently produced by fermentation with Aspergillus niger and several species in the genera Candida [81], Aspergillus [82], and Rhizopus [83], respectively. Certain fungi are also industrially used to produce enzymes on a large scale. A few examples are lipases for the food, detergent, and pharmaceutical sectors [84]; cellulase for the production of cellulose as veterinary foods, wood and paper, fibers and clothes, cosmetics, and pharmaceutical excipients [85]; and amylases for the food, textile, paper, and detergent industries [86].

Advertisement

3. Fungal infections, epidemiology, and treatment

As mentioned before, there are tens of thousands if not millions of different species of fungus [1, 5]. Many live in air, soil, water, and plants, but some live naturally in the human body [1, 5]. And indeed, only a relative minority—about 300—is pathogenic to humans, and this usually occurs when the invading fungus takes over an area of the body and cannot be effectively dealt with by the immune system [20, 21]. Examples of common types of fungal infections in humans are athlete’s foot, vaginal yeast infections, Candida toenail infections and diaper rash, jock itch, and ringworm [20, 21]. Familiar symptoms of many fungal infections include skin changes, redness, itching, and cracking or peeling skin [20, 21]. Fortunately, these infections are usually not very serious if treated promptly and properly [22, 23], often with over-the-counter or prescription creams [22, 23]. On the other hand, fungal infections are more severe in individuals with a weakened immune system [22, 23]. The same holds for fungal infections in people taking antibiotics [22, 23], undergoing treatment for cancer [22, 23], or suffering from diabetes mellitus [22, 23]. These infections may require additional methods of treatment [5, 6].

3.1 Types of fungal infections

Fungal infections or mycoses can be classified on the basis of the route of acquisition of the pathogen, the degree of virulence exhibited by the fungus, and the site of the infection [20, 21]. The route of acquisition of a fungal infection may either be exogenous or endogenous, i.e., resulting from airborne, cutaneous, or percutaneous fungi, or colonization by a member of the normal flora or reactivation of a previous infection [20, 21]. With regards to the classification based on virulence, primary pathogens can establish infections in normal hosts while opportunistic pathogens cause disease in individuals with compromised host defense mechanisms [20, 21]. More often, fungal infections are classified by the site of the infection, i.e., as superficial, cutaneous, subcutaneous, or systemic (deep) infections depending on the type and degree of tissue involvement and the host response to the pathogen [20, 21].

Superficial mycoses are fungal infections of the skin, hair, and nails that invade only the stratum corneum and the superficial layers of the skin and neither invade living tissue nor provoke an immune response by the host. A well-known superficial fungal infection is pityriasis versicolor, a condition characterized by light spots on the chest, back, upper arms, and legs that are usually caused by Malassezia globosa, a yeast species that normally lives in the pores of the skin [19].

Cutaneous mycoses (also referred to as dermatophytoses or dermatomycoses) are fungal infections of skin, hair, and nails that extend deeper into the epidermis and may evoke host immune responses that result in pathologic changes in the deeper layers of the skin. Common cutaneous mycoses are tinea corporis (ringworm of the body), tinea unguium or onychomycosis (fungal infection of the fingernails, toenails, and the nail bed; Figure 3), tinea pedis (athlete’s foot), tinea cruris (jock itch), and tinea capitis (fungal infection of the scalp and hair) [87]. These conditions are rather common and are mostly caused by parasitic dermatophytes in the genus Trichophyton, in addition to the genera Epidermophyton and Microsporum [87]. Other widespread cutaneous fungal infections are oropharyngeal candidiasis (oral thrush) and vulvovaginal candidiasis, common fungal infection of the mouth and vagina caused by the opportunistic pathogenic yeast Candida albicans [88, 89]. A less frequent but distinctive fungal skin infection that is prevalent in tropical regions is caused by the yeast-like fungus Lacazia loboi, which is transmitted by direct contact with contaminated water, soil, vegetation, or an infected dolphin, and usually presents with bumps in the skin, firm swellings, deep skin lesions, or even malignant tumors [90].

Figure 3.

Tinea unguium or onychomycosis mostly caused by dermatophytes in the genus Trichophyton (from: https://upload.wikimedia.org/wikipedia/commons/5/57/Onychomycosis.JPG).

Subcutaneous mycoses are chronic, localized fungal infections of the dermis, subcutaneous tissues, muscle, and fascia. These lesions are caused by a variety of soil saprophytes following trauma to the skin which allows the fungi to enter the body. Common subcutaneous mycoses are sporotrichosis (rose gardener’s disease) caused by Sporothrix schenkii in soil and decomposing plant material [91], and intertriginous candidiasis, an opportunistic infection of the skin characterized by dermatitis, localized rashes, and/or yellowing of the nails caused by yeasts in the genus Candida [92]. Subcutaneous fungal infections are in general difficult to treat and may require surgical interventions such as debridement [20].

Systemic infections can be caused by primary pathogens or opportunistic pathogens. As mentioned above, systemic mycoses due to primary pathogens can occur in people with a normal immune system [20, 21]. They generally originate in the lungs, tend to develop at a slow rate, and may spread to the brain, heart, lungs, liver, spleen, and/or kidneys [20, 21]. Fungi that cause systemic mycoses are often dimorphic, inherently virulent, and can cause serious health problems. Examples of primary fungal infections are paracoccidioidomycosis (characterized by asymptomatic lung infection preceding in 5% of cases acute or subacute forms in children and young adults, and the chronic form in adults), coccidioidomycosis, and histoplasmosis, caused by soil fungi in the genera Paracoccidioides [93], Coccidioides [94], and Histoplasma capsulatum [95], respectively. These conditions primarily affect the respiratory organs, may remain asymptomatic, but may cause acute, subacute, or chronic infections [20, 21].

Systemic mycoses due to opportunistic pathogens are infections of patients with immunocompromising conditions who would otherwise not be infected [20, 21, 96]. Patients with a high risk of developing such invasive fungal infections are those suffering from HIV/AIDS, those with an altered flora caused by antibiotics, those suffering from diabetes mellitus, those undergoing chemotherapy for metastatic cancer or organ transplantation, and those undergoing major surgical procedures [20, 21, 96]. Common opportunistic mycoses are candidiasis, cryptococcosis, and aspergillosis [20, 21, 96]. Candidiasis is caused by species in the genus Candida (particularly Candida albicans) which normally live on the skin as well as in the mouth, throat, gut, and vagina, without causing any problems in healthy individuals [88, 89]. Cryptococcosis is a pulmonary or disseminated infection acquired by inhalation of soil or dried bird’s feaces (particularly pigeon’s) contaminated with the encapsulated yeasts Cryptococcus neoformans or Cryptococcus gattii [97]. And aspergillosis is caused by species in the ubiquitous genus of Aspergillus and also affects the respiratory system [98]. These fungal infections are in general rather aggressive, often rapidly spread to other organs, and can be life-threatening, while early diagnosis and efficacious treatment are in general difficult [20, 21, 96].

3.2 Epidemiology

Despite the astonishing number of fungal species, only some 300 are known to cause illness in humans. Nevertheless, the number of individuals on Earth having a fungal infection is astonishing [99]. Fortunately, many cases are aymptomatic to mild mucocutaneous infections rather than potentially life-threatening systemic infections [99]. Even so, nearly a billion people on our planet have skin, nail and hair fungal infections; tens of millions suffer from mucosal candidiasis; and more than 150 million fall victim to debilitating and potentially serious fungal diseases [99, 100, 101, 102].

Among the latter conditions are roughly 3,000,000 cases of chronic pulmonary aspergillosis, about 223,100 cases of cryptococcal meningitis complicating HIV/AIDs, about 700,000 cases of invasive candidiasis, about 500,000 cases of pneumocystis pneumonia, about 250,000 cases of invasive aspergillosis, about 100,000 cases of disseminated histoplasmosis, over 10,000,000 cases of fungal asthma, and about 1,000,000 cases of fungal keratitis per year [99, 100, 101, 102]. Notably, over 1.6 million people die each year from their disease, which is comparable to the mortality due to tuberculosis and more than three-fold that caused by malaria [99]. These disturbingly high numbers are important causes of the emergence of the HIV/AIDS pandemic as well as the increasing incidence of chronic obstructive pulmonary disease and cancers, conditions that promote the virulence of opportunistic and potentially pathogenic fungi [103, 104, 105].

3.3 Treatment

The five main classes of drugs for treating fungal infections are polyenes, azoles, allylamines, echinocandins, and the antimetabolite flucytosine [22, 23]. Polyene antimycotics are macrolides derived from certain species of Streptomyces bacteria. These compounds bind to ergosterol in the fungal plasma membrane, perturbing membrane integrity and causing leakage of K+ and Na+ ions [106]. Well-known examples are nystatin from Streptomyces noursei, amphotericin B from Streptomyces nodosus, and natamycin from Streptomyces natalensis [106]. Nystatin is mainly used for the topical, oral, or intravaginal treatment of Candida infections including diaper rash, oral thrush, esophageal candidiasis, and vaginal yeast infections [106]. Amphotericin B is parenterally given against a broad range of invasive fungal infections such as aspergillosis, candidiasis, coccidioidomycosis, and cryptococcosis [106]. Natamycin is mainly used topically as cream and in eye drops for treating fungal infections of the eyelids, conjunctiva, and cornea [106].

Antifungal azoles are synthetic five-membered heterocyclic compounds that can be distinguished into imidazoles and triazoles. Imidazoles have two atoms of nitrogen in the azole ring, triazoles have three. The primary mechanism of action of these compounds is inhibition of lanosterol 14-α-demethylase, an enzyme required for the biosynthesis of ergosterol [107]. Imidazoles such as miconazole, ketoconazole, and clotrimazole are topically used for treating fungal infections of the skin and mucous membranes such as ringworm, pityriasis versicolor, vaginal yeast infections, oral thrush, dandruff, and diaper rash [107]. ‘First-generation’ triazoles such as fluconazole and itraconazole can be given both orally and intravenously against a range of superficial infections, but also against invasive fungal infections such as candidiasis, coccidiodomycosis, cryptococcosis, histoplasmosis, dermatophytosis, aspergillosus, and/or pityriasis versicolor [107]. ‘Second-generation’ triazoles such as voriconazole, posaconazole and ravuconazole have broad-spectrum activity against yeasts and molds including Aspergillus species, and are more potent and more active against resistant fungi when compared to their first-generation counterparts [107].

Allylamine antifungal compounds inhibit squalene epoxidase (one of the rate-limiting enzymes in sterol biosynthesis), resulting in the intracellular accumulation of toxic concentrations of squalene, depletion of ergosterol, and fungal cell death [108]. The two most important representatives of this class of antifungal compounds are naftifine and terbinafine. Naftifine is mainly used for the topical treatment of athlete’s foot, jock itch, and ringworm [108]. Terbinafine, either taken by mouth or applied to the skin as a cream or ointment, is used to treat pityriasis versicolor, ringworm including jock itch and athlete's foot, as well as fungal nail infections [108]. Of note, terbinafine is the treatment of choice for fungal infection of the nails but is only efficacious in this condition when taken as oral tablets, not when used as a topical cream or ointment [108].

A more recent class of antifungal drugs is that of the semisynthetic echinocandin derivatives caspofungin, micafungin, and anidulafungin [109]. These compounds act by inhibiting the synthesis of β-glucan in the fungal cell wall, preventing cross-linking of the cell wall components, perturbing the integrity of the cell wall [109]. Because of this mechanism of action, toxicity associated with echinocandins is infrequent (glucan is not found in mammalian cells) [109]. Furthermore, they are not metabolized by cytochrome P450 enzymes, which virtually excludes drug-drug interactions [109]. Echinocandins are administered intravenously, and are effective for treating (drug-resistant) mucosal and systemic Candida and Aspergillus infections [109].

The fluorinated cytosine analog 5-fluorocytosine or flucytosine is an antimetabolite that, following penetration through the fungal cell via cytosine permease, is intracellularly converted into 5-fluorouracil that perturbs DNA and RNA synthesis, resulting in the death of the fungus [110]. It was initially developed as an antineoplastic agent, but is now indicated in combination with amphotericin B for the treatment of septicemia, endocarditis and urinary system infections caused by Candida spp., as well as meningitis and pulmonary infections due to Cryptococcus spp. [110]. It can be used singly or with other antifungals for chromomycosis [109] and can be given orally or intravenously [110]. Chromomycosis (also known as chromoblastomycosis) is a chronic fungal infection that mainly occurs in subtropical and tropical areas and that is caused by traumatic inoculation of pigmented fungi (generally the ascomycetous soil saprotroph Fonsecaea pedrosoi) and is characterized by slow-growing, raised, crusted small nodular to papillary-like lesions of the skin and subcutaneous tissues [111].

Advertisement

4. Suriname: the country profile, health and health care, and fungal infections

Suriname is a presidential republic located on the northeast coast of South America, bordering the Atlantic Ocean to the north, French Guiana to the east, Brazil to the south, and Guyana to the west (Figure 4). Although, situated in South America, Suriname is considered a Caribbean country rather than a Latin American country [112]. The climate is tropical moderated by trade winds, with dry and rainy seasons [113]. The average daily temperature throughout the year varies between 21 and 32°C (70–90°F), the average annual percentage of humidity is 72.0%, and the yearly rain average is 2200 mm [113]. The capital city Paramaribo lies roughly 15 km from the Atlantic Ocean along the Suriname River. Paramaribo’s historical center has been designated a World Heritage status by the United Nations Educational, Scientific, and Cultural Organization (UNESCO) in 2002 [114].

Figure 4.

Map of Suriname (from: https://images.app.goo.gl/nEEqreFZcJy1XHJz8). Insert: location of Suriname in South America (from: https://images.app.goo.gl/75w3iEDBm44i3W259).

The country’s land area of about 165,000 km2 is divided into ten administrative districts [113, 115], and can be distinguished into northern urban-coastal and rural-coastal plains consisting of sandbanks and mudbanks as well as swampland, and a southern rural interior with mostly savanna grassland, rolling hills that rise to over 1000 m [113, 115], and dense, pristine, tropical rain forest with a great diversity of flora and fauna [116]. The relatively narrow urban-coastal area—that includes Paramaribo and its suburbs—harbors approximately 80% of the population of over 600,000 [113, 115]. The hinterland encompasses more than three-quarters of Suriname’s land surface and is home to the remaining 20% of Suriname’s inhabitants [113, 115]. The urban areas are characterized by a “western” lifestyle, modern health-care facilities, and an economy that is mainly based on commerce, services, and industry [113]. The rural societies have a more traditional way of living, lack comprehensive public health services, and have agriculture, forestry, crude oil drilling, bauxite and gold mining, as well as ecotourism as major economic activities [113].

The official language of government, business, media, and education is Dutch, reminiscent of the nearly 300 years Suriname was a colony under Dutch rule [117]. The common language is Surinamese or Sranantongo has evolved from an English- and Portuguese-based lingua franca in the seventeenth century to a language with its own official orthography [117]. Suriname’s economy is dominated by the mining industry, with oil and gold accounting for approximately 85% of exports and 27% of government revenues [118]. Other means of sustenance are agriculture, fisheries, forestry, and ecotourism [118]. These activities have led to a gross domestic product (GDP) in 2019 of USD 4221 billion and an average gross national income per capita in that year of USD 6340 [118, 119]. This positions Suriname on the World Bank’s list of upper-middle-income economies [119].

Suriname’s population is among the most varied in the world, comprising the Indigenous Amerindians, the original inhabitants; descendants from enslaved Africans imported between the seventeenth and the nineteenth century (called Maroons and Creoles); descendants from indentured workers from China, India (called Hindustanis), and the island of Java, Indonesia (called Javanese) who had been attracted between the second half of the nineteenth century and the first half of the twentieth century; descendants from settlers from several European and Middle Eastern countries; and more recently, immigrants from various Latin American and Caribbean countries including Brazil, Guyana, French Guiana, Haiti, and Cuba [115, 120, 121]. According to the 2012-census, the largest groups are represented by the Hindustanis, Maroons, Creoles, and Javanese, making up roughly 27, 22, 17, and 16%, respectively, of the total Surinamese population [115, 122].

4.1 Health and health care in Suriname

Like most other South American and Caribbean countries [123], Suriname can be characterized as a demographically transitioning country with declining mortality and infertility rates as well as a growing and aging population [124, 125]. These changes are for an important part attributable to considerable progress in health care, nutrition, sanitation, and drinking water quality; the eradication of various infectious diseases including malaria; as well as improvements in average living and working conditions, education, and income [124, 125]. The result was a decline of the death rate from 24 per 1000 in 1923 to 8 per 1000 in 2017 and the attainment of an average life expectancy of 72 years in 2019 [126]. The latter estimate is slightly below those for the rest of South America [126].

On the other hand, as also seen in many demographically transitioning countries [123], during the past decades, non-communicable diseases (NCDs) have largely replaced infectious diseases as the most important causes of morbidity and mortality in Suriname [127, 128]. According to estimates of the World Health Organization, cardiovascular ailments, cancer, and diabetes mellitus, along with their complications, were among the leading causes of morbidity and mortality in Suriname in 2014 [127]. Notably, 68% of the total number of deaths in the country in that year were attributable to these NCDs [127], and the probability of dying from these conditions between age 30 and 70 years was estimated at 14% [127]. These NCDs were also among the main causes of mortality in 2012–2013, accounting for almost 1000 cases or about one-third of the total number of 3260 deaths in that period [127, 128]. Nevertheless, Suriname is not entirely free from infectious diseases, important ones being bacterial and protozoal diarrhea, hepatitis A, and typhoid fever; vector-borne diseases such as dengue fever and chikungunya; and more recently, HIV/AIDS and covid-19 [129, 130]. The fungal infections in Suriname are addressed further in this section.

The responsibility of all aspects of health care in Suriname is in the hands of the Minister of Health and the Director of Health (the Chief Medical Officer) [131]. About 6% of the country’s GDP is available for this purpose [119]. This sum covers, among others, the health costs for the economically weakest individuals, insurance for government employees and employees of government-related companies, as well as import and distribution of essential pharmaceuticals [131]. Specialized institutions under the responsibility of the Ministry of Health are the Regional Health Services and the Medical Mission [131]. The Regional Health Services run about forty community health centers in Suriname’s coastal area and are, together with approximately 250 general practitioners, responsible for primary care in that part of Suriname [131]. The Medical Mission is a non-governmental organization that provides health services to people in the country’s hinterland through a network of almost fifty clinics spread over the interior [131].

These institutions are supported by the Dermatology Services, the Office of Public Health, and Stichting Lobi Health Center [19]. The Dermatology Services is a governmental institution that specifically deals with dermatological diseases, sexually transmitted diseases, and HIV/AIDS [131]. The Office of Public Health is also government-owned and is principally involved in programs to prevent and fight parasitic and microbial diseases [131]. Stichting Lobi Health Center is a non-governmental organization for Sexual and Reproductive Health Services [131]. Secondary care is provided by two private and two government-supported hospitals in Paramaribo and one public hospital in the western district of Nickerie [131]. All hospitals have modern clinical laboratory facilities at their disposal including a microbial culture laboratory at the Academic Hospital Paramaribo [131]. There are, in addition, four private clinical laboratories and three private radiology clinics in Suriname [131]. The Academic Hospital Paramaribo also functions as a training facility for both general practitioners and medical specialists, and provides tertiary care at, among others, a Thorax Center, a Neurology High-Care Unit, and a Neonatal Care Unit [131].

4.2 Fungal infections in Suriname

Until now, there is no registry of fungal and other infections in Suriname, and only a relative handful of scientific papers have addressed these topics. As a result, there are no accurate data available about either the prevalence and incidence of, and mortality due to mycoses in the country. Still, according to several general practitioners, dermatologists, gynecologists, infectologists, and epidemiologists, rather prevalent mycoses in Suriname are infections of the skin and mucous membranes caused by Malassezia spp., those due to Candida spp., as well as tineas caused by dermatophytic fungi in the genera Trichophyton, Epidermophyton, and Microsporum. These infections mainly include pityriasis versicolor (called ‘lota’ in Suriname); infections of the mouth, throat, gut, and vagina; ringworm; infections of fingernails, toenails, and nail bed; athlete’s foot; jock itch; as well as infections of scalp and hair [132, 133], and are treated according to the standard medical protocols addressed in the previous section of this monograph [134, 135].

In addition, a relatively recent overview of infectious diseases of Suriname [136] has mentioned the occurrence in the country of chromomycosis, cryptococcosis, dermatophytosis, histoplasmosis, lobomycosis, as well as invasive fungal infections. As mentioned before, chromomycosis or chromoblastomycosis is a chronic fungal infection of the skin and subcutaneous tissues that is caused by traumatic inoculation of pigmented fungi (often Fonsecaea pedrosoi) [111]; cryptococcosis is a pulmonary or disseminated infection acquired by inhalation of soil or dried pigeon feces contaminated with the encapsulated yeasts Cryptococcus neoformans or Cryptococcus gattii [97]; dermatophytoses are tineas that affect hair, skin, or nails and are most commonly caused by the dermatophytic mold Trichophyton genus [87]; histoplasmosis is caused by inhaling spores of the dimorphic fungus Histoplasma capsulatum that is often found in bird and bat droppings [95]; and lobomycosis is a skin infection that is caused by the yeast-like fungus Lacazia loboi and is transmitted by direct contact with contaminated water, soil, vegetation, or an infected dolphin [90].

Invasive fungal infections reported in Suriname are those involving major syndromes such as candidiasis, blastomycosis, and histoplasmosis [136]. These conditions are often life-threatening and are in general seen in critically ill, aged patients as well as immunosuppressed individuals such as those suffering from AIDS and recipients of transplanted abdominal organs [137]. Invasive candidiasis occurs when endogenous Candida organisms that are normally found in the digestive tract, enter the bloodstream, spread through the body, and damage the heart, brain, eyes, bones, and other parts of the body [138]. Blastomycosis is mainly caused by the thermally dimorphic fungi Blastomyces dermatitidis and Blastomyces gilchristii which cause pneumonia and disseminated disease following inhalation of mycelial fragments and spores [139]. Disseminated histoplasmosis occurs—as indicated in the preceding alinea—after inhaling the spores of Histoplasma capsulatum from droppings of birds and bats, and develops progressively, spreading from the lungs to other organs through the bloodstream, and affecting multiple organs [140]. This syndrome is not uncommon in Suriname, where it has particularly been associated with immunocompromised patients suffering from HIV/AIDS [141]. These infections are also treated as mentioned before in this monograph. Incidentally, an assessment of six families and fifteen genera by the Zoological Collection of Suriname suggested that the amphibian skin disease chytridiomycosis, caused by the pathogenetic fungus Batrachochytrium dendrobatidis, is not present in the country [142].

Advertisement

5. Traditional herbal treatments of fungal infections in Suriname

Despite the availability of affordable and accessible health care [131], traditional medicines are abundantly used in Suriname [143]. The various ethnic groups of Suriname have largely preserved their beliefs, values, behaviors, language, religion, ancestry, and other characteristics, including much of their specific ethnopharmacological traditions [120, 131]. This probably was a means of strengthening the ethnic identity during the secluded lifestyle the former colonial authorities had forced them into [120, 131]. As a result, the use of various forms of traditional medicine is deeply rooted in the entire Surinamese population [143]. Consequently, many diseases are often treated with traditional plant-based medicines instead of, or together with prescription drugs [143]. This also holds for several fungal infections, particularly the relatively mild superficial and cutaneous mycoses.

All ethnic groups in Suriname have made important contributions to the traditional treatment of mycoses. The Indigenous Trio Amerindians recognize various skin infections including those from the fungal origin, and have identified medicinal plants for treating these conditions [144]. Maroon and Creole females regularly use genital steam baths for their personal hygiene [145], but presumably also to prevent and treat vaginal candidiasis [145]. Chinese traditional healers have prescribed curcumin, the yellow-colored secondary metabolite in the turmeric Curcuma longa L. (Zingiberaceae), for centuries against fungal infections [146]. Practitioners of Indian Ayurveda, Unani, and Siddha are for over two millennia familiar with the antifungal properties of the neem tree Azadirachta indica A. Juss., 1830 (Meliaceae) [147]. And Javanese Jamu has recognized centuries ago the apparent efficacy of the rhizomes from the blue ginger Alpinia galanga (L.) Willd. (Zingiberaceae) against pityriasis versicolor [148].

The next sections address nine plants that have less thoroughly been evaluated in the scientific literature but are commonly used in Suriname for treating fungal infections. The plants have been selected from several comprehensive publications describing various aspects of medicinal plants in the country [148, 149, 150, 151, 152, 153, 154, 155]. Their relevant aspects have been summarized in Table 1. Hereunder, the plants have in detail been assessed for their phytochemical and pharmacological properties, to support their traditional use against fungal infections.

Plant familyPlant species (vernacular name in English; Surinamese)Part(s)
Used
Presumed active constituent(s)Antifungal activity [references]
AraceaeColocasia esculenta (L.) Schott. (taro; snesi taya)Leaf
Corm
Flavonoids
Phytosteroids
Aspergillus niger, Candida albicans [156]
Athelia rolfsii [157]
EuphorbiaceaeEuphorbia hirta L. (milkweed; sabana merkiwiwiri)Leaf
Flower
Root
Shoots
Stem, stem latex
Aerial parts
Whole plant
Tannins
Flavonoids
Alkaloids
Glycosides
Proteins
Sterols
Saponins
Aspergillus spp., Fusarium spp., Trichophyton spp. [158, 159, 160, 161, 162, 163]
Saccharomyces cerevisiae [164]
Colletotrichum capsici, Lasiodiplodia theobromae, Phomopsis caricae-papayae [160]
Rhizopus oryzae [161, 163]
Candida albicans [165, 166]
LamiaceaeOcimum tenuiflorum L. (holy basil; tulsi)Leaf, leaf essential oil
Root
Eugenol; linaloolAlternaria spp. Aspergillus spp., Penicillium chrysogenum [167, 168, 169].
Candida spp. [169, 170]
Trichophyton spp., Microsporum spp., Epidermophyton floccosum [171]
Fusarium oxysorum, Cochliobolus lunatus, Rhizoctonia solani [169]
LauraceaePersea americana Mill.
(avocado; afkati)
Seed, epicarp
Flesh of unripe fruit, peel
Leaf
Persin
1-acetoxy-2,4-dihydroxy-n-heptadeca-16-ene and 1-acetoxy-2-hydroxy-4-oxo-heneicosa-12,15-diene
Candida spp. [172, 173, 174]
Cryptococcus neoformans [175, 176]
Malassezia pachydermatis in dogs [177]
Zygosaccharomyces bailii [178]
Rhizopus stolonifera, Botryodiploidia theobromae, Fusarium oxysporum, Geothricum candidum [179]
Aspergillus spp. [179, 180]
Colletotrichum gloeosporioides [181, 182, 183]
LythraceaePunica granatum L. (pome granate; granaatappel)Fruit, fruit peel, fruit juice
Seed
Phenolic compoundsAspergillus fumigatus [184]
Candida spp. [184, 185] including Candida associated with denture stomatitis [186]
Mucor indicus, Penicillium citrinum, Rhizopus oryzae, Trichoderma reesei [187]
Various dermatophytic fungi [188]
Botrytis cinerea [185, 186, 188, 189, 190, 191, 192]
Pyricularia oryzae, Colletotrichum falcatum, Dreschlera rostrata, Curvularia lunata [188]
Penicillium digitatum [190]
Fusarium sambucinum [193]
MyristicaceaeVirola surinamensis (Rol. ex Rottb.) Warb. (baboonwood; babunudu)Root
Stembark
Leaf
Seed
Flavonoids; (neo)lignans; capric acidCladosporium cladosporioides [194]
Candida spp. [195, 196]
Microsporum gypseum [197]
MyrtaceaePsidium guajava L. (common guava; guyaba)Leaf
Ripe fruit
Bark
Twigs
Phenolic compounds (tannins, coumarins, flavonoids), terpenoidsCandida spp., Saccharomyces spp., Cryptococcus spp., Trichosporon spp., Aspergillus spp., Sporothrix spp., Microsporum spp., Trichosporon spp. [198, 199, 200, 201, 202, 203, 204, 205]
Arthrinium sacchari, Chaetomium funicola [206]
RananculaceaeNigella sativa L. black caraway; zwarte komijnSeed, seed essential oilPeptidergic defensins Ns-D1 and Ns-D2Aspergillus spp., Curvularia spp., Microsporum spp., Penicillium spp., Trichoderma spp., Candida spp., Chaetomium spp., Fusarium spp., Trichophyton spp., Chrysosporium spp. [207, 208, 209, 210, 211, 212, 213, 214, 215, 216]
Various dermatophytic fungi [217, 218]
RutaceaeAegle marmelos (L.) Corrêa (golden apple; belLeaf, leaf essential oil
Fruit
TerpenoidsVarious dermatophytic fungi [219, 220, 221, 222, 223, 224, 225, 226, 227]
Fusarium spp. [227, 228]
Trichophyton spp., Microsporum spp., Epidermophyton spp. [181, 229, 230, 231, 232, 233].
Pythium debaryanum [183]
Aspergillus spp., Epidermophyton floccosum, Candida albicans [229, 230, 231, 232, 233]

Table 1.

Plants with antifungal activity addressed in this section, parts preferentially used, presumed antifungal constituents, and proven antifungal activity.

References are placed between [ ].

5.1 Araceae—Colocasia esculenta (L.) Schott

The taro Colocasia esculenta (L.) Schott. is believed to originate from south-eastern Asia (hence its Surinamese vernacular name ‘snesi taya’ meaning ‘Chinese taro’), but is now grown in many tropical regions including Suriname for its edible, starchy corm. The plant bears characteristic large green leaves which resemble elephant ears (Figure 5) and have long, erect petioles of 40–200 cm tall growing in clusters from a large, tuberous rootstock. C. esculenta is presumably one of the earliest plants human beings have started to cultivate and has become one the most extensively grown members of the plant family Araceae. Similarly to yam, C. esculenta corm is an important food staple in many African, Oceanic, and South Asian cultures. The corm can be roasted, baked, or boiled, and is also used for baby food. Furthermore, the nutritious young leaves and stems are eaten after the pungent flavor and the stinging and burning calcium oxalate- or calcium carbonate-containing raphides [234] have been removed by boiling twice or by steeping overnight in cold water.

Figure 5.

The taro Colocasia esculenta (L.) Schott. (Araceae) (from: https://images.app.goo.gl/HH8Zt3mnRgyfVqhSA).

C. esculenta corm, leaf, leaf sap, and stem are traditionally used for treating a variety of conditions such as parasitic infections; asthma; diabetes mellitus; hypertension; neurological disorders; arthritis and other inflammatory disorders; body ache; amenorrhea; stomach problems and liver ailments; internal haemorrhages; cysts, boils, and wounds; baldness; as well as bacterial infections including conjunctivitis [235, 236]. In Suriname, a raw peeled corm is eaten with salt against jaundice [153]; the grated whole plant is made into an ointment for treating skin conditions including acute dermatitis that may be caused by Malassezia yeasts [237]; the stem is rubbed between the toes to treat fungal infections such as tinea pedis after the raphides have been broken down by heating [148]; and the corm is used as an ingredient of genital steam baths to recuperate the uterus post-partum and treat vulvovaginal candidiasis [155].

The results from several pharmacological studies provided support for some of these traditional uses. Aqueous and ethanolic leaf extracts decreased blood glucose levels in alloxan-induced diabetic rats to a comparable degree as the oral hypoglycemic medication metformin [238]. Cooked corm decreased tissue cholesterol and triglycerides in cholesterol-fed rats [239]. Ethanolic leaf extracts exerted anti-inflammatory activity, inhibiting carrageenan-induced hind paw edema, carrageenan-induced pleurisy, and cotton pellet-induced granuloma in Wistar rats [240]. Ethanolic corm extracts elicited hepatoprotective effects in paracetamol- and carbon tetrachloride-treated laboratory rats [241] while ethanolic leaf extracts had similar effects in precision-cut liver slices from rats [242]. And aqueous and ethanolic leaf extracts showed anthelmintic activity against earthworms [243].

Several studies also reported antimicrobial activity of C. esculenta including antifungal activity. For instance, an aqueous extract of the leaves displayed remarkable in vitro activity against various bacterial strains as well as the common food contaminant Aspergillus niger and the opportunistic pathogenic gastrointestinal yeast Candida albicans when compared to the reference compounds chloramphenicol and rifampicine [156]. Incidentally, a chloroform extract of C. esculenta leaf also showed antibacterial activity in an evaluation of fifteen coastal medicinal plants for antibacterial activity against bacterial fish pathogens [244]. Furthermore, comparing 130 ethanol extracts of lyophilized vegetables for inhibitory activity on recombinant human lanosterol synthase activity in a cell-free assay, a C. esculenta corm preparation showed the highest activity [245]. This is of relevance when considering the key role of lanosterol synthase in the viability of fungi [246].

A wide range of chemical compounds including flavonoids, β-sitosterol, and steroids have been isolated from C. esculenta [235, 236]. Still, there are no clear indications about the biochemical identity of the antifungal compound in this plant. However, in a study with the phytopathogenic crust fungus Athelia rolfsii—the causative agent of southern blight in many crops including tomato, onion, snapbean, and pea—the antifungal activity could be related to a phytocystatin in the corm that potently inhibited cysteine protease [157]. The phytocystatin inhibited mycelium growth and lyzed sclerotia by acting against the endogenous cysteine proteinase in the mycelia [157]. Notably, the inhibition of exogenous proteinases by phytocystatins have been suggested to represent a defense mechanism of plants against, among others, pathogenic fungi [247].

5.2 Euphorbiaceae—Euphorbia hirta L

The milkweed Euphorbia hirta L. (Figure 6) is a pantropical weed that is presumably native to India but is now encountered in many tropical parts of the world in gardens, abandoned estates, open grasslands, and along roadsides. It can grow up to 60 cm tall and has a solid, hairy stem. The vernacular name ‘milkweed’ stems from the white latex the stem produces when damaged, a characteristic shared by most plants in the genus Euphorbia [248]. This distinctive feature is also reflected by the Surinamese vernacular name ‘sabana merkiwiwiri’ that means ‘the herb from the savanna that emits a milky substance’. The latex is believed to represent a deterrent to herbivores, is rather toxic, and is used as an ingredient of arrow poisons and antimicrobial substances [249].

Figure 6.

The milkweed Euphorbia hirta L. (Euphorbiaceae) (from: https://images.app.goo.gl/1HjVzdfUghX9AyiH7).

E. hirta has also been named ‘asthma-plant’ because of the traditional use of preparations from the leaf against asthma, bronchitis, and hay fever [250]. Preparations from various parts of the plant are, furthermore, traditionally used for treating, among others, gastrointestinal disorders; worm infestations; hypertension; genito-urinary conditions including kidney stones; diabetes mellitus; lactation insufficiency; conjunctivitis; several skin diseases as well as swellings and boils; and as a gargle for oral thrush [155, 251, 252]. In Suriname, E. hirta is used, in addition, as an essential ingredient of vaginal steam baths, in herbal baths to make child delivery proceed smoothly, and as an ingredient of cleansing baths to protect against evil forces [155].

Pharmacological studies with various laboratory models supported the traditional claims of E. hirta preparations against asthma [251], diarrhea [253], bowel cramps [254], hypertension [255], and diabetes mellitus [256], and as a diuretic [257]. There was also preclinical evidence for sedative and anxiolytic activity [258]; analgesic, antipyretic, and anti-inflammatory effects [259, 260]; as well as antiamoebic [254] and antimalarial actions [261, 262]. Several of these effects have been associated with the presence in the plant of polyphenolic compounds such as quercitin [253, 254, 263, 264]. In addition, preparations from several of its parts exhibited meaningful and broad antibacterial activity in laboratory assays (see, for instance, references [158, 164, 165] including some activity against Helicobacter pylori [265].

There is also ample evidence for antifungal activity of E. hirta. For instance, aqueous extracts from the leaf inhibited aflatoxin contamination of rice, wheat, maize, and mustard crops by the cosmopolitan saprotrophic and pathogenic fungus Aspergillus flavus [159] and the growth of Saccharomyces cerevisiae in culture [164]. Ethanolic leaf extracts displayed meaningful activity against the plant pathogenic molds Colletotrichum capsici, Fusarium pallidoroseum, Lasiodiplodia theobromae, and Phomopsis caricae-papayae [160] as well as Aspergillus fumigatus, Aspergillus niger, Aspergillus flavus, and Rhizopus oryzae [161]. Importantly, some combinations of a methanol extract of the leaf and nystatin exhibited synergistic activity against clinical isolates of Candida albicans [266].

A preparation from the whole plant also inhibited aflatoxin production by Aspergillus flavus [162], while methanolic and acetonic extracts from the whole plant and the latex from the stem showed meaningful activity against Aspergillus flavus; Fusarium incarnatum that affects crops such as sorghum, rice, and maize; as well as Trichophyton spp., the causative agents of athlete's foot, ringworm, jock itch, and similar infections of nail, beard, skin and scalp [158]. Furthermore, ethanolic extracts of the aerial parts of the plant were active against Aspergillus niger, Aspergillus fumigatus, Aspergillus flavus, and Rhizopus oryzae [163]; methanolic extracts of leaf, flower, stem, and root were active against Candida albicans [166]; and water extracts of the root and shoots were active against a clinical isolate of Candida albicans, although not against Penicillium spp., Aspergillus spp., and Microsporum spp. [165].

So far, only a few studies have assessed the ingredient(s) and/or the mechanism(s) involved in the antifungal activity of E. hirta preparations. For instance, the antimicrobial activity of the crude plant extract has been attributed to tannins, flavonoids, alkaloids, glycosides, proteins, sterols, and/or saponins [163]. Nevertheless, these observations support the use of E. hirta in traditional medicine against certain fungal infections, providing a possible explanation for the use of a decoction of fresh leaves as a treatment for oral thrush [251] and in genital steam baths for cleansing the vagina from, among others, fungal infections [155].

5.3 Lamiaceae—Ocimum tenuiflorum L

Ocimum tenuiflorum L. (Figure 7), commonly known as holy basil or tulsi in Surinamese-Hindi or Sarnami, is a perennial plant growing between 30 and 100 cm tall that is particularly valued for its strong-scented and sharp-tasting but edible leaves. It is native to India but has spread throughout the south-eastern parts of Asia as a cultivated plant, and is also grown in various other tropical parts of the world with an Indian diaspora such as Suriname. As indicated by the vernacular ‘holy basil’, it is a sacred plant in Hinduism and is widely grown around Hindu houses and temples for its perceived protective influence [267]. Leaf preparations are regarded as ‘elixirs of life’ and believed to promote longevity [267], and the flowers are used as a holy cleanser for food offerings during prayers.

Figure 7.

The holy basil Ocimum tenuiflorum L. (Lamiaceae) (from: https://images.app.goo.gl/F1HdJrRmdGUcjxZM6).

O. tenuiflorum leaf essential oil and seed extracts contain antimicrobial, insecticidal, and antifeedant compounds and have for centuries been added to stored grains to repel insects [268]. The seeds also yield an essential oil containing pleasantly scented monoterpes, sesquiterpenes, and phenols which are used in the cosmetic industry and food technology [269]. In addition, preparations from all parts of the plant have a myriad of traditional medical applications. A few indications are asthma and bronchitis; fevers, colds, influenza, sinusitis, and headaches; inflammatory conditions such as rheumatism and arthritis; pains, cramps, and spasms; diabetes mellitus; hypertension; convulsions; stress and a disturbed homeostasis; parasitic infections such as leishmaniasis; as well as antibacterial and antifungal infections such as ringworm [153, 155, 270]. Some of these activities have been attributed to eugenol, the major compound in the essential oil, as well as other terpenoids such as β-elemene, β-caryophyllene, and germacrene [271, 272].

Several pharmacological investigations supported some of the traditional claims for O. tenuiflorum. For instance, in line with its presumed usefulness against asthma and related conditions, a leaf extract displayed anti-anaphylactic, antihistaminic, and mast cell-stabilizing activities in laboratory rodents [273]. Indications for its anti-inflammatory, analgesic, and antipyretic activities came from the inhibitory effects in laboratory animals of the non-volatile oil from leaf and seed on formaldehyde-, carrageenan-, inflammatory mediators-, or turpentine oil-induced arthritis, paw edema, and joint edema [274, 275, 276]; acetic acid-induced tail flicking, tail clipping, tail immersion, and writhing [277]; and typhoid-paratyphoid A/B vaccine-provoked pyrexia [277].

Furthermore, oral administration of an ethanolic leaf extract led to marked lowering of blood sugar in animal and laboratory models of diabetes mellitus [278, 279], and the O. temuiflorum-containing polyherbal Ayurvedan supplement Diabecon® seemed efficacious and safe in helping control blood sugar levels in diabetics [280]. In addition, ethanol and chloroform leaf and stem extracts protected rats to a comparable extent as phenytoin from tonic convulsions induced by transcorneal electroshock [281]. And ethanolic leaf extracts led to normalization of stress hormone and neurotransmitter levels in animals which had been exposed to noise stress [282, 283].

As well, both the essential oil from O. tenuiflorum leaf and purified eugenol showed potent anthelmentic activity against Caenorhabditis elegans [284]. Aqueous extracts, several organic extracts, and the essential oil of the leaf and the root of the plant also exerted meaningful antibacterial activity in vitro against a broad range of bacterial strains (see, for instance, references [167, 285]. O. tenuiflorum preparations were also active against isolates of various bacterial strains from urine, stool, and sputum from infected individuals [286], and attacked both Gram-positive and Gram-negative pathogenic bacteria [287] as well as methicillin-resistant Staphylococcus aureus [288].

There are also several studies reporting antifungal activity of O. temuiflorum. Aqueous leaf extracts and the leaf essential oil were active against cultures of Candida albicans and Candida tropicalis [170] as well as those of Aspergillus flavus [168]. The fractions obtained after successive extraction of the crushed and dried leaf with hexane, benzene, chloroform, ethyl acetate, methanol, and water were active against five different clinical isolates of dermatophytic fungi, namely Trichophyton mentagrophytes, Trichophyton rubrum, Microsporum canis, Microsporum gypseum, and Epidermophyton floccosum [171]. Hexane, acetone, and ethanol extracts of leaf and root were active against the plant pathogenic fungal strains Alternaria alternata, Fusarium oxysorum, Cochliobolus lunatus, and Rhizoctonia solani, as well as Aspergillus flavus and Candida albicans [169]. And a methanol leaf extract was active against cultures of the fungal species Alternaria porri; Aspergillus flavus, Aspergillus niger, and Aspergillus oryzae; as well as Penicillium chrysogenum [167]. Some investigators attributed the antifungal activity of the essential oil to eugenol [168], but others to linalool [170]. Markedly, both the leaf essential oil and eugenol not only inhibited the growth of Aspergillus flavus but also completely inhibited aflatoxin B1 production by the fungus [168].

5.4 Lauraceae—Persea americana Mill

The avocado Persea americana Mill. is a large, spreading, evergreen tree that can reach a height of approximately 30 m and bears large fruits (Figure 8) that are surrounded by a leathery peel and contain a single large seed. The plant presumably originates from south-central Mexico but is now cultivated in various tropical and subtropical areas for the commercially valuable and very nutritious fruit [289]. The dried or fresh leaves are prepared as a tea but also used as a spice in stews, giving a flavor that resembles that of anise.

Figure 8.

The avocado Persea americana Mill. (Lauraceae) (from: https://images.app.goo.gl/LoRCDzk7Z8bW61A28).

The fresh fruit pulp is massaged into the hair and scalp as a vitamin-rich hair tonic and restorer, and the cosmetic industry uses the fruit oil as an ingredient of soaps and skin moisturizer products [290]. Avocado oil has a favorable lipid composition and is very suitable for cooking due to its high smoke point [290, 291], but is relatively expensive when compared to common salad and cooking oils. Leaf and seed contain cyanide and the fatty acid fungicidal compound persin in amounts that can cause intoxications in small animals but at too low concentrations to be hazardous to humans [292]. However, more recent studies showed that methanolic extracts of fruit and leaf caused chromosomal aberrations in cultured human peripheral lymphocytes [293].

Almost all parts of P. americana are medicinally used in many traditional systems including those in Suriname. Conditions treated with preparations from this plant are, among others, dysentery, gut parasites, gastritis, gastroduodenal ulcers, and diarrhea; liver and bilious disorders; anemia; hypertension and hypercholesterolemia; coughing; menstrual problems; cystitis; exhaustion; nervousness; to evoke abortion; against scabies, purulent wounds, lesions of the scalp, dandruff, and hair loss; as well as various microbial infections [149, 152, 153, 155, 289]. Some of these applications may be related to the presence in the plant of certain aliphatic acetogenins such as persin, avocadenols, 1,2,4-trihydroxyheptadec-16-ene, and 1,2,4-trihydroxynonadecane; terpenoid glycosides such as glycosylated abscisic acid derivatives; furan ring-containing derivatives called ‘avocadofurans’; flavonoids such as quercetin, catechin, and epicatechin; as well as the plant coumarin scopoletin [294].

The results from pharmacological studies support some of the ethnomedical uses of P. americana. Aqueous leaf and seed extracts displayed vasorelaxant and blood pressure-lowering activities in laboratory rats and isolated rat aortic rings precontracted with KCl or norepinephrine [295, 296, 297]. Aqueous and/or methanolic leaf and/or seed extracts reduced plasma glucose levels in alloxan-treated rats [298, 299] and influenced lipid metabolism in normal or hypercholesterolemic rats [297, 298, 300]. The fruit extract exerted hepatoprotective effects in rats which might be attributed to certain constituents in the oil such as the fatty acid acids [301]. A methanol leaf extract also exerted hepatoprotective activity in rats [302], while aqueous leaf extracts showed analgesic and anti-inflammatory [303] as well as anticonvulsant effects in laboratory mice [304].

Furthermore, chloroformic and ethanolic extracts of the seed were active against Giardia lamblia and Entamoeba histolytica, the causative agents of giardiasis and amoebic dysentery, respectively [305]. Seed preparations also showed in vitro activity against the trophozoites of Trichomonas vaginalis (that is responsible for trichomoniasis) [305], the epimastigotes of Trypanosoma cruzi (the etiological agent of Chagas’ disease) [306], and the larvae of Aedes aegypti (that spreads, among others, yellow fever, dengue fever, chikungunya, and Zika fever viruses) [172]. The antitrypanosomal activity was tentatively localized to 1,2,4-trihydroxyheptadecane and 1,2,4-trihydroxynonadecane derivatives [306].

Evidence for antibacterial activity of P. americana came, among others, from the in vitro activity of organic seed extracts against both Gram-positive and Gram-negative bacteria (except Escherichia coli) [307], and drug-sensitive as well as mono-resistant and multidrug resistant strains of Mycobacterium tuberculosis [305]; and that of a petroleum ether root extract against several bacterial strains including methicillin-resistant Staphylococcus aureus [175]. Some of the antibacterial effects might be due to the high phenolic content of the plant parts tested [173] and/or to avocadenols, 1,2,4-trihydroxyheptadec-16-ene, and 1,2,4-trihydroxynonadecane in the (unripe) fruit pulp [308].

There is also abundant evidence supporting that P. americana has antifungal activity. Hexane, chloroform, and methanol seed extracts displayed in vitro activity against yeast species including Cryptococcus neoformans [175, 176], Malassezia pachydermatis in dogs [177], and strains of Candida [172]. Ethanolic extracts from the epicarp and seed of several P. americana cultivars were active against the yeast Zygosaccharomyces bailii that is responsible for the spoilage of many normally shelf-stable acidic and/or high-sugar products such as fruit concentrates, wine, soft drinks, syrups, ketchup, mayonnaise, pickles, and salad dressings [178], but not against Penicillium spp. and Aspergillus flavus [307]. Seed extracts also showed in vitro activity against the black bread mold Rhizopus stolonifer as well as the plant pathogenic fungi Aspergillus flavus, Botryodiploidia theobromae, Fusarium oxysporum, and Geothricum candidum [179]. In addition, a hydroethanol leaf extract and ethyl acetate and butanol fractions derived therefrom, and an acetonic leaf extract showed substantial activity against both drug-sensitive and drug-resistant strains of Candida glabrata [173] and Candida albicans [174] as well as Aspergillus fumigatus, Aspergillus niger, and Aspergillus flavus [180].

The compounds responsible for the antifungal activities have not conclusively been identified. However, persin isolated from P. americana idioblast cells inhibited spore germination of Colletotrichum gloeosporioides that causes anthracnose and fruit rotting diseases in many economically important plants [181]. And 1-acetoxy-2,4-dihydroxy-n-heptadeca-16-ene and 1-acetoxy-2-hydroxy-4-oxo-heneicosa-12,15-diene in peel and flesh of unripe avocado fruits may act synergistically to prevent Colletotrichum gloeosporioides of infecting and causing bitter rot in P. americafruit and various other economically valuable crops [182, 183].

5.5 Lythraceae—Punica granatum L

The pomegranate Punica granatum L. is a slow-growing, deeply rooting, drought-tolerant, fruit-bearing deciduous shrub or small tree that grows to an average height of 5 meters, and is originally from the region extending from modern-day Iran to northern India. It is presumably one of the first fruit trees to be domesticated in the eastern Mediterranean region (around the fifth millennium BC), and is now cultivated in warm temperate to subtropical and tropical zones as an economically highly valued fruit crop (Figure 9). The inner, spongy mesocarp of the fruit houses around six hundred seeds, each surrounded by a juicy, deep red- or purple-colored sour-tasting pulp, the edible part of the fruit. The red color is attributable to anthocyanins such as delphinidin, cyanidin, and pelargonidin glycosides, the sour notes are from the acidic ellagitannins [229].

Figure 9.

The pomegranate Punica granatum L. (Lythraceae) (from: https://images.app.goo.gl/4bBBQ5FahGprBx4FA).

P. granatum has a history of traditional medical use that dates back more than 3000 years, and is still abundantly used today in various traditional medical systems. Indications are, among others, worm infestations, diarrhea and jaundice; throat infections, colds, fevers, cough, and a runny nose; nose bleeds; mouth sores and bleeding gums; tuberculosis and shortness of breath; hypertension; diabetes mellitus; syphilis; menstrual problems and vaginal discharge including leucorrhoea; and various other microbial and viral infections [149, 150, 152, 230, 231].

The results from phytochemical and pharmacological studies supported some of the traditional uses. For instance, leaf, stem bark, and peel extracts exerted both in vitro and in vivo activity against the gastrointestinal parasites Schistosoma mansoni [232], Ascaris suum [233], and Giardia lamblia [309]. These effects might be due to pelletierine alkaloids, unusual alkaloids which, among others, paralyze tapeworms so that they are easily expelled from the body by using a laxative [310]. Other compounds isolated from the fruit such as the phenolics gallagic acid and punicalagins exhibited in vitro activity against the malaria parasite Plasmodium falciparum [184].

As well, orally administered P. granatum juice or seed or flower extracts led to a significant reduction of blood glucose levels as well as those of blood lipids and lipoproteins in streptozotocin- or alloxan-induced diabetic rats [311, 312] and type II diabetic patients with hyperlipidemia [313]. These effects have been attributed to oleanolic, ursolic, and gallic acids [314], and might be associated with inhibition of the reabsorption of glucose in the proximal tubuli of the kidneys [315], an increased utilization of glucose by the peripheral tissues [311], and/or improved sensitivity of the insulin receptor [314]. Furthermore, P. granatum fresh, fermented or concentrated fruit juice as well as cold-pressed seed oil elicited meaningful anti-inflammatory activities in vitro and in experimental mouse and rat models of colitis and osteoarthritis [316, 317].

There are many pharmacological indications for the antimicrobial activity of P. granatum. Extracts from leaves, roots, bark, rhizomes, seeds, fruit peels, fruit juice, whole fruit, as well as flowers prepared with solvents of different polarities, showed broad and strong activity against various bacterial strains (see, for instance, references [184, 187, 189, 318, 319]), including multidrug-resistant pathogenic bacteria [184, 318]. The antibacterial activity was more pronounced against Gram-positive than against Gram-negative bacteria [319], was often highest in the methanol extracts or fractions [246], and has also been attributed to several phenolic compounds [184, 189, 318, 319].

There is ample evidence for antifungal activity of P. granatum. For instance, extracts of various parts of the plant were active against dermatophytic fungi [188]. An extract of the fruit formulated as a gel elicited antifungal activity in individuals with candidosis associated with denture stomatitis [186]. Extracts from the fruit peel, seed, juice, and whole fruit were active against industrially important fungi such as Mucor indicus, Penicillium citrinum, Rhizopus oryzae, and Trichoderma reesei [187]. Furthermore, several fractions of the fruit juice obtained by column chromatography, as well as purified ellagic acid, gallagic acid, punicalins, and punicalagins exerted in vitro activity against the common pathogens Candida albicans and Candida neoformans as well as Aspergillus fumigatus [184]. Punicalagin isolated from the fruit peel also showed strong in vitro activity against Candida albicans and Candida parapsilosis, and acted synergistically with fluconazole [185].

In addition, extracts from various parts of the plant were active against the plant pathogenic fungi Pyricularia oryzae, Colletotrichum falcatum, Dreschlera rostrata, and Curvularia lunata, completely inhibiting spore germination of the latter two species [188]. Particularly the fruit peel seemed to possess potent activity against plant pathogenic fungi. Preparations from this part of the plant were active against the plant pathogenic fungi Penicillium digitatum [190], Botrytis cinerea [185, 186, 188, 189, 190, 191, 192], and Fusarium sambucinum [193], both in vitro and in vivo. These fungal species are responsible for green rot in citrus fruit, bunch rot in grapes, and dry rot on potato tubers, respectively. Again, methanolic extracts seemed to be more effective than water extracts [191], and the main antifungal constituents of P. granatum seemed to be phenolic compounds such as ellagic acid derivatives like punicalagin, castagalagin, and granatin; gallotannins and procyanidins; cyanidin 3-glucoside and pelargonidin 3-glucoside; as well as catechins, kaempferol, and quercetin [187, 190, 192, 318].

5.6 Myristicaceae—Virola surinamensis (Rol. ex Rottb.) Warb

Virola surinamensis (Rol. ex Rottb.) Warb, commonly known as baboonwood or (white) ucuba is probably native to the tropical and subtropical parts of northern Brazil, Venezuela, Guyana, French Guiana, Suriname, Panama, Costa Rica, and the West Indies, where it can be found in moist lowland forests, swamps, and heavily degraded former forests. It is an evergreen tree that grows 25–35 m tall and has a straight, widening cylindrical trunk that is free of branches for 15–18 m (Figure 10). V. surinamensis is called ‘babun udu’ in Suriname, because the reddish-brown-color of the hardwood (‘udu’ in Surinamse) resembles the fur color of the red howler monkey called ‘babun’ in the country [145]. The tree is much in demand for its light-weight wood and is listed as globally ‘endangered’ on the IUCN Red List of Threatened Species [320].

Figure 10.

The baboonwood Virola surinamensis (Rol. ex Rottb.) Warb. (Myristicaceae) (from: https://images.app.goo.gl/kjANY1chTGChw2DF6).

V. surinamensis produces grey seeds of about 2 cm long and 1.5 cm wide which are surrounded by a brilliant red nutritious aril that attracts several species of birds and monkeys which function as its primary seed dispersers. The seeds contain 60–70% fat (consisting of, among others, 69% myristic acid, 13% lauric acid, and 7% palmitic acid), as well as vitamins A and C and traces of oleic acid and linoleic acid [321]. The fat is called ucuúba seed butter (‘ucuúba’ is the indigenous name of the tree, ‘ucu’ meaning ‘grease’ and ‘yba’ meaning ‘tree’) and is edible, but is also used in a wide assortment of skin-conditioning products [322].

Leaf, fruit, stembark, and root of V. surinamensis are extensively used for preparing traditional medicines, particularly by the Indigenous peoples of the Amazon including those from Suriname. Some of the many indications are rheumatism, arthritis, and inflammation; coughing and other symptoms of respiratory disease; aphthae, mouth sores, and abscessed teeth; conjunctivitis; skin problems including rash caused by scabies, infected wounds and bacterial infections such as erysipelas; gastrointestinal troubles such as dyspepsia, ulcers, gastritis, intestinal worms, and bloody diarrhea; hemorrhoids and piles; malaria; bladder problems; infertility; and fungal infections [145, 151, 323, 324]. Interestingly, several Amazonian tribes process the red sap from the inner bark into a hallucinogenic snuff or an intoxicating paste [325]. This substance contains psychoactive substances including hallucinogenic tryptamines such as N, N-dimethyltryptamine and 5-methoxy-N, N-dimethylpritmamine, and psychedelic β-carbolines such as harmine, harmaline, and tetrahydroharmine [325]. The sap would also be efficacious against ringworm, oral thrush, and jock itch [323].

Pharmacological studies have shown substantial activity of V. surinamensis preparations against the cercaria of the parasitic flatworm Schistosoma mansoni that causes bilharzia [326]; the tripomastigote forms of Trypanosoma cruzi, the causative agent of Chagas disease [327, 328]; cultured promastigotes and/or amastigotes of several species of Leishmania that are responsible for the severe forms of leishmaniasis including kala-azar [329, 330]; the larvae of Aedes aegypti, a known factor of dengue fever, chikungunya, Zika fever, and yellow fever viruses [331]; those of the oriental latrine fly Chrysomya megacephala that can cause accidental myiasis [332]; and the trophozoite stages of Plasmodium falciparum that causes malaria [324]. These activities might be attributable to various lignans and neolignans such as grandisin in the leaf and twig of the plant and/or constituents in the leaf essential oil such as the sesquiterpenoid nerolidol [327, 328, 329, 330, 331, 332]. Grandisin also elicited anti-inflammatory and antinociceptive effects in laboratory mice [333]. And an ethanolic extract of the resin stem bark (that contains the antioxidant flavonoid epicatechin) prevented the development of ethanol-, indomethacin-, stress-, or pylorus ligature-induced ulcers in laboratory mice [334].

Pharmacological support for the folkloristic claims of antifungal effects of V. surinamensis is scant. A dichloromethane extract from the root and the fractions obtained from this extract, were active against cultures of Cladosporium cladosporioides that can cause fruit rot disease in red wine grapevine but also allergies and asthma in humans [194]. Furthermore, an ethanol extracts from the stembark was active against several species of Candida [239]. The antifungal activity has been attributed to the flavonoids 7-hydroxyflavanone and 7-hydroxy-4′-methoxyisoflavone and was ten times greater than that found for nystatin [194], but also to (neo)lignans in leaf and seed of the plant [335]. In the latter case, the antifungal activity has been associated with the inhibition of the polymerization or assembly of the fungal cell wall [336]. Interestingly, the saturated fatty acid capric acid (or decanoic acid) in V. surinamensis seed, leaf, and bark elicited meaningful activity against the parasitic dermatophyte Microsporum gypseum [197] and forty isolates of Candida [196]. Capric acid also displayed synergism with fluconazole and nystatin against drug-resistant Candida isolates from oral thrush in neonates [196]. However, the amounts of this compound in at least V. surinamensis seed (around 1%) [321] seem too low to be responsible for the antifungal activity.

5.7 Myrtaceae—Psidium guajava L

The common guava Psidium guajava L. is an evergreen tree that grows to a height of 3–10 m and is native to South America, Central America, and the Caribbean. It is widely cultivated for its edible fruit in many tropical and subtropical regions. Depending on the cultivar, the fruit can be ovoid or pear-shaped; have a green, yellow, or red skin and white-, pink-, or red-colored fleshy pulp; and range in size from as small as an apricot to as large as a grapefruit (Figure 11). Edible oil is obtained from the seeds [337], and extracts from the fruit are included in various cosmetic products such as shampoos to confer a pleasantly fresh scent [338].

Figure 11.

The guava Psidium guajava L. (Myrtaceae) (from: https://images.app.goo.gl/Cdw93GHRJaJ2jHYg7).

Preparations from the young and ripe fruit as well as leaf, stem, stembark, and rootbark from P. guajava are used in many traditional medical systems in Africa, Asia, Central America, and the Caribbean. A few indications are diarrhea, constipation, spasms, and ulcers; colds, sore throat, coughs, laryngitis, and bronchitis; menstrual disorders; gonorrhea; inflammatory disorders such as rheumatism; diabetes mellitus; hypertension; sprains and pains; vertigo, epilepsy, and convulsions; a wide variety of skin complaints; as well as parasitic and microbial infections including those causing vaginal discharge and ringworm [339, 340]. In Suriname, P. guajava preparations are, in addition, used in genital steam baths for cleansing the vagina and preventing microbial infections [155].

Some of the traditional uses of P. guajava have been validated by phytochemical and pharmacological studies (see, for instance, references [341, 342]). Aqueous and ethanolic leaf extracts elicited antidiarrheal activity in mice and rats treated with Castor oil [343] that was comparable to that caused by loperamide and might occur through inhibition of gut motility [343]. The leaf essential oil was efficacious in diarrhea associated with infantile rotaviral enteritis [344]. And the polyphenolic fraction of an aqueous leaf extract displayed spasmolytic activity in an isolated guinea-pig ileum preparation precontracted with acetylcholine and/or KCl [345]. Notably, a double-blind clinical study with a phytodrug called QG5 developed from P. guajava leaf, produced a decrease in the duration of abdominal pain, presumably due to the antispasmodic activity of quercetin in the preparation [346].

The presumed efficacy of P. guajava against respiratory disorders was supported by the antitussive activity of leaf extracts in rats and guinea pigs exposed to a capsaicin aerosol [347]. Evidence for anti-inflammatory and antinociceptive activity of the plant was provided by the inhibitory effects of an aqueous leaf extract on fresh egg albumin-induced paw edema in rats and thermally and chemically induced nociceptive pain in mice [348]. And indications for central modulatory effects of P. guajava came from the suppression of central nervous system activity and the prolongation of pentobarbital-induced sleeping time in mice that had been given methanol leaf extracts or non-polar fractions therefrom [349, 350]. These observations have been attributed to terpenes and possibly also flavonoids in the extracts [349, 351].

Studies with both laboratory animals and human subjects also supported the antihypotensive and antidiabetic activities of P. guajava. Leaf preparations produced vasorelaxation in spontaneously-contracting portal veins as well as rat aortic ring preparations of normotensive rats [352]. Importantly, randomized clinical studies found that the consumption of the fruit led to small but statistically significant positive effects on blood pressure, total blood cholesterol and triglyceride levels, as well as HDL cholesterol in hypertensive patients [353, 354]. And intraperitoneal administration of an aqueous extract of the unripe fruit or from the fruit peel led to clear hypoglycemic and antidiabetic activities in streptozotocin-treated diabetic rats [355]. The ripe fruit without peel was more efficacious in diabetic patients when compared to the fruit with peel [356].

Evidence for antiparasitic activity of P. guajava was provided by the in vitro inhibitory effect of a polyphenolic fraction of an aqueous leaf extract on the growth of Entamoeba histolytica (that causes amoebic dysentery) [345]; the potent activity of an aqueous stembark extract against the malaria parasite Plasmodium falciparum in a lactate dehydrogenase assay [357]; and the meaningful anticestodal efficacy of an aqueous leaf extract against experimental Hymenolepis diminuta (tapeworm) infection in rats [358].

Support for antibacterial effects of P. guajava came from the activity of aqueous and organic extracts from the leaf and the stembark against the main causative agent of acne lesions, Propionibacterium acnes, and other organisms isolated from these lesions [359]; two clinical isolates of the gastrointestinal tract, Escherichia coli and Staphylococcus aureus [360]; a wide variety of clinically important bacterial strains (see, for instance, references [198, 361]); as well as multidrug-resistant clinical isolates of Staphylococcus aureus [362] and a multidrug-resistant strain of Vibrio cholera [363]. The antibacterial activity might be associated with inhibition of adherence by certain lectins in the fruit [364] or phenolic compounds including flavonoids in the leaves [198, 361].

Most studies on the antifungal activity of P. guajava have been carried out with preparations of the leaf from the plant. Thus, extracts from this part of the plant obtained with various organic solvents or hot water elicited broad and meaningful activity against species in the genera Candida, Saccharomyces, Cryptococcus, Trichosporon, Aspergillus, Sporothrix, and Microsporum [198, 199, 200, 201, 202, 203]. The antifungal activities in the leaf preparations potentiated that of fluconazole [203, 204] and have been attributed to phenolic compounds including tannins, coumarins, and flavonoids such as quercetin, and/or terpenoids [198, 201, 203, 361]. The former supposition is supported by the activity of tannic and flavonoid fractions from the leaf against several species of Candida [204] and the inhibitory effects of plant phenolic compounds on the biosynthesis of ergosterol for assembling the fungal plasma membrane [365].

As well, a methanolic extract from the ripe fruit was active against strains of Arthrinium sacchari that can cause damping-off disease of wheat (collapse and overgrowth of emerging seedlings), as well as the melanin-containing ‘black yeast’ Chaetomium funicola that can cause pheohyphomycosis, a dark-brown-colored infection of skin and subcutis, paranasal sinuses, or central nervous system [206]. Furthermore, a tincture of P. guajava bark effectively killed various dermatophytes in the genera Trichophyton and Microsporum [205], while organic extracts from the twigs inactivated Candida albicans and Aspergillus niger [204].

5.8 Rananculaceae—Nigella sativa L

The black caraway (or black cumin) Nigella sativa L. is an annual flowering plant that is native to a large region that includes eastern Mediterranean, northern Africa, the Indian Subcontinent, and western Asia. It grows 20–30 cm tall, and bears relatively large fruits made up of an inflated capsule composed of three to seven united follicles, each containing numerous black seeds. The plant is mainly cultivated for the edible seeds (called ‘black cumin seeds’; Figure 12) which have a pungent, bitter taste and smell, and are used as a spice, flavoring, and food preservative [366].

Figure 12.

The black caraway Nigella sativa L. (Rananculaceae) (from: https://images.app.goo.gl/s29DmisCFu3opVT78).

In addition, the seeds and the seed oil (called ‘black seed oil’) are widely used for treating, among others, inflammatory conditions such as rheumatism, as well as respiratory conditions, gastrointestinal problems, diabetes mellitus; elevated blood cholesterol levels, a weakened immune system, parasitic infections, and microbial infections such as athlete’s foot, nail infections, ringworm, and thrush [367, 368, 369]. Relatively recently, Ayurvedic supplements of N. sativa seed oil in capsule or softgel form have been promoted in various countries including Suriname as a remedy for several of these conditions including skin, hair, and nail problems.

Some of the therapeutic properties of N. sativa have been attributed to the monoterpene thymoquinone, a major bioactive component of the seed essential oil [207] that is virtually non-toxic to humans [208]. For instance, exposure of cultured pancreatic cancer cells to thymoquinone led to a reduction in markers of inflammation [209]. This was consistent with the inhibitory effects of a ground seed extract on inflammation in the cerebellum and medulla of a rat model of multiple sclerosis [210] and with the improvement of inflammation markers in the serum of patients with rheumatoid arthritis who had received the seed oil [211]. Furthermore, N. sativa seed oil protected rats from developing stomach ulcers following exposure to acetylsalicylic acid [370] and from liver damage caused by aflatoxin B1 [212], and caused a decrease in levels of blood sugar [213], serum triglycerides and LDL [214]. N. sativa seed preparations and seed oil also elicited meaningful antimicrobial activity. Antibacterial effects were seen with a topical formulation against a staphylococcal skin infection in children [215] and against many cultured bacterial strains including methicillin-resistant Staphylococcus aureus isolates from the wounds of diabetic patients [216, 371].

Support for antifungal activity of N. sativa was provided by the inhibitory effects of diethyl ether, methanol, and aqueous seed extracts on the growth of Candida albicans in vitro [372] and on the infestation of liver, spleen, and kidneys of mice inoculated with the fungus [373]. The extracts also inactivated various dermatophytic fungi [217, 218], albeit to a lesser extent when compared to griseofulvin [217]. However, another study only found an inhibitory effect of organic seed extracts on the formation of Candida albicans colonies in the organs of experimental animals, not of aqueous extracts [374]. Thymoquinone also inhibited the growth of dermatophytes [217] as well as that of intravaginally administered Candida albicans in mice immunosuppressed by subcutaneously injected methyl prednisolone [375]. The antifungal activities of thymoquinone (that is rather lipophilic) presumably occurred as a result of the perturbation of fungal plasma membrane proteins [375].

Furthermore, the seed essential oil of N. sativa exerted excellent and broad activity against various species of Aspergillus, Curvularia, Microsporum, Penicillium, Trichoderma, Candida, Chaetomium, Fusarium, Trichophyton, and Chrysosporium [376, 377, 378, 379, 380]. However, in another study, the oil did not affect the growth of Aspergillus flavus and Aspergillus parasiticus in vitro, but inhibited the production of aflatoxin B1 by at least one-third [381]. The antifungal activity of N. sativa seeds has been hypothesized to be attributable to (an) ingredient(s) that kill(s) fungi by stimulating the production of nitric oxide by host granulocytes and monocytes [382]. A subsequent study reported two peptides called defensins and designated Ns-D1 and Ns-D2 that displayed strong activity against phytopathogenic fungi, presumably by interacting with specific sphingolipids in the fungal plasma membrane [383].

5.9 Rutaceae—Aegle marmelos (L.) Corrêa

The bael tree or Aegle marmelos (L.) Corrêa is a slow-growing deciduous tree that can reach a height of 10–15 m and is native to the northern parts of India. It is mainly cultivated for its edible aromatic, sweet-tasting, yellow-colored fruit (Figure 13) in that country as well as in other south-eastern Asian countries such as Sri Lanka, Thailand, and Malaysia. Young leaves and shoots of A. marmelos are eaten as salad greens and contain the alkaloid aegeline (N-[2-hydroxy-2 (4-methoxyphenyl) ethyl]-3-phenyl-2-propenamide) that would reduce appetite, produce weight loss, improve athletic performance, and increase energy [384]. For these reasons, aegeline has been incorporated in dietary supplements that have been marketed for weight loss and muscle building [385], even though there is no sound scientific evidence to support these uses. More importantly, aegeline has been associated with potentially fatal liver damage [386], and the more than fifty cases of acute hepatitis including several instances of fatal, acute liver failure have led to the withdrawal from the market of an aegeline-based slimming product called OxyElite Pro® [386].

Figure 13.

The Indian bael tree Aegle marmelos (L.) Corrêa (Rutaceae) (from: https://images.app.goo.gl/BkkX6CZgxx1q5Whv7).

Nevertheless, A. marmelos is extensively used in the traditional medical systems of the Indian subcontinent [387] and those of the Indian diaspora in Suriname [155]. Preparations from leaf, stembark, root, fruit, and seed are used against, among others, stomach pain, peptic ulcers, diarrhea, amoebic dysentery, constipation, and jaundice; rheumatoid arthritis, gout, and swollen joints; diabetes mellitus; high blood pressure; asthma; anemia; bone fractures; agitation and insomnia; wounds; skin problems such as leucoderma; as well as microbial infections [387, 388].

Pharmacological studies have validated many of the ethnomedicinal uses of A. marmelos (see, for instance, references [388, 389]). Thus, a chloroform root extract was effective against castor oil-induced diarrhea in laboratory rodents and inhibited the in vitro growth of various cultured diarrhea-causing species of bacteria—particularly Vibrio cholerae, Escherichia coli, and Shigella spp.—at a comparable degree as the fluoroquinolone antibiotic ciprofloxacin [390]. A methanolic leaf extract elicited considerable anti-inflammatory, antinociceptive, and antipyretic activities in rats and mice [391]. And leaf, callus, and fruit extracts displayed notable hypoglycemic and antihyperlipidemic effects in alloxan- and streptozotocin-treated mice, rats, and rabbits [392, 393, 394]. This was presumably due to the stimulation of glucose uptake by the peripheral tissues [392] and/or insulin secretion from the pancreatic β-cells [392, 393].

A methanolic leaf extract formulated as an ointment or an injectable preparation for intraperitoneal administration, stimulated the healing of excision and incision wounds in rats to a comparable degree as the topical nitrofuran antimicrobial compound nitrofurazone [395]. Preparations from A. marmelos leaf, fruit, seed, and root also displayed antibacterial and antifungal activity. The antibacterial activity was directed against a broad range of Gram-positive as well as and Gram-negative bacteria (see, for instance, references [219, 220, 396]) including multidrug resistant strains [396]. Some of the pharmacological activities of A. marmelos have been attributed to the presence of coumarins, monoterpenoids (such as cuminaldehyde), phenolic compounds (such as flavonoids and eugenol), alkaloids, tannins, and essential oils in the plant [221, 222].

Evidence for antifungal properties of A. marmelos was provided by the meaningful activity of the leaf essential oil against a variety of yeast-like and filamentous fungal species including clinical isolates of dermatophytic fungi such as those causing ringworm [219, 220, 221, 222, 223, 224, 225, 226, 227]. Furthermore, the essential oil of the plant completely inhibited spore germination of dermatophytic and plant pathogenic Fusarium fungal species in vitro, including that of a highly resistant species, Fusarium udum, the causative agent of wilt disease in the pigeon pea (characterized by the disruption of the flow of water in the xylem transport tissue of the plant) [227]. In addition, water, methanol, and ethanol leaf extracts as well as fractions from them, exerted potent antifungal activity against clinical isolates of dermatophytic fungi in the genera Trichophyton, Microsporum, and Epidermophyton [397].

As well, fractions from chloroform leaf and fruit extracts showed substantial activity in pot cultures against the plant pathogenic fungi Fusarium oxysporum f.sp. ciceris [228] and Pythium debaryanum [398], the causative agents of wilt of chickpea, and damping-off in peanut, beet, and tobacco, respectively. Notably, the addition of the leaf methanol extract to the bio-agents Trichoderma viride and/or Pseudomonas fluorescens suppressed chickpea wilt disease more when compared to either substance alone [228]. Preparations from A. marmelos seed also exhibited considerable in vitro activity against various species of Trichophyton and Aspergillus, as well as Epidermophyton floccosum (that causes skin and nail infections in humans) and Candida albicans [399, 400, 401, 402]. And an ethanolic extract of the root showed activity against Aspergillus fumigatus and Trichophyton mentagrophytes [403]. The antifungal activity of the leaf essential oil has been attributed to certain terpenoids that inhibited spore germination by interfering with Ca2+-dipicolonic acid metabolism [220, 227], a metabolic pathway involved in the regulation of the resilience of macromolecules including DNA to stressors such as heat [404].

Advertisement

6. Concluding remarks

Since their emergence on Earth, humans have explored the biodiversity to improve their well-being [405]. For this purpose, various plants, animals, and micro-organisms have been used as food, spices, shelter, poisons on arrow- and spearheads for hunting and warfare, cosmetics, stimulants, as well as medicines. Insight into the latter uses has provided unlimited opportunities for new drug leads because of nature’s enormous chemical diversity and the production of a myriad of drugs that became an essential part of health care systems throughout the world. For instance, the antineoplastic agents vincristine and paclitaxel have first been identified in the periwinkle plant Catharanthus roseus [406] and the Pacific yew Taxus brevifolia [407], respectively. The angiotensin-converting enzyme inhibitor captopril and its derivatives for treating hypertension and some types of congestive heart failure, have been developed from the venom of the Brazilian viper Bothrops jararaca [408]. And exenatide for treating type 2 diabetes mellitus was originally isolated from the venom of the Gila monster Heloderma suspectum [409].

The bacterial species Saccharopolyspora erythraea and Streptomyces nodosus gave the antibacterial agent erythromycin [410], and the antifungal and antileishmanial agent amphotericin B [411], respectively. And as mentioned throughout this monograph, the fungal species Penicillium chrysogenum, Tolypocladium inflatum, and Aspergillus terreus were at the basis of identifying β-lactam antibiotics such as the penicillins [12], the immunosuppressive agent tacrolimus [13], and the specific LDL-lowering HMG-CoA reductase-inhibiting statins that reduce the risk of arterial blockage, a heart attack, a stroke, and diabetes mellitus [14].

This monograph has presented data about the antifungal activities of nine plants that are traditionally used for treating mycoses in Suriname. The plants were the taro Colocasia esculenta (Araceae), the milkweed Euphorbia hirta (Euphorbiaceae), the holy basil Ocimum tenuiflorum (Lamiaceae), the avocado Persea americana (Lauraceae), the pomegranate Punica granatum (Lythraceae), the baboonwood Virola surinamensis (Myristicaceae), the guava Psidium guajava (Myrtaceae), the black caraway Nigella sativa (Rananculaceae), and the golden apple Aegle marmelos (Rutaceae). Preparations from these plants showed good and broad antifungal properties in vitro. In addition, most of the samples also displayed various other pharmacological activities including antiparasitic, antibacterial, and antiviral activity. An extract of P. granatum fruit formulated as a gel even elicited antifungal activity in individuals with candidosis associated with denture stomatitis [186]. And there are in most cases also valuable indications about the pharmacologically active compound(s) and the mechanism(s) of action.

These data are encouraging, providing preliminary indications of the usefulness and potential toxicity of the plant samples, but do not reflect their clinical efficacy and usefulness. It is therefore imperative to properly evaluate the plant preparations for their chemical stability, spectrum of antifungal activity in the clinical setting, susceptibility to efflux pumps, bioavailability and degree of protein binding and inactivation by serum and hepatic enzyme systems, and many other pharmacokinetic and pharmacodynamics aspects. This is all the more important because of two reasons. First, the development of resistance to available antifungal drugs including the spread of multidrug-resistant fungal strains, is becoming a growing problem in many parts of the world [412]. Second, despite the availability of modern allopathic antifungal drugs, about 80% of the world population residing in the developing world rely on herbal medicinal products as a primary source of healthcare [413]. It is just and fair to provide sound scientific evidence on the clinical efficacy of these products for treating fungal infections.

References

  1. 1. Heitman J, Howlett B, Crous PW, Stukenbrock E, James T, Gow NAR, editors. The Fungal Kingdom. Washington, DC: American Society for Microbiology; 2017. DOI: 10.1128/9781555819583
  2. 2. Berbee ML, Taylor JW. Dating the molecular clock in fungi—How close are we? Fungal Biology Reviews. 2010;24:1-16. DOI: 10.1016/j.fbr.2010.03.001
  3. 3. Redecker D, Kodner R, Graham LE. Glomalean fungi from the Ordovician. Science. 2000;289:1920-1921. DOI: 10.1126/science.289.5486.1920
  4. 4. Anonymous. Stop neglecting fungi. Nature. Microbiology. 2017;2:17120. DOI: 10.1038/nmicrobiol.2017.120
  5. 5. O'Brien BL, Parrent JL, Jackson JA, Moncalvo JM, Vilgalys R. Fungal community analysis by large-scale sequencing of enviromental samples. Applied and Environmental Microbiology. 2005;71:5544-5550. DOI: 10.1128/AEM.71.9.5544-5550.2005
  6. 6. Raspor P, Zupan J. Yeasts in extreme environments. In: Rosa C, Gábor P, editors. Biodiversity and Ecophysiology of Yeasts. Berlin, Germany: Springer-Verlag; 2006. pp. 372-417
  7. 7. Arnold AE, Lutzoni F. Diversity and host range of foliar fungal endophytes: Are tropical leaves biodiversity hotspots? Ecology. 2007;88:541-549. DOI: 10.1890/05-1459
  8. 8. Miller OK, Henkel TW, James TY, Miller SL. Pseudotulostoma, a remarkable new genus in the Elaphomycetaceae from Guyana. Mycological Research. 2001;105:1268-1272. DOI: 10.1017/S095375620100466X
  9. 9. Sterflinger K, Tesei D, Zakharova K. Fungi in hot and cold deserts with particular reference to microcolonial fungi. Fungal Ecology. 2012;5:453-462. DOI: 10.1016/j.funeco.2011.12.007
  10. 10. Murgia M, Fiamma M, Barac A, Deligios M, Mazzarello V, Paglietti B, et al. Biodiversity of fungi in hot desert sands. Microbiology Open. 2019;8:e595. DOI: 10.1002/mbo3.595
  11. 11. Frąc M, Hannula SE, Bełka M, Jędryczka M. Fungal biodiversity and their role in soil health. Frontiers in Microbiology. 2018;9:707. DOI: 10.3389/fmicb.2018.00707
  12. 12. Aminov RI. A brief history of the antibiotic era: Lessons learned and challenges for the future. Frontiers in Microbiology. 2010;1:134. DOI: 10.3389/fmicb.2010.00134
  13. 13. Tedesco D, Haragsim L. Cyclosporine: A review. Journal of Transplantation. 2012;2012:230386. DOI: 10.1155/2012/23038
  14. 14. Stossel TP. The discovery of statins. Cell. 2008;134:903-905. DOI: 10.1016/j.cell.2008.09.008
  15. 15. Legras JL, Merdinoglu D, Cornuet JM, Karst F. Bread, beer and wine: Saccharomyces cerevisiae diversity reflects human history. Molecular Ecology. 2007;16:2091-2102. DOI: 10.1111/j.1365-294X.2007.03266.x
  16. 16. Botstein D, Fink GR. Yeast: An experimental organism for 21st century biology. Genetics. 2011;189:695-704. DOI: 10.1534/genetics.111.130765
  17. 17. Goffeau A, Barrell BG, Bussey H, Davis RW, Dujon B, Feldmann H, et al. Life with 6000 genes. Science. 1996;274:563-567. DOI: 10.1126/science.274.5287.546
  18. 18. Horowitz NH, Berg P, Singer M, Lederberg J, Susman M, Doebley J, et al. A centennial: George W. Beadle, 1903-1989. Genetics. 2004;166:1-10. DOI: 10.1534/genetics.166.1.1
  19. 19. Morishita N, Sei Y. Microreview of pityriasis versicolor and Malassezia species. Mycopathologia. 2006;162:373-376. DOI: 10.1007/s11046-006-0081-2
  20. 20. Badiee P, Hashemizadeh Z. Opportunistic invasive fungal infections: Diagnosis and clinical management. Indian Journal of Medical Research. 2014;139:195-204
  21. 21. Magnussen A, Parsi MA. Aflatoxins, hepatocellular carcinoma and public health. World Journal of Gastroenterology. 2013;19:1508-1512. DOI: 10.3748/wjg.v19.i10.1508
  22. 22. Carris LM, Little CR, Stiles CM. Introduction to fungi. The Plant Health Instructor. 2012. DOI: 10.1094/ PHI-I-2012-0426-01. Available from: https://www.apsnet.org/edcenter/disandpath/fungalasco/intro/Pages/IntroFungi,aspx [Accessed: August 02, 2021]
  23. 23. Sant D, Tupe S, Ramana C, Deshpande M. Fungal cell membrane—Promising drug target for antifungal therapy. Journal of Applied Microbiology. 2016;121:1498-1510. DOI: 10.1111/jam.13301
  24. 24. Garcia-Rubio R, de Oliveira HC, Rivera J, Trevijano-Contador N. The fungal cell wall: Candida, Cryptococcus, and Aspergillus species. Frontiers in Microbiology. 2020;10:2993. DOI: 10.3389/fmicb.2019.02993
  25. 25. Kurtzman CP, Piškur J. Taxonomy and phylogenetic diversity among the yeasts (Chapter 15). In: Sunnerhagen P, Piskur J, editors. Comparative Genomics: Using Fungi as Models. Topics in Current Genetics. Berlin, Germany: Springer; 2006. pp. 29-46. DOI: 10.1007/b106654.
  26. 26. Yong E. Yeast suggests speedy start for multicellular life. Nature. 2012. DOI: 10.1038/nature.2012.9810. Available from: https://www.nature.com/articles/nature.2012.9810 [Accessed: August 03, 2021]
  27. 27. Chavez JA, Brat DJ, Hunter SB, Vega JV, Guarner J. Practical diagnostic approach to the presence of hyphae in neuropathology specimens with three illustrative cases. American Journal of Clinical Pathology. 2018;149:98-104. DOI: 10.1093/ajcp/aqx144
  28. 28. Walker K, Skelton H, Smith K. Cutaneous lesions showing giant yeast forms of Blastomyces dermatitidis. Journal of Cutaneous Pathology. 2002;29:616-618. DOI: 10.1034/j.1600-0560.2002.291009.x
  29. 29. Bennett JW. An overview of the genus Aspergillus (Chapter 1). In: Machida M, Katsuya Gomi K, editors. Aspergillus: Molecular Biology and Genomics. Norfolk, UK: Caister Academic Press; 2010. pp. 1-17
  30. 30. Yadav AN, Verma P, Kumar V, Sangwan P, Mishra S, Panjiar N, et al. Biodiversity of the genus Penicillium in different habitats (Chapter 1). In: Gupta VK, editor. New and Future Developments in Microbial Biotechnology and Bioengineering. Amsterdam, The Netherlands: Elsevier; 2018. pp. 3-18. DOI: 10.1016/B978-0-444-63501-3.00001-6
  31. 31. Zheng R-Y, Chen G-Q, Huang H, Liu X-Y. A monograph of Rhizopus. Sydowia. 2007;59:273-372
  32. 32. Ely JW, Rosenfeld S, Stone MS. Diagnosis and management of tinea infections. American Family Physician. 2014;90:702-711
  33. 33. Parniske M. Arbuscular mycorrhiza: the mother of plant root endosymbioses. Nature Reviews. Microbiology. 2008;6:763-775. DOI: 10.1038/nrmicro1987
  34. 34. Gauthier GM. Fungal dimorphism and virulence: molecular mechanisms for temperature adaptation, immune evasion, and in vivo survival. Mediators of inflammation. 2017;2017:8491383. DOI: 10.1155/2017/8491383
  35. 35. Chandler JM, Treece ER, Trenary HR, Brenneman JL, Flickner TJ, Frommelt JL, et al. Protein profiling of the dimorphic, pathogenic fungus. Penicillium marneffei. Proteome Science. 2008;6:17. DOI: 10.1186/1477-5956-6-17
  36. 36. Dhaliwal MS, Jindal SK, Sharma A, Prasanna HC. Tomato yellow leaf curl virus disease of tomato and its management through resistance breeding: A review. Journal of Horticultural Science and Biotechnology. 2020;95:425-444. DOI: 10.1080/14620316.2019.1691060
  37. 37. Purdy LH, Schmidt RA. Status of cacao witches’ broom: Biology, epidemiology, and management. Annual Review of Phytopathology. 1996;34:573-594. DOI: 10.1146/annurev.phyto.34.1.573
  38. 38. Huerta-Espino J, Singh RP, Germán S, McCallum BD, Park RF, Chen WQ, et al. Global status of wheat leaf rust caused by Puccinia triticina. Euphytica. 2011;179:143-160. DOI: 10.1007/s10681-011-0361-x
  39. 39. Sanders WB. Lichens: The interface between mycology and plant morphology. BioScience. 2001;51:1025-1036. DOI: 10.1641/0006-3568(2001)051[1025:ltibma]2.0.co;2
  40. 40. Huang G-M, Zou Y-N, Wu Q-S, Xu Y-J, Kuča K. Mycorrhizal roles in plant growth, gas exchange, root morphology, and nutrient uptake of walnuts. Plant, Soil and Environment. 2020;66:295-302. DOI: 10.17221/240/2020-PSE
  41. 41. Stajich JE, Berbee ML, Blackwell M, Hibbett DS, Taylor JW. The fungi. Current Biology. 2009;19:R840-R845
  42. 42. Hyde KD, Al-Hatmi AMS, Andersen B, Boekhout T, Buzina W, Dawson TL, et al. The world's ten most feared fungi. Fungal Diversity. 2018;93:161-194. DOI: 10.1007/s13225-018-0413-9
  43. 43. Hibbett DS, Binder M, Bischoff JF, Blackwell M, Cannon PF, Eriksson OE, et al. A higher-level phylogenetic classification of the fungi. Mycological Research. 2007;111:509-547. DOI: 10.1016/j.mycres.2007.03.004
  44. 44. Didier ES. Microsporidiosis: An emerging and opportunistic infection in humans and animals. Acta Tropica. 2005;94:61-76. DOI: 10.1016/j.actatropica.2005.01.010
  45. 45. Longcore JE, Pessier AP, Nichols DK. Batrachochytirum dendrobatidis gen. et sp. nov., a chytrid pathogenic to amphibians. Mycologia. 1999;91:219-227. DOI: 10.1080/00275514.1999.12061011
  46. 46. Letcher PM, Lee PA, Lopez S, Burnett M, McBride RC, Powell MJ. An ultrastructural study of Paraphysoderma sedebokerense (Blastocladiomycota), an epibiotic parasite of microalgae. Fungal Biology. 2016;120:324-337. DOI: 10.1016/j.funbio.2015.11.003
  47. 47. Orpin CG. Studies on the rumen flagellate Neocallimastix frontalis. Journal of General Microbiology. 1975;91:249-262. DOI: 10.1099/00221287-91-2-249
  48. 48. Rodríguez M, Pérez D, Chaves FJ, Esteve E, Garcia PM, Xifra G, et al. Obesity changes the human gut mycobiome. Scientific Reports. 2015;5:14600. DOI: 10.1038/srep14600
  49. 49. Srinivasan K, Murakami M, Nakashimada Y, Nishio N. Efficient production of cellulolytic and xylanolytic enzymes by the rumen anaerobic fungus, Neocallimastix frontalis, in a repeated batch culture. Journal of Bioscience and Bioengineering. 2001;91:153-158. DOI: 10.1016/S1389-1723(01)80058-X
  50. 50. Brundrett M. Mycorrhizal associations and other means of nutrition of vascular plants: Understanding the global diversity of host plants by resolving conflicting information and developing reliable means of diagnosis. Plant and Soil. 2009;320:37-77. DOI: 10.1007/s11104-008-9877-9
  51. 51. Mandyam KG, Jumpponen A. Mutualism-parasitism paradigm synthesized from results of root-endophyte models. Frontiers in Microbiology. 2014;5:1-13. DOI: 10.3389/fmicb.2014.00776
  52. 52. Osherov N, May GS. The molecular mechanisms of conidial germination. FEMS Microbiology Letters. 2001;199:153-160. DOI: 10.1111/j.1574-6968.2001.tb10667.x
  53. 53. Ferreira JA, Mahboubi A, Lennartsson PR, Taherzadeh MJ. Waste biorefineries using filamentous ascomycetes fungi: Present status and future prospects. Bioresource Technology. 2016;215:334-345
  54. 54. Gmoser R, Ferreira JA, Lennartsson PR, Taherzadeh MJ. Filamentous ascomycetes fungi as a source of natural pigments. Fungal Biology and Biotechnology. 2017;4:4. DOI: 10.1186/s40694-017-0033-2
  55. 55. Kolmer JA, Ordonez M, Groth J. The rust fungi (Chapter 1). In: John Wiley & Sons Ltd., editor. Encyclopedia of Life Sciences. Hoboken, NJ, USA: Wiley; 2019. pp. 1-9. DOI: 10.1002/9780470015902.a0021264.pub2
  56. 56. Valverde ME, Paredes-López O, Pataky JK, Guevara-Lara F. Huitlacoche (Ustilago maydis) as a food source—Biology, composition, and production. Critical Reviews in Food Science and Nutrition. 1995;35:191-229. DOI: 10.1080/10408399509527699
  57. 57. Cabañes FJ. Malassezia yeasts: how many species infect humans and animals? PLOS Pathogens. 2014;10:e1003892. DOI: 10.1371/journal.ppat.1003892
  58. 58. Perfect JR. Cryptococcus neoformans: The yeast that likes it hot. FEMS Yeast Research. 2006;6:463-468. DOI: 10.1111/j.1567-1364.2006.00051.x
  59. 59. Rivera-Mariani FE, Bolaños-Rosero B. Allergenicity of airborne basidiospores and ascospores: Need for further studies. Aerobiologia. 2011;28:83-97. DOI: 10.1007/s10453-011-9234-y
  60. 60. McLaughlin DJ, Frieders EM, Lü H. A microscopist’s view of heterobasidiomycete phylogeny. Studies in Mycology. 1995;38:91-109
  61. 61. Beard M, North JA, Price SRF. Religions of Rome: Volume 1, a History. Cambridge, UK: Cambridge University Press; 1998
  62. 62. Sharma M, Kulshrestha S. Colletotrichum gloeosporioides: An anthracnose causing pathogen of fruits and vegetables. Biosciences Biotechnology Research Asia. 2015;12:1233-1246. DOI: 10.13005/bbra/1776
  63. 63. Williamson B, Tudzynski B, Tudzynski P, van Kan JAL. Botrytis cinerea: The cause of grey mould disease. Molecular Plant Pathology. 2007;8:561-580. DOI: 10.1111/j.1364-3703.2007.00417.x
  64. 64. Gryndler M, Krofta K, Gryndlerová H, Soukupová L, Hršelová H, Gabriel J. Potentially dangerous fusarioid microorganisms associated with rot of hop (Humulus lupulus L.) plants in field culture. Plant, Soil and Environment. 2008;54:149-154
  65. 65. Pérez-García A, Romero D, Fernández-Ortuño D, López-Ruiz F, De Vicente A, Torés Montosa JA. The powdery mildew fungus Podosphaera fusca (synonym Podosphaera xanthii), a constant threat to cucurbits. Molecular Plant Pathology. 2009;10:53-60. DOI: 10.1111/j.1364-3703.2008.00527
  66. 66. Capasso L. 5300 years ago, the Ice Man used natural laxatives and antibiotics. The Lancet. 1998;352:1864-1864. DOI: 10.1016/S0140-6736(05)79939-6
  67. 67. Sahoo AK, Mahajan R. Management of tinea corporis, tinea cruris, and tinea pedis: A comprehensive review. Indian Dermatology Online Journal. 2016;7:77-86. DOI: 10.4103/2229-5178.178099
  68. 68. Fujie A. Discovery of micafungin (FK463): A novel antifungal drug derived from a natural product lead. Pure and Applied Chemistry. 2007;79:603-614. DOI: 10.1351/pac200779040603
  69. 69. Liang H. Sordarin, an antifungal agent with a unique mode of action. Beilstein Journal of Organic Chemistry. 2008;4:31. DOI: 10.3762/bjoc.4.31
  70. 70. Spadaro D, Lorè A, Amatulli MT, Garibaldi A, Gullino ML. First report of Penicillium griseofulvum causing blue mold on stored apples in Italy (Piedmont). Plant Disease. 2011;95:76. DOI: 10.1094/PDIS-08-10-0568
  71. 71. Tadych M, Bergen MS, Johnson-Cicalese J, Polashock JJ, Vorsa N, White JF Jr. Endophytic and pathogenic fungi of developing cranberry ovaries from flower to mature fruit: Diversity and succession. Fungal Diversity. 2012;54:101-116. DOI: 10.1007/s13225-012-612 0160-2
  72. 72. Barrasa JM, Lundqvist G, Moreno G. Notes on the genus Sordaria in Spain. Sordaria elongatispora, a new coprophilous species (Pyrenomycetes). Persoonia—Molecular Phylogeny and Evolution of Fungi. 1986;13:83-88
  73. 73. Deb D, Khan A, Dey N. Phoma diseases: Epidemiology and control. Plant Pathology. 2020;69:1203-1217. DOI: 10.1111/ppa.13221
  74. 74. Baxter A, Fitzgerald BJ, Hutson JL, McCarthy AD, Motteram JM, Ross BC, et al. Squalestatin 1, a potent inhibitor of squalene synthase, which lowers serum cholesterol in vivo. Journal of Biological Chemistry. 1992;267:11705-11708
  75. 75. Tfelt-Hansen P, Saxena PR, Dahlöf C, Pascual J, Láinez M, Henry P, et al. Ergotamine in the acute treatment of migraine: A review and European consensus. Brain. 2000;123:9-18. DOI: 10.1093/brain/123.1.9
  76. 76. Van Dongen PW, de Groot AN. History of ergot alkaloids from ergotism to ergometrine. European Journal of Obstetrics, Gynecology, and Reproductive Biology. 1995;60:109-116. DOI: 10.1016/0028-2243(95)02104-z
  77. 77. Ropars J, Cruaud C, Lacoste S, Dupont J. A taxonomic and ecological overview of cheese fungi. International Journal of Food Microbiology. 2012;155:199-210. DOI: 10.1016/j.ijfoodmicro.2012.02.005
  78. 78. McGee CF. Microbial ecology of the Agaricus bisporus mushroom cropping process. Applied Microbiology and Biotechnology. 2018;102:1075-1083. DOI: 10.1007/s00253-017-8683-9
  79. 79. Pegler DN. Useful fungi of the world: Morels and truffles. Mycologist. 2003;17:174-175. DOI: 10.1017/S0269915X04004021
  80. 80. Wiebe M. Mycoprotein from Fusarium venenatum: A well-established product for human consumption. Applied Microbiology and Biotechnology. 2002;58:421-427. DOI: 10.1007/s00253-002-0931-x
  81. 81. Grewal HS, Kalra KL. Fungal production of citric acid. Biotechnology Advances. 1995;13:209-234. DOI: 10.1016/0734-9750(95)00002-8
  82. 82. Peleg Y, Stieglitz B, Goldberg I. Malic acid accumulation by Aspergillus flavus. I. Biochemical aspects of acid biosynthesis. Applied Microbiology and Biotechnology. 1988;28:69-75. DOI: 10.1007/BF00250501
  83. 83. Zhang ZY, Jin B, Kelly JM. Production of lactic acid from renewable materials by Rhizopus fungi. Biochemical Engineering Journal. 2007;35:251-263. DOI: 10.1016/j.bej.2007.01.028
  84. 84. Mahadik ND, Puntambekar US, Bastawde KB, Khire JM, Gokhale DV. Production of acidic lipase by Aspergillus niger in solid-state fermentation. Process Biochemistry. 2002;38:715-721
  85. 85. Ahmed A, Bibi A. Fungal cellulase; production andapplications: minireview. LIFE: International Journal of Health and Life Sciences. 2018;4:19-36. DOI: 10.20319/lijhls.2018.41.1936
  86. 86. Akpan I, Bankoley MO, Adesemowo AM. Production of α-amylase from Aspergillus niger using cheap medium. Tropical Science. 1999;39:77-79
  87. 87. Zhan P, Liu W. The changing face of dermatophytic infections worldwide. Mycopathologia. 2016;182:77-86. DOI: 10.1007/s11046-016-0082-8
  88. 88. Singh A, Verma R, Murari A, Agrawal A. Oral candidiasis: An overview. Journal of Oral and Maxillofacial Pathology. 2014;18:S81-S85. DOI: 10.4103/0973-029X.141325
  89. 89. Gonçalves B, Ferreira C, Alves CT, Henriques M, Azeredo J, Silva S. Vulvovaginal candidiasis: Epidemiology, microbiology and risk factors. Critical Reviews in Microbiology. 2016;42:905-927. DOI: 10.3109/1040841X.2015.1091805
  90. 90. Francesconi V, Klein AP, Santos AP, Ramasawmy R, Francesconi F. Lobomycosis: Epidemiology, clinical presentation, and management options. Therapeutics and Clinical Risk Management. 2014;10:851-860. DOI: 10.2147/TCRM.S46251
  91. 91. Orofino-Costa R, Macedo PM, Rodrigues AM, Bernardes-Engemann AR. Sporotrichosis: An update on epidemiology, etiopathogenesis, laboratory and clinical therapeutics. Anais Brasileiros de Dermatologia. 2017;92:606-620. DOI: 10.1590/abd1806-4841.2017279
  92. 92. Metin A, Dilek N, Bilgili SG. Recurrent candidal intertrigo: Challenges and solutions. Clinical, Cosmetic and Investigational Dermatology. 2018;11:175-185. DOI: 10.2147/CCID.S127841
  93. 93. Marques SA. Paracoccidioidomycosis: Epidemiological, clinical, diagnostic and treatment up-dating. Anais Brasileiros de Dermatologia. 2013;88:700-711. DOI: 10.1590/abd1806-4841.20132463
  94. 94. Nguyen C, Barker BM, Hoover S, Nix DE, Ampel NM, Frelinger JA, et al. Recent advances in our understanding of the environmental, epidemiological, immunological, and clinical dimensions of coccidioidomycosis. Clinical Microbiology Reviews. 2013;26:505-525. DOI: 10.1128/CMR.00005-13
  95. 95. Kauffman CA. Histoplasmosis: A clinical and laboratory update. Clinical Microbiology Reviews. 2007;20:115-132. DOI: 10.1128/CMR.00027-06
  96. 96. Rodrigues ML, Nosanchuk JD. Fungal diseases as neglected pathogens: A wake-up call to public health officials. PLoS Neglected Tropical Diseases. 2020;14:e0007964. DOI: 10.1371/journal.pntd.0007964
  97. 97. Setianingrum F, Rautemaa-Richardson R, Denning DW. Pulmonary cryptococcosis: A review of pathobiology and clinical aspects. Medical Mycology. 2019;57:133-150. DOI: 10.1093/mmy/myy086
  98. 98. Russo A, Tiseo G, Falcone M, Menichetti F. Pulmonary aspergillosis: An evolving challenge for diagnosis and treatment. Infectious Diseases and Therapy. 2020;9:511-524. DOI: 10.1007/s40121-020-00315-4
  99. 99. Bongomin F, Gago S, Oladele RO, Denning DW. Global and multi-national prevalence of fungal diseases—Estimate precision. Journal of Fungi. 2017;3:57. DOI: 10.3390/jof3040057
  100. 100. Brown GD, Denning DW, Gow NAR, Levitz SM, Netea MG, White TC. Hidden killers: Human fungal infections. Science Translational Medicine. 2012;4:165rv13. DOI: 10.1126/scitranslmed.3004404
  101. 101. Denning DW. Global fungal burden. Mycoses. 2013;56:13
  102. 102. Rajasingham R, Rachel MS, Benjamin JP, Joseph NJ, Nelesh PG, Tom MC, et al. Global burden of disease of HIV-associated cryptococcal meningitis: An updated analysis. The Lancet, Infectious Diseases. 2017;17:873-881. DOI: 10.1016/S1473-3099(17)30243-8
  103. 103. Marr KA, Carter RA, Boeckh M, Martin P, Corey L. Invasive aspergillosis in allogeneic stem cell transplant recipients: Changes in epidemiology and risk factors. Blood. 2002;100:4358-4366. DOI: 10.1182/blood-2002-05-1496
  104. 104. Guinea J, Torres-Narbona M, Gijón P, Muñoz P, Pozo F, Peláez T, et al. Pulmonary aspergillosis in patients with chronic obstructive pulmonary disease: Incidence, risk factors, and outcome. Clinical Microbiology and Infection. 2010;16:870-877. DOI: 10.1111/j.1469-0691.2009.03015.x
  105. 105. Limper AH, Adenis A, Le T, Harrison TS. Fungal infections in HIV/AIDS. The Lancet, Infectious Diseases. 2017;17:e334-e343. DOI: 10.1016/S1473-3099(17)30303-1
  106. 106. Te Welscher YM, van Leeuwen MR, de Kruijff B, Dijksterhuis J, Breukink E. Polyene antibiotic that inhibits membrane transport proteins. Proceedings of the National Academy of Sciences USA. 2012;109:11156-11159. DOI: 10.1073/pnas.1203375109
  107. 107. Sheehan DJ, Hitchcock CA, Sibley CM. Current and emerging azole antifungal agents. Clinical Microbiology Reviews. 1999;12:40-79
  108. 108. Stütz A. Allylamine derivatives—A new class of active substances in antifungal chemotherapy. Angewandte Chemie. 1987;26:320-328. DOI: 10.1002/anie.198703201
  109. 109. Denning DW. Echinocandins: A new class of antifungal. Journal of Antimicrobial Chemotherapy. 2002;49:889-891. DOI: 10.1093/jac/dkf045
  110. 110. Vermes A, Guchelaar HJ, Dankert J. Flucytosine: A review of its pharmacology, clinical indications, pharmacokinetics, toxicity and drug interactions. Journal of Antimicrobial Chemotherapy. 2000;46:171-179. DOI: 10.1093/jac/46.2.171
  111. 111. Queiroz-Telles F, de Hoog S, Santos DWCL, Salgado CG, Vicente VA, Bonifaz A, et al. Chromoblastomycosis. Clinical Microbiology Reviews. 2017;30:233-276. DOI: 10.1128/CMR.00032-16
  112. 112. Jessen A, Katona A. Breaking from Isolation: Suriname’s Participation in Regional Integration Initiatives. Buenos Aires: Institute for the Integration of Latin America and the Caribbean; 2001
  113. 113. Algemeen Bureau voor de Statistiek/Conservation International Suriname. Suriname in cijfers 286-2012/04. (General Bureau of Statistics/Conservation International Suriname. Suriname in numbers 286-2012/04). Milieustatistieken (Environment Statistics). Paramaribo (Suriname): Algemeen Bureau voor de Statistiek; 2012
  114. 114. United Nations Educational, Scientific and Cultural Organisation. Historic Inner City of Paramaribo. Paris, France: United Nations; 2002. Available from: https://whc.unesco.org/en/lis/940/documents/ [Accessed: August 03, 2021]
  115. 115. Algemeen Bureau voor de Statistiek/Censuskantoor. Suriname in cijfers 2013/05. Resultaten achtste (8ste) volks- en woningtelling in Suriname (volume 1) (General Bureau of Statistics/Census office. Suriname in numbers 2013/05. Results of the eight general census of Suriname). Demografische en sociale karakteristieken en migratie (Demographic and social characteristics and migration). Paramaribo (Suriname): Algemeen Bureau voor de Statistiek; 2013
  116. 116. Hammond DS. Forest conservation and management in the Guiana Shield (Chapter 1). In: Hammond DS, editor. Tropical Rainforests of the Guiana Shield. Wallingford, UK: CABI Publishing; 2005. pp. 1-14
  117. 117. Diepeveen J, Hüning M. The status of Dutch in post-colonial Suriname. In: Schmidt-Brücken D, Schuster S, Wienberg M, editors. Aspects of (Post)colonial Linguistics. Current Perspectives and New Approaches. Berlin, Germany: De Gruyter; 2016. pp. 131-155. DOI: 10.1515/9783110436907-007
  118. 118. Algemeen Bureau voor de Statistiek. Suriname in cijfers 303-2014-04 (General Bureau of Statistics Suriname in numbers 303-2014-04). Basis Indicatoren (Basic Indicators). Paramaribo (Suriname): Algemeen Bureau voor de Statistiek; 2014
  119. 119. The World Bank Group. Data - Suriname [Internet]. Washington, DC, USA: World Bank Group; 2021. Available from: https://data.worldbank.org/country/suriname?view=chart [Accessed: August 03, 2021]
  120. 120. Helman A. Cultureel mozaïek van Suriname. Bijdrage tot onderling begrip (Cultural mosaic of Suriname. a contribution to mutual understanding). Zutphen, The Netherlands: De Walburg Pers; 1977
  121. 121. Bakker E, Dalhuisen L, Donk R, Hassankhan M, Steegh F. Geschiedenis van Suriname: van stam tot staat (History of Suriname: from tribe to state). Zutphen, The Netherlands: Walburg Pers; 1998
  122. 122. Algemeen Bureau voor de Statistiek. Suriname in cijfers 345/2019-03 (General Bureau of Statistics. Suriname in Numbers 345/2019-03). Demographische Data 2015-2018 (Demographic Data 2015-2018). Paramaribo, Suriname: Algemeen Bureau voor de Statistiek; 2019
  123. 123. Chan KY, Adeloye D, Grant L, Kolčić I, Marušić A. How big is the ‘next big thing’? Estimating the burden of non-communicable diseases in low- and middle-income countries. Journal of Global Health. 2012;2:020101. DOI: 10.7189/jogh.02.0
  124. 124. Oehlers GP, Lichtveld MY, Brewster LM, Algoe M, Irving ER. Health life in Suriname (Chapter 6). In: Hassankhan MS, Roopnarine L, White C, Mahase R, editors. Legacy of Slavery and Indentured Labour. Historical and Contemporary Issues in Suriname and the Caribbean. New Delhi, India: Manohar; 2016. pp. 111-150
  125. 125. Eersel MGM, Vreden SGS, van Eer ED, Mans DRA. Fifty years of primary health care in the rainforest: Temporal trends in morbidity and mortality in indigenous Amerindian populations of Suriname. Journal of Global Health. 2018;8:020423. DOI: 10.7189/jogh.08.020403
  126. 126. The World Bank Group. Data—Suriname [Internet]. 2021. Available from: https://data.worldbank.org/indicator/SP.DYN.LE00.IN?locations=SR [Accessed: August 03, 2021]
  127. 127. World Health Organization. Non-communicable Diseases (NCD) Country Profiles. Suriname. Geneva (Switzerland): World Health Organization; 2014
  128. 128. Punwasi W. Doodsoorzaken in Suriname 2009-2011 (Causes of Daeth in Suriname 2009-2011). Paramaribo, Suriname: Ministerie van Volksgezondheid, Bureau Openbare Gezondheidszorg; 2012
  129. 129. Pan American Health Organization. Health in the Americas, 2007. Volume II—Countries. Suriname. [Internet]. 2007. Available from: https://www3.paho.org/hia2007/archivosvol2/paisesing/Suriname%20English.pdf [Accessed: August 03, 2021]
  130. 130. Worldometer. Suriname [Internet]. 2021. Available from: https://www.worldometers.info/coronavirus/country/suriname/ [Accessed: August 06, 2021]
  131. 131. Ministry of Health. Report of the Director of Health 2005-2007. Paramaribo, Suriname: Ministry of Health Republic of Suriname; 2008
  132. 132. Walsh TJ, Dixon DM. Spectrum of mycoses (Chapter 75). In: Baron S, editor. Medical Microbiology. 4th ed. Galveston, TX, USA: University of Texas Medical Branch at Galveston; 1996
  133. 133. Garber G. An overview of fungal infections. Drugs. 2001;61:1-12. DOI: 10.2165/00003495-200161001-00001
  134. 134. Segal E, Elad D. Special issue: Treatments for fungal infections. Journal of Fungi (Basel). 2018;4:135. DOI: 10.3390/jof4040135
  135. 135. Wall G, Lopez-Ribot JL. Current antimycotics, new prospects, and future approaches to antifungal therapy. Antibiotics (Basel). 2020;9:445. DOI: 10.3390/antibiotics9080445
  136. 136. Berger S. Infectious Diseases of Suriname. Los Angeles, CA, USA: Gideon Informatics, Inc.; 2020
  137. 137. Schmiedel Y, Zimmerli S. Common invasive fungal diseases: An overview of invasive candidiasis, aspergillosis, cryptococcosis, and Pneumocystis pneumonia. Swiss Medical Weekly. 2016;146:w14281. DOI: 10.4414/smw.2016.1428
  138. 138. Pappas P, Lionakis M, Arendrup M, Ostrosky-Zeichner L, Kullberg BJ. Invasive candidiasis. Nature Reviews. Disease Primers. 2018;4:18026. DOI: 10.1038/nrdp.2018.26
  139. 139. McBride JA, Gauthier GM, Klein BS. Clinical manifestations and treatment of blastomycosis. Clinics in Chest Medicine. 2017;38:435-449. DOI: 10.1016/j.ccm.2017.04.006
  140. 140. Goodwin RA Jr, Shapiro JL, Thurman GH, Thurman SS, Des Prez RM. Disseminated histoplasmosis: Clinical and pathologic correlations. Medicine (Baltimore). 1980;59:1
  141. 141. Nacher M, Adenis A, Mc Donald S, Do Socorro Mendonca Gomes M, Singh S, Lima IL, et al. Disseminated histoplasmosis in HIV-infected patients in South America: A neglected killer continues on its rampage. PLoS Neglected Tropical Diseases. 2013;7:e2319. DOI: 10.1371/journal.pntd.0002319
  142. 142. Jairam R. A historical overview of Batrachochytrium dendrobatidis infection from specimens at the National Zoological Collection Suriname. PLoS ONE. 2020;15:e0239220. DOI: 10.1371/journal.pone.0239220
  143. 143. Mans DRA, Ganga D, Kartopawiro J. Meeting of the minds: traditional herbal medicine in multiethnic Suriname (chapter 6). In: El-Shemy H, editor. Aromatic and medicinal plants—Back to nature. Rijeka: InTech; 2017. pp. 111-132. DOI: 10.5772/66509
  144. 144. Herndon CN, Uiterloo M, Uremaru A, Plotkin MJ, Emanuels-Smith G, Jitan J. Disease concepts and treatment by tribal healers of an Amazonian forest culture. Journal of Ethnobiology and Ethnomedicine. 2009;5:27. DOI: 10.1186/1746-4269-5-27
  145. 145. Van Andel TR, de Korte S, Koopmans D, Behari-Ramdas J, Ruysschaert S. ‘Wasi ondrosei’; het gebruik van vaginale stoombaden in Suriname. OSO, tijdschrift voor Surinamistiek. 2008;27:52-71
  146. 146. Liu X, Ma Z, Zhang J, Yang L. Antifungal compounds against Candida infections from traditional Chinese medicine. BioMed Research International. 2017;2017:4614183. DOI: 10.1155/2017/4614183
  147. 147. Mahmoud DA, Hassanein NM, Youssef KA, Abou Zeid MA. Antifungal activity of different neem leaf extracts and the nimonol against some important human pathogens. Brazilian Journal of Microbiology. 2011;42:1007-1016. DOI: 10.1590/S1517-838220110003000021
  148. 148. Tjong AG. Het gebruik van medicinale planten door de Javaanse bevolkingsgroep in Suriname (The use of medicinal plants by the Javanese in Suriname). Paramaribo, Suriname: Instituut voor de Opleiding van Leraren; 1989
  149. 149. Stephen HJM. Geneeskruiden van Suriname: hun toepassing in de volksgeneeskunde en in de magie (Herbal medicines from suriname: their applications in folk medicine and wizardry). Amsterdam, The Netherlands: De Driehoek; 1979
  150. 150. May AF, Sranan oso dresi. Surinaams kruidenboek [Surinamese Folk Medicine. A Collection of Surinamese Medicinal Herbs]. Paramaribo, Suriname: De Walburg Pers; 1982
  151. 151. Heyde H. Surinaamse medicijnplanten (Surinamese Medicinal Plants). 2nd ed. Paramaribo, Suriname: Westfort; 1987
  152. 152. Sedoc NO. Afrosurinaamse natuurgeneeswijzen: Bevattende meer dan tweehonderd meest gebruikelijke geneeskrachtige kruiden (AfroSurinamese Natural Remedies: Over Two Hundred Commonly Used Medicinal Herbs). Paramaribo, Suriname: Vaco Press; 1992
  153. 153. Raghoenandan UPD. Etnobotanisch onderzoek bij de Hindoestaanse bevolkingsgroep in Suriname (An ethnobotanical investigation among Hindustanis in Suriname) [thesis]. Paramaribo, Suriname: Anton de Kom University of Suriname; 1994
  154. 154. DeFilipps RA, Maina SL, Crepin J. Medicinal Plants of the Guianas (Guyana, Surinam, French Guiana). Washington, DC, USA: Smithsonian Institution; 2004
  155. 155. Van Andel TR, Ruysschaert S. Medicinale en rituele planten van Suriname (Medicinal and Ritual Plants of Suriname). Amsterdam, The Netherlands: KIT Publishers; 2011
  156. 156. Singh B, Namrata KL, Dwivedi SC. Antibacterial and antifungal activity of Colocasia esculenta aqueous extract: An edible plant. Journal of Pharmacy Research. 2011;4:1459-1460
  157. 157. Yang AH, Yeh KW. Molecular cloning, recombinant gene expression and antifungal activity of cystain from taro. Planta Medica. 2005;221:493-501. DOI: 10.1007/s00425-004-1462-8
  158. 158. Nazeer BS. Antimicrobial activity of Euphorbia hirta L. Paripex—Indian Journal of Research. 2017;6:1-2
  159. 159. Singh P, Sinha KK. Inhibition of aflatoxin production on some agricultural commodities through aqueous plant extracts. Journal of the Indian Botanical Society. 1986;65:30-32
  160. 160. Mohamed S, Saka S, El-Sharkawy SH, Ali AM, Muid S. Antimycotic screening of 58 Malaysian plants against plant pathogens. Pesticide Science. 1996;47:259-264. DOI: 10.1002/(SICI)1096-9063(199607)47:3<259::AID-PS413>3.0.CO;2-N
  161. 161. Rao KVB, Karthik L, Elumalai EK, Srinivasan K, Kumar G. Antibacterial and antifungal activity of Euphorbia hirta L. leaves: A comparative study. Journal of Pharmacy Research. 2010;3:548-549
  162. 162. Masood A, Rajan KS. The effect of aqueous plant extracts on growth and aflatoxin production by Aspergillus flavus. Letters in Applied Microbiology. 1991;13:32-34. DOI: 10.1111/j.1472-765X.1991.tb00562.x
  163. 163. Suresh K, Deepa P, Harisaranraj R, Vaira AV. Antimicrobial and phytochemical investigation of the leaves of Carica papaya L., Cynodon dactylon (L.) Pers., Euphorbia hirta L., Melia azedarach L. and Psidium guajava L. Ethnobotanical Leaflets. 2008;12:1184-1189
  164. 164. Vaidya M. Antimicrobial and antifungal activity of the plant extract of Euphorbia hirta L. World Journal of Pharmaceutical Research. 2017;6:2043-2047
  165. 165. Adjeroh LA, Nwachukwu MO, Abara PN, Nnokwe JC, Azorji JN, Osinomumu IO. Phytochemical screening and antibacteria/antifungi activities of root and shoot extracts of Euphorbia hirta (asthma weed). International Journal of Tropical Disease and Health. 2020;41:1-10. DOI: 10.9734/ijtdh/2020/v41i630281
  166. 166. Rajeh MA, Zuraini Z, Sasidharan S, Latha LY, Amutha S. Assessment of Euphorbia hirta L. leaf, flower, stem and root extracts for their antibacterial and antifungal activity and brine shrimp lethality. Molecules. 2010;15:6008-6018. DOI: 10.3390/molecules15096008
  167. 167. Sanguri S, Kapil S, Gopinathan P, Pandey FK, Bhatnagar T. Comparative screening of antibacterial and antifungal activities of some weeds and medicinal plants leaf extracts: an in vitro study. Elixir International Journal (Applied Botany). 2012;47:8903-8905
  168. 168. Kumar A, Shukla R, Singh P, Dubey NK. Chemical composition, antifungal and anti-aflatoxigenic activities of Ocimum sanctum L. essential oil and its safety assessment as plant based antimicrobial. Food and Chemical Toxicology. 2010;48:539-543. DOI: 10.1016/j.fct.2009.11.028
  169. 169. Sermakkani M, Thangapandian V. Studies on preliminary phytochemical constituents and antimicrobial activity of Ocimum tenuiflorum L. leaves. International Journal of Institutional Pharmacy and Life Sciences. 2011;1:1-12
  170. 170. Khan A, Ahmad A, Manzoor N, Khan LA. Antifungal activities of Ocimum sanctum essential oil and its lead molecules. Natural Product Communications. 2010;5:345-349
  171. 171. Balakumar S, Rajan S, Thirunalasundari T, Jeeva S. Antifungal activity of Ocimum sanctum Linn. (Lamiaceae) on clinically isolated dermatophytic fungi. Asian Pacific Journal of Tropical Medicine. 2011;4:654-657. DOI: 10.1016/S1995-7645(11)60166-1
  172. 172. Leite JJ, Brito EH, Cordeiro RA, Brilhante RS, Sidrim JJ, Bertini LM, et al. Chemical composition, toxicity and larvicidal and antifungal activities of Persea americana (avocado) seed extracts. Revista da Sociedade Brasileira de Medicina Tropical. 2009;42:110-113. DOI: 10.1590/s0037-86822009000200003
  173. 173. Deuschle VCKN, Cruz RD, Flores VC, Denardi LB, Deuschle RAN, Rossi GG, et al. Persea americana: Phenolic profile, antioxidant potential, antimicrobial activity and in silico prediction of pharmacokinetic and toxicological properties. Indian Journal of Pharmaceutical Sciences. 2019;81:766-775. DOI: 10.36468/pharmaceutical-sciences
  174. 174. Martins N, Ferreira IC, Barros L, Silva S, Henriques M. Candidiasis: Predisposing factors, prevention, diagnosis and alternative treatment. Mycopathologia. 2014;177:223-240. DOI: 10.1007/s11046-014-9749-1
  175. 175. Falodun A, Imieje V, Erharuyi O, Ahomafor J, Jacob MR, Khan SI, et al. Evaluation of three medicinal plant extracts against Plasmodium falciparum and selected microganisms. African Journal of Traditional, Complementary, and Alternative Medicines. 2014;11:142-146. DOI: 10.4314/ajtcam.v11i4.22
  176. 176. Manfredi R, Calza L, Chiodo F. AIDS-associated Cryptococcus infection before and after the highly active antiretroviral therapy era: Emerging management problems. International Journal of Antimicrobial Agents. 2003;22:449-452. DOI: 10.1016/s0924-8579(03)00113-4
  177. 177. Bond R, Saijonmaa-Koulumies LEM, Lloyd DH. Population sizes and frequency of Malassezia pachydermatis at skin and mucosal sites on healthy dogs. Journal of Small Animal Practice. 1995;36:147-150. DOI: 10.1111/j.1748-5827.1995.tb02865.x
  178. 178. Thomas DS, Davenport RR. Zygosaccharomyces bailii—A profile of characteristics and spoilage activities. Food Microbiology. 1985;2:157-169. DOI: 10.1016/s0740-0020(85)80008-3
  179. 179. Akalazu JN, Uchegbu RI. Biochemical composition and antimicrobial activities of seed extracts of avocado (Persea americana). The FASEB Journal (Supplement: Experimental Biology 2020 Meeting Abstracts). 2020;34:1-1. DOI: 10.1096/fasebj.2020.34.s1.02097
  180. 180. Ajayi OE, Awala SI, Olalekan OT, Alabi OA. Evaluation of antimicrobial potency and phytochemical screening of Persea americana leaf extracts against selected bacterial and fungal isolates of clinical importance. Microbiology Research Journal International. 2017;20:1-11. DOI: 10.9734/MRJI/2017/24508
  181. 181. Domergue F, Helms GL, Prusky D, Browse J. Antifungal compounds from idioblast cells isolated from avocado fruits. Phytochemistry. 2000;54:183-189. DOI: 10.1016/s0031-9422(00)00055-8
  182. 182. Prusky D, Keen NT, Sims JJ, Midland SL. Possible involvement of an antifungal diene in the latency of Colletotrichum gloeosporioides on unripe avocado fruits. Phytopathology. 1982;72:1578-1582
  183. 183. Prusky D, Kobiler I, Fishman Y, Sims J, Midland S, Keen N. Identification of an antifungal compound in unripe avocado fruits and its possible involvement in the quiescent infections of Colletotrichum gloeosporioides. Journal of Phytopathology. 1991;132:319-327. DOI: 10.1111/j.1439-0434.1991.tb00127.x
  184. 184. Reddy MK, Gupta SK, Jacob MR, I Khan SI, Ferreira D. Antioxidant, antimalarial and antimicrobial activities of tannin-rich fractions, ellagitannins and phenolic acids from Punica granatum L. Planta Medica. 2007;73:461-467. DOI: 10.1055/s-2007-967167
  185. 185. Endo EH, Cortez DA, Ueda-Nakamura T, Nakamura CV, Dias Filho BP. Potent antifungal activity of extracts and pure compound isolated from pomegranate peels and synergism with fluconazole against Candida albicans. Research in Microbiology. 2010;161:534-540. DOI: 10.1016/j.resmic.2010.05.002
  186. 186. De Souza Vasconcelos LC, Sampaio MCC, Sampaio FC, Higino JS. Use of Punica granatum as an antifungal agent against candidosis associated with denture stomatitis. Mycoses. 2003;46:192-196. DOI: 10.1046/j.1439-0507.2003.00884.x
  187. 187. Dahham SS, Ali MN, Tabassum H, Khan M. Studies on antibacterial and antifungal activity of pomegranate (Punica granatum L.). American-Eurasian Journal of Agricultural and Environmental Sciences. 2010;9:273-281
  188. 188. Dutta BK, Rahman I, Das TK. Antifungal activity of Indian plant extracts. Mycoses. 1998;41:535-606. DOI: 10.1111/j.1439-0507.1998.tb00718.x
  189. 189. Wafa BA, Makni M, Ammar S, Khannous L, Hassana AB, Bouaziz M, et al. Antimicrobial effect of the Tunisian Nana variety Punica granatum L. extracts against Salmonella enterica (serovars Kentucky and Enteritidis) isolated from chicken meat and phenolic composition of its peel extract. International Journal of Food Microbiology. 2017;241:123-131. DOI: 10.1016/j.ijfoodmicro.2016.10.007
  190. 190. Tayel AA, El-Baz AF, Salem MF, El-Hadary MH. Potential applications of pomegranate peel extracts for the control of citrus green mould. Journal of Plant Diseases and Protection. 2009;116:252-256
  191. 191. Shuhua Q, Hongyun J, Yanning Z, Weizhi H. Inhibitory effects of Punica granatum peel extracts on Botrytis cinerea. Journal of Plant Diseases and Protection. 2010;36:148-150
  192. 192. Romeo FV, Ballistreri G, Fabroni S, Pangallo S, Nicosia MG, Schena L, et al. Chemical characterization of different sumac and pomegranate extracts effective against Botrytis cinerea rots. Molecules. 2015;20:11941-11958. DOI: 10.3390/molecules200711941
  193. 193. Elsherbinya EA, Aminb BH, Baka ZA. Efficiency of pomegranate (Punica granatum L.) peels extract as a high potential natural tool towards Fusarium dry rot on potato tubers. Postharvest Biology and Technology. 2016;111:256-263
  194. 194. Lopes NP, Kato MJ, Yoshida M. Antifungal constituents from roots of Virola surinamensis. Phytochemistry. 1999;51:29-33
  195. 195. Costa ES, Hiruma-Lima CA, Lima EO, Sucupira GC, Bertolin AO, Lolis SF, et al. Antimicrobial activity of some medicinal plants of the cerrado, Brazil. Phytotherapy Research. 2008;22:705-707. DOI: 10.1002/ptr.2397
  196. 196. Khalandi H, Masoori L, Farahyar S, Delbandi AA, Raiesi O, Farzanegan A, et al. Antifungal activity of capric acid, nystatin, and fluconazole and their in vitro interactions against candida isolates from neonatal oral thrush. Assay and Drug Development Technologies. 2020;18:195-201. DOI: 10.1089/adt.2020.971
  197. 197. Chadeganipour M, Haims A. Antifungal activities of pelargonic and capric acid on Microsporum gypseum. Mycoses. 2001;44:109-112. DOI: 10.1046/j.1439-0507.2001.00609.x
  198. 198. Das M, Goswami S. Antifungal and antibacterial property of guava (Psidium guajava) leaf extract: Role of phytochemicals. International Journal of Health Sciences and Research. 2019;9:39-45
  199. 199. Nair R, Chanda S. In vitro antimicrobial activity of Psidium guajava L. leaf extracts against clinically important pathogenic microbial strains. Brazilian Journal of Microbiology. 2007;38:452-458
  200. 200. Metwally AM, Omar AA, Harraz FM, El Sohafy SM. Phytochemical investigation and antimicrobial activity of Psidium guajava L. leaves. Pharmacognosy Magazine. 2010;6:212-218. DOI: 10.4103/0973-1296.66939
  201. 201. Padrón-Márquez B, Viveros-Valdez E, Oranday-Cárdenas A, Carranza-Rosales P. Antifungal activity of Psidium guajava organic extracts against dermatophytic fungi. Journal of Medicinal Plants Research. 2012;6:5435-5438. DOI: 10.5897/JMPR12.240
  202. 202. Saleh B, Al-Halab L, Al-Mariri A. In vitro leaves and twigs antimicrobial properties of Psidium guajava L. (Myrtaceae). Herba Polonica. 2015;61:93-104. DOI: 10.1515/hepo-2015-0025
  203. 203. Morais-Braga MFB, Sales DL, Carneiro JNP, Machado AJT, Dos Santos ATL, de Freitas MA, et al. Psidium guajava L. and Psidium brownianum Mart ex DC.: Chemical composition and anti-Candida effect in association with fluconazole. Microbial Pathogenesis. 2016;95:200-207. DOI: 10.1016/j.micpath.2016.04.013
  204. 204. Bezerra CF, Rocha JE, Nascimento Silva MKD, de Freitas TS, de Sousa AK, Dos Santos ATL, et al. Analysis by UPLC-MS-QTOF and antifungal activity of guava (Psidium guajava L.). Food and Chemical Toxicology. 2018;119:122-132. DOI: 10.1016/j.fct.2018.05.021
  205. 205. Dutta BK, Das TK. In vitro study on antifungal property of common fruit plants. Biomedicine. 2000;20:187-189
  206. 206. Sato J, Goto K, Nanjo F, Hawai S, Murata K. Antifungal activity of plant extracts against Arthrinium sacchari and Chaetomium funicola. Journal of Biochemical Engineering and Sciences. 2000;90:442-446
  207. 207. Gharby S, Harhar H, Guillaume D, Roudani A, Boulbaroud S, Ibrahimi M, et al. Chemical investigation of Nigella sativa L. seed oil. Journal of the Saudi Society of Agricultural Sciences. 2015;14:172-177. DOI: 10.1016/j.jssas.2013.12.001
  208. 208. Al-Ali A, Alkhawajah AA, Randhawa MA, Shaikh NA. Oral and intraperitoneal LD50 of thymoquinone, an active principle of Nigella sativa, in mice and rats. Journal of Ayub Medical College, Abbottabad. 2008;20:25-27
  209. 209. Chehl N, Chipitsyna G, Gong Q, Yeo CJ, Arafat HA. Anti-inflammatory effects of the Nigella sativa seed extract, thymoquinone, in pancreatic cancer cells. HPB: The Official Journal of the International Hepato-Pancreato-Biliary Association (Oxford). 2009;11:373-381. DOI: 10.1111/j.1477-2574.2009.00059.x
  210. 210. Noor NA, Fahmy HM, Mohammed FF, Elsayed AA, Radwan NM. Nigella sativa ameliorates inflammation and demyelination in the experimental autoimmune encephalomyelitis-induced Wistar rats. International Journal of Clinical and Experimental Pathology. 2015;8:6269-6286
  211. 211. Hadi V, Kheirouri S, Alizadeh M, Khabbazi A, Hosseini H. Effects of Nigella sativa oil extract on inflammatory cytokine response and oxidative stress status in patients with rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Avicenna Journal of Phytomedicine. 2016;6:34-43
  212. 212. Al-Ghasham A, Ata HS, El-Deep S, Meki AR, Shehada S. Study of protective effect of date and Nigella sativa on aflatoxin B1 toxicity. International Journal of Health Sciences (Qassim). 2008;2:26-44
  213. 213. Daryabeygi-Khotbehsara R, Golzarand M, Ghaffari MP, Djafarian K. Nigella sativa improves glucose homeostasis and serum lipids in type 2 diabetes: A systematic review and meta-analysis. Complementary Therapies in Medicine. 2017;35:6-13. DOI: 10.1016/j.ctim.2017.08.016
  214. 214. Sahebkar A, Beccuti G, Simental-Mendía LE, Nobili V, Bo S. Nigella sativa (black seed) effects on plasma lipid concentrations in humans: A systematic review and meta-analysis of randomized placebo-controlled trials. Pharmacological Research. 2016;106:37-50. DOI: 10.1016/j.phrs.2016.02.008
  215. 215. Rafati S, Niakan M, Naseri M. Antimicrobial effect of Nigella sativa seed extract against staphylococcal skin infection. Medical Journal of the Islamic Republic of Iran. 2014;28:42. eCollection 2014
  216. 216. Morsi NM. Antimicrobial effect of crude extracts of Nigella sativa on multiple antibiotics-resistant bacteria. Acta Microbiologica Polonica. 2000;49:63-74
  217. 217. Aljabre SH, Randhawa MA, Akhtar N, Alakloby OM, Alqurashi AM, Aldossary A. Antidermatophyte activity of ether extract of Nigella sativa and its active principle, thymoquinone. Journal of Ethnopharmacology. 2005;101:116-119. DOI: 10.1016/j.jep.2005.04.002
  218. 218. Sheik Noor MM, Jaikumar K, Babu A, Anand D, Saravanan P. A study on the in vitro antifungal activity of Nigella sativa (Linn.) seed extract and its phytochemical screening using GC-MS analysis. World Journal of Pharmacy and Pharmaceutical Sciences. 2015;4:1003-1011
  219. 219. Pattnaik S, Subramanyam VR, Kole C. Antibacterial and antifungal activity of ten essential oils in vitro. Microbios. 1996;86:237-246
  220. 220. Ibrahim NA, El-Sakhawy FS, Mohammed MMD, Farid MA, Abdel Wahed NAM, Deabes DAH. Chemical composition, antimicrobial and antifungal activities of essential oils of the leaves of Aegle marmelos (L.) Correa growing in Egypt. Journal of Applied Pharmaceutical Science. 2015;5:1-5. DOI: 10.7324/JAPS.2015.50201
  221. 221. Maity P, Hansda D, Bandyopadhyay U, Mishra DK. Biological activities of crude extracts and chemical constituents of bael, Aegle marmelos (L.) Corr. Indian Journal of Experimental Biology. 2009;47:849-861
  222. 222. Bansal Y, Bansal G. Analytical methods for standardization of Aegle marmelos: A review. Journal of Pharmaceutical Education and Research. 2011;2:37-44
  223. 223. Jain NK. Antifungal activity of essential oil of Aegle marmelos Correa (Rutaceae). Indian Drugs and Pharmaceutical Industry. 1977;12:55
  224. 224. Dubey NK, Mishra AK. Evaluation of some essential oil against dermatophytes. Indian Drugs. 1990;27:529-531
  225. 225. Mishra DN, Dixit V, Mishra AK. Mycotoxic evaluation of some higher plants against ringworm causing fungi. Indian Drugs. 1991;28:300-303
  226. 226. Yadav P, Dubey NK. Screening of some essential oils against ringworm fungi. Indian Journal of Pharmaceutical Sciences. 1994;56:227-230
  227. 227. Rana BK, Singh UP, Taneja V. Antifungal activity and kinetics of inhibition by essential oil isolated from leaves of Aegle marmelos. Journal of Ethnopharmacology. 1997;57:29-34. DOI: 10.1016/S0378-8741(97)00044-5
  228. 228. More YD, Gade RM, Shitole AV. In vitro antifungal activity of Aegle marmelos, Syzygium cumini and Pongamia pinnata extracts against Fusarium oxysporum f. sp. cicero. Indian Journal of Pharmaceutical Sciences. 2017;79:457-462. DOI: 10.4172/pharmaceutical-sciences.1000249
  229. 229. Gómez Caravaca AM, Verardo V, Toselli M, Segura Carretero A, Fernández Gutiérrez A, Caboni MF. Determination of the major phenolic compounds in pomegranate juices by HPLC−DAD−ESI-MS. Journal of Agricultural and Food Chemistry. 2013;61:5328-5337. DOI: 10.1021/jf400684n
  230. 230. Bhowmik D, Gopinath H, Kumar BP, Duraivel S, Aravind G, Kumar KPS. Medicinal uses of Punica granatum and its health benefits. Journal of Pharmacognosy and Phytochemistry. 2013;1:28-35
  231. 231. Shaygannia E, Bahmani M, Zamanzad B, Rafieian-Kopaei M. A review study on Punica granatum L. Journal of Evidence-Based Complementary and Alternative Medicine. 2016;21:221-227. DOI: 10.1177/2156587215598039
  232. 232. Yones DA, Badary DM, Sayed HM, Bayoumi SA, Khalifa AA, El-Moghazy AM. Comparative evaluation of anthelmintic activity of edible and ornamental pomegranate ethanolic extracts against Schistosoma mansoni. BioMed Research International. 2016;2016:2872708. DOI: 10.1155/2016/2872708
  233. 233. Amelia M, Jasaputra D, Tjokropranoto R. Effects of pomegranate peel (Punica granatum L.) extract as an anthelmintic. Journal of Medicine and Health. 2017;1. DOI: 10.28932/jmh.v1i5.537
  234. 234. Bradbury JH, Nixon RW. The acridity of raphides from the edible aroids. Journal of the Science of Food and Agriculture. 1998;76:608-616. DOI: 10.1002/(SICI)1097-0010(199804)76:4<608::AID-JSFA996>3.0.CO;2-2
  235. 235. Reyad-ul-Ferdous M, Arman MSI, Tanvir MMI, Sumi S, Siddique KMMR, Billah MM, et al. Biologically potential for pharmacologicals and phytochemicals of medicinal plants of Colocasia esculenta: A comprehensive review. American Journal of Clinical and Experimental Medicine. 2015;3:7-11. DOI: 10.11648/j.ajcem.s.2015030501.12
  236. 236. Pawar HA, Choudhary PD, Kamat SR. An overview of traditionally used herb, Colocasia esculenta, as a phytomedicine. Medicinal and Aromatic Plants. 2018;7:317. DOI: 10.4172/2167-0412.1000317
  237. 237. Plowman T. Folk uses of New World aroids. Economic Botany. 1969;23:97-122
  238. 238. Kumawat NS, Chaudhari SP, Wani NS, Deshmukh TA, Patil VR. Antidiabetic activity of ethanol extract of Colocasia esculenta leaves in alloxan-induced diabetic rats. International Journal of PharmTech Research. 2010;2:1246-1249
  239. 239. Prema P, Kurup PA. Effect of feeding cooked whole tubers on lipid metabolism in rats fed cholesterol free and cholesterol containing diet. Indian Journal of Experimental Biology. 1979;17:1341-1345
  240. 240. Biren NS, Nayak BS, Bhatt SP, Jalalpure SS, Seth AK. The anti-inflammatory activity of Colocasia esculenta. Saudi Pharmaceutical Journal. 2007;15:228-232
  241. 241. Tuse TA, Harle UN, Bore VV. Hepatoprotective activity of Colocasia antiquorum against experimentally induced liver injury in rat. Malaysian Journal of Pharmaceutical Sciences. 2009;2:99-112
  242. 242. Bhagyashree RP, Hussein MA. Antihepatotoxic activity of Colocasia esculenta leaf juice. International Journal of Advanced Biotechnology and Research. 2011;2:296-304
  243. 243. Kubde MS, Khadabadi SS, Farooqui IA, Deore SL. In vitro anthelmintic activity of Colocasia esculenta. Archives of Applied Science Research. 2010;2:82-85
  244. 244. Ravikumar S, Gracelin N, Anitha A, Selvan PG, Kalaiarasi A. In vitro antibacterial activity of coastal medicinal plants against isolated bacterial fish pathogens. International Journal of Pharmaceutical Research and Development. 2011;3:109-116
  245. 245. Sakano Y, Mutsuga M, Tanaka R, Suganuma H, Inakuma T, Toyoda M, et al. Inhibition of human lanosterol synthase by the constituents of Colocasia esculenta (taro). Biological and Pharmaceutical Bulletin. 2005;28:299-304. DOI: 10.1248/bpb.28.299
  246. 246. Dupont S, Lemetais G, Ferreira T, Cayot P, Gervais P, Beney L. Ergosterol biosynthesis: A fungal pathway for life on land? Evolution. 2012;66:2961-2968. DOI: 10.1111/j.1558-5646.2012.01667.x
  247. 247. Pernas M, Lopez-Solanilla E, Sanchez-Monge R, Salcedo G, Rodriguez-Palenzuela P. Antifungal activity of a plant cystatin. Molecular Plant-Microbe Interactions. 1999;12:624-627. DOI: 10.1094/MPMI.1999.12.7.624
  248. 248. Pintus F, Medda R, Rinaldi AC, Spanò D, FlorisG. Euphorbia latex biochemistry: Complex interactions in a complex environment. Plant Biosystems. 2010;144:381-391. DOI: 10.1080/11263500903396016
  249. 249. Hussain M, Farooq U, Rashid M, Bakhsh H, Majeed A, Khan IA, et al. Antimicrobial activity of fresh latex, juice and extract of Euphorbia hirta and Euphorbia thymifolia—An in vitro comparative study. International Journal of Pharma Sciences. 2014;4:546-553
  250. 250. Ekpo OE, Pretorius E. Asthma, Euphorbia hirta and its anti-inflammatory properties. South African Journal of Science. 2007;103:201-203
  251. 251. Kumar S, Malhotra R, Kumar D. Euphorbia hirta: Its chemistry, traditional and medicinal uses, and pharmacological activities. Pharmacognosy Reviews. 2010;4:58-61. DOI: 10.4103/0973-7847.65327
  252. 252. Asha S, Deevika B, Sadiq AM. Euphorbia hirta Linn—A review on traditional uses, phytochemistry and pharmacology. World Journal of Pharmaceutical Research. 2014;3:180-205
  253. 253. Galvez J, Zarzuelo A, Crespo ME, Lorente MD, Ocete MA, Jimenez J. Antidiarrhoeic activity of Euphorbia hirta extract and isolation of an active flavanoid constituent. Planta Medica. 1993;59:333-336. DOI: 10.1055/s-2006-959694
  254. 254. Ali MZ, Mehmood MH, Saleem M, Gilani A-H. The use of Euphorbia hirta L. (Euphorbiaceae) in diarrhea and constipation involves calcium antagonism and cholinergic mechanisms. BMC Complementary Medicine and Therapies. 2020;20:14. DOI: 10.1186/s12906-019-2793-0
  255. 255. Williams LAD, Williams MG, Sajabi A, Barton EN, Fleischhacker R. Angiotensin converting enzyme inhibiting and anti-dipsogenic activities of Euphorbia hirta. extracts. Phytotherapy Research. 1997;11:401-402
  256. 256. Devi S, Kumar M. In vivo antidiabetic activity of methanolic extract of Euphorbia hirta L. International Journal of Diabetes and Endocrinology. 2017;23:36-39. DOI: 10.11648/j.ijde.20170203.11
  257. 257. Johnson PB, Abdurahman EM, Tiam EA, Abdu-Aguye I, Hussaini IM. Euphorbia hirta leaf extracts increase urine output and electrolytes in rats. Journal of Ethnopharmacology. 1999;65:63-69. DOI: 10.1016/s0378-8741(98)00143-3
  258. 258. Lanhers MC, Fleurentin J, Cabalion P. Behavioral effects of Euphorbia hirta L.: Sedative and anxiolytic properties. Journal of Ethnopharmacology. 1990;29:189-198. DOI: 10.1016/0378-8741(90)90055-x
  259. 259. Lanhers MC, Fleurentin J, Dorfman P, Mortier F, Pelt JM. Analgesic, antipyretic and antiinflammatory properties of Euphorbia hirta. Planta Medica. 1991;57:225-231. DOI: 10.1055/s-2006-960079
  260. 260. Singh GD, Kaiser P, Youssouf MS, Singh S, Khajuria A, Koul A, et al. Inhibition of early and late phase allergic reactions by Euphorbia hirta L. Phytotheraoy Research. 2006;20:316-321. DOI: doi.org/10.1002/ptr.1844
  261. 261. Tona L, Ngimbi NP, Tsakala M, Mesia K, Cimanga K, Apers S, et al. Antimalarial activity of 20 crude extracts from nine African medicinal plants in Kinshasa, Congo. Journal of Ethnopharmacology. 1999;68:193-203. DOI: 10.1016/s0378-8741(99)00090-2
  262. 262. Tona L, Cimanga RK, Mesia GK, Musuamba CT, de Bruyne T, Apers S, et al. In vitro antiplasmodial activity of extracts and fractions from seven medicinal plants used in Democratic Republic of Congo. Journal of Ethnopharmacology. 2004;93:27-32. DOI: 10.1016/S1995-7645(11)60052-7
  263. 263. Koli MC, Choudhary R, Kumar S. An isoflavone glycoside from the stem of Euphorbia hirta Linn as antimalarial compound. Asian Journal of Chemistry. 2002;14:1673-1677
  264. 264. Liu Y, Murakami N, Ji H, Abreu P, Zhang S. Antimalarial flavonol glycosides from Euphorbia hirta. Pharmaceutical Biology. 2007;45:278-281. DOI: 10.1080/13880200701214748
  265. 265. Wang YC, Huang TL. Screening of anti-Helicobacter pylori herbs deriving from Taiwanese folk medicinal plants. FEMS Immunology and Medial Microbiology. 2005;43:295-300. DOI: 10.1016/j.femsim.2004.09.008
  266. 266. Jackson C, Agboke A, Nwoke V. In vitro evaluation of antimicrobial activity of combinations of nystatin and Euphorbia hirta leaf extract against Candida albicans by the checkerboard method. Journal of Medicinal Plants Research. 2009;3:666-669
  267. 267. Mondal S, Mirdha BR, Mahapatra SC. The science behind sacredness of Tulsi (Ocimum sanctum Linn.). Indian Journal of Physiology and Pharmacology. 2009;53:291-306
  268. 268. Biswas NP, Biswas AK. Evaluation of some leaf dusts as grain protectant against rice weevil Sitophilus oryzae (Linn.). Environment and Ecology. 2005;23:485-488
  269. 269. Rahman S, Islam R, Kamruzzaman M, Alam K, Jamal ARM. Ocimum sanctum L.: A review of phytochemical and pharmacological profile. American Journal of Drug Discovery and Development. 2011;1:15. DOl: 10.3923/ajdd.2011
  270. 270. Ravi P, Elumalai A, Eswaraiah MC, Kasarla R. A review on krishna tulsi, Ocimum tenuiflorum Linn. International Journal of Research in Ayurveda and Pharmacy. 2012;3:291-293
  271. 271. Padalia RC, Verma RS. Comparative volatile oil composition of four Ocimum species from northern India. Natural Product Research. 2011;25:569-575. DOI: 10.1080/14786419.2010.482936
  272. 272. Singh D, Chaudhuri PK. A review on phytochemical and pharmacological properties of holy basil (Ocimum sanctum L.). Industrial Crops and Products. 2018;118:367-382. DOI: 10.1016/j.indcrop.2018.03.048
  273. 273. Sridevi G, Gopkumar P, Ashok S, Shastry CS. Pharmacological basis for antianaphylactic, antihistaminic and mast cell stabilization activity of Ocimum sanctum. Internet Journal of Pharmacology. 2008;7. DOI: 10.5580/bc4
  274. 274. Singh S, Majumdar DK, Rehan HMS. Evaluation of anti-inflammatory potential of Ocimum sanctum (holy basil) and its possible mechanism of action. Journal of Ethnopharmacology. 1996;54:19-26. DOI: 10.1016/0378-8741(96)83992-4
  275. 275. Singh S, Majumdar DK. Evaluation of antiinflammatory activity of fatty acids of Ocimum sanctum fixed oil. Indian Journal of Experimental Biology. 1997;35:380-383
  276. 276. Singh S. Comparative evaluation of antiinflammatory potential of fixed oil of different species of Ocimum and its possible mechanism of action. Indian Journal of Experimental Biology. 1998;36:1028-1031
  277. 277. Singh S, Majumdar DK. Analgesic activity of Ocimum sanctum and its possible mechanism of action. International Journal of Pharmacognosy. 1995;33:188-192. DOI: 10.3109/13880209509065361
  278. 278. Rao SA, Vijay Y, Deepthi T, Lakshmi CS, Rani V. Antidiabetic effect of ethanolic extract of leaves of Ocimum sanctum in alloxan-induced diabetes in rats. International Journal of Basic and Clinical Pharmacology. 2013;2:613-616
  279. 279. Hannan JMA, Marenah L, Ali L, Rokeya B, Flatt PR, Abdel YHA. Ocimum sanctum leaf extracts stimulate insulin secretion from perfused pancreas, isolated islets and clonal pancreatic beta-cells. Journal of Endocrinology. 2006;189:127-136. DOI: 10.1677/joe.1.06615
  280. 280. Kundu PK, Chatterjee PS. Meta-analysis of Diabecon tablets: efficacy and safety. Outcomes from 15 clinical trials in diabetes mellitus. Indian Journal of Clinical Practice. 2010;20:653-659
  281. 281. Jaggi RK, Madaan R, Singh B. Anticonvulsant potential of holy basil, Ocimum sanctum Linn. and its cultures. Indian Journal of Experimental Biology. 2003;41:1329-1333
  282. 282. Ravindran R, Devi RS, Samson J, Senthilvelan M. Noise stress induced brain neurotransmitter changes and the effect of Ocimum sanctum (Linn) treatment in albino rats. Journal of Pharmacological Sciences. 2005;98:354-360. DOI: 10.1254/jphs.fp0050127
  283. 283. Sembulingam K, Sembulingam P, Namasivayam A. Effect of Ocimum sanctum Linn on the changes in central cholinergic system induced by acute noise stress. Journal of Ethnopharmacology. 2005;96:477-482. DOI: 10.1016/j.jep.2004.09.047
  284. 284. Asha MK, Prashanth D, Murali B, Padmaja R, Amit A. Anthelmintic activity of essential oil of Ocimum sanctum and eugenol. Fitoterapia. 2001;72:669-670. DOI: 10.1016/s0367-326x(01)00270-2
  285. 285. Seleman NS, Amri E. Antibacterial activity of aqueous, ethanol and acetone extracts of Ocimum sanctum Linn. American Journal of BioScience. 2015;3:256-261. DOI: 10.11648/j.ajbio.20150306.18
  286. 286. Rahman MS, Khan MMH, Jamal MAHM. Antibacterial evaluation and minimum inhibitory concentration analysis of Oxalis corniculata and Ocimum sanctum against bacterial pathogens. Biotechonology. 2010;9:533-536. DOI: 10.3923/biotech.2010.533.536
  287. 287. Mishra P, Mishra S. Study of antibacterial activity of Ocimum sanctum extract against gram positive and gram negative bacteria. American Journal of Food Technology. 2011;6:336-341. DOI: 10.3923/ajft.2011.336.341
  288. 288. Yamani HA, Pang EC, Mantri N, Deighton MA. Antimicrobial activity of tulsi (Ocimum tenuiflorum) essential oil and their major constituents against three species of bacteria. Frontiers in Microbiology. 2016;7:681. DOI: 10.3389/fmicb.2016.00681
  289. 289. Dreher ML, Davenport AJ. Hass avocado composition and potential health effects. Critical Reviews in Food Science and Nutrition. 2013;53:738-750. DOI: 10.1080/10408398.2011.556759
  290. 290. Flores M, Saravia C, Vergara CE, Avila F, Valdés H, Ortiz-Viedma J. Avocado oil: Characteristics, properties, and applications. Molecules. 2019;24:2172. DOI: 10.3390/molecules24112172
  291. 291. Eyres L. Frying oils: Selection, smoke points and potential deleterious effects for health. Food New Zealand. 2015;15:30-31
  292. 292. Stadler PI, van Rensburg B, Naudé TW. Suspected avocado (Persea americana) poisoning in goats. Journal of the South African Veterinary Association. 1991;62:186-188
  293. 293. Kulkarni P, Paul R, Ganesh N. In vitro evaluation of genotoxicity of avocado (Persea americana) fruit and leaf extracts in human peripheral lymphocytes. Journal of Environmental Science and Health, Part C. 2010;28:172-187. DOI: 10.1080/10590501.2010.504979
  294. 294. Yasir M, Das S, Kharya MD. The phytochemical and pharmacological profile of Persea americana Mill. Pharmacognosy Reviews. 2010;4:77-84. DOI: 10.4103/0973-7847.65332
  295. 295. Owolabi MA, Jaja SI, Coker HAB. Vasorelaxant action of aqueous extract of the leaves of Persea americana on isolated thoracic rat aorta. Fitoterapia. 2005;76:567-573. DOI: 10.1016/j.fitote.2005.04.020
  296. 296. Anaka ON, Ozolua RI, Okpo SO. Effect of the aqueous seed extract of Persea americana Mill (Lauraceae) on the blood pressure of Sprague-Dawley rats. African Journal of Pharmacy and Pharmacology. 2009;3:485-490
  297. 297. Imafidon KE, Amaechina FC. Effects of aqueous seed extract of Persea americana Mill. (avocado) on blood pressure and lipid profile in hypertensive rats. Advances in Biological Research. 2010;4:116-121
  298. 298. Antia BS, Okokon JE, Okon PA. Hypoglycemic activity of aqueous leaf extract of Persea americana Mill. Indian Journal of Pharmacology. 2005;37:325-326. DOI: 10.4103/0253-7613.16858
  299. 299. Brai BI, Odetola AA, Agomo PU. Hypoglycemic and hypocholesterolemic potential of Persea americana leaf extracts. Journal of Medicinal Food. 2007;10:356-360. DOI: 10.1089/jmf.2006.291
  300. 300. Pahua-Ramos ME, Ortiz-Moreno A, Chamorro-Cevallos G, Hernández-Navarro MD, Garduño-Siciliano L, Necoechea-Mondragón H, et al. Hypolipidemic effect of avocado (Persea americana Mill) seed in a hypercholesterolemic mouse model. Plant Foods for Human Nutrition. 2012;67:10-16. DOI: 10.1007/s11130-012-0280-6
  301. 301. Kawagishi H, Fukumoto Y, Hatakeyama M, He P, Arimoto H, Matsuzawa T, et al. Liver injury suppressing compounds from avocado (Persea americana). Journal of Agricultural and Food Chemistry. 2001;49:2215-2221. DOI: 10.1021/jf0015120
  302. 302. Ekor M, Adepoju GKA, Epoyun AA. Protective effect of the methanolic leaf extract of Persea americana (avocado) against paracetamol-induced acute hepatotoxicity in rats. International Journal of Pharmacology. 2006;2:416-420. DOI: 10.3923/ijp.2006.416.420
  303. 303. Adeyemi OO, Okpo SO, Ogunti OO. Analgesic and antiinflammatory effects of the aqueous extract of leaves of Persea americana Mill (Lauraceae). Fitoterapia. 2002;73:375-380. DOI: 10.1016/s0367-326x(02)00118-1
  304. 304. Ojewole JA, Amabeoku GJ. Anticonvulsant effect of Persea americana Mill (Lauraceae) (avocado) leaf aqueous extract in mice. Phytotherapy Research. 2006;20:696-700. DOI: 10.1002/ptr.1940
  305. 305. Jimenez-Arellanes A, Luna-Herrera J, Ruiz-Nicolas R, Cornejo-Garrido J, Tapia A, Yépez-Mulia L. Antiprotozoal and antimycobacterial activities of Persea americana seeds. BMC Complementary and Alternative Medicine. 2013;13:109. DOI: 10.1186/1472-6882-13-109
  306. 306. Abe F, Nagafuji S, Okawa M, Kinjo J, Akahane H, Ogura T, et al. Trypanocidal constituents in plants 5. Evaluation of some Mexican plants for their trypanocidal activity and active constituents in the seeds of Persea americana. Biological and Pharmaceutical Bulletin. 2005;28:1314-1317
  307. 307. Chia TWR, Dykes GA. Antimicrobial activity of crude epicarp and seed extracts from mature avocado fruit (Persea americana) of three cultivars. Pharmaceutical Biology. 2010;48:753-756. DOI: 10.3109/13880200903273922
  308. 308. Lu YC, Chang HS, Peng CF, Lin CH, Chen IS. Secondary metabolites from the unripe pulp of Persea americana and their antimycobacterial activities. Food Chemistry. 2012;135:2904-2909. DOI: 10.1016/j.foodchem.2012.07.073
  309. 309. Al-Megrin WA. In vivo study of pomegranate (Punica granatum) peel extract efficacy against Giardia lamblia in infected experimental mice. Asian Pacific Journal of Tropical Biomedicine. 2017;7:59-63. DOI: 10.1016/j.apjtb.2016.08.018
  310. 310. Al-Hemiri AA, Abed KM, Al-Shahwany AW. Extraction of pelletierine from Punica granatum L. by liquid membrane technique and modelling. Iraqi Journal of Chemical and Petroleum Engineering. 2012;13:1-9
  311. 311. Jafri MA, Aslam M, Javed K, Singh S. Effect of Punica granatum Linn. (flowers) on blood glucose level in normal and alloxan-induced diabetic rats. Journal of Ethnopharmacology. 2000;70:309-314. DOI: 10.1016/s0378-8741(99)00170-1
  312. 312. Bagri P, Ali M, Aeri V, Bhowmik M, Sultana S. Antidiabetic effect of Punica granatum flowers: Effect on hyperlipidemia, pancreatic cells lipid per oxidation and antioxidant enzymes in experimental diabetes. Food and Chemical Toxicology. 2009;47:50-54. DOI: 10.1016/j.fct.2008.09.058
  313. 313. Esmaillzadeh A, Tahbaz F, Gaieni I, Alavi-Majd H, Azadbakht L. Cholesterol-lowering effect of concentrated pomegranate juice consumption in type II diabetic patients with hyperlipidemia. International Journal Vitamin Nutritional Resources. 2006;76:147-151. DOI: 10.1024/0300-9831.76.3.147
  314. 314. Katz SR, Newman RA, Lansky EP. Punica granatum: Heuristic treatment for diabetes mellitus. Journal of Medicinal Food. 2007;10:213-217. DOI: 10.1089/jmf.2006.290
  315. 315. Sharma MK, Khare AK, Feroz H. Effect of neem oil on blood sugar levels of normal hyperglycemic and diabetic animals. Indian Medikcal Gazette. 1983;11:380-383
  316. 316. Danesi F, Ferguson LR. Could pomegranate juice help in the control of inflammatory diseases? Nutrients. 2017;9:958. DOI: 10.3390/nu9090958
  317. 317. Wang D, Özen C, Abu-Reidah IM, Chigurupati S, Patra JK, Horbanczuk JO, et al. Vasculoprotective effects of pomegranate (Punica granatum L.). Frontiers in Pharmacology. 2018;9:544. DOI: 10.3389/fphar.2018.00544
  318. 318. Ahmad I, Beg AZ. Antimicrobial and phytochemical studies on 45 Indian medicinal plants against multi-drug resistant human pathogens. Journal of Ethnopharmocology. 2001;74:113-123. DOI: 10.1016/s0378-8741(00)00335-4
  319. 319. Naz S, Siddiqi R, Ahmad S, Rasool SA, Sayeed SA. Antibacterial activity directed isolation of compounds from Punica granatum. Journal of Food Science. 2007;72:M341-M345. DOI: 10.1111/j.1750-3841.2007.00533.x
  320. 320. IUCN. The IUCN Red List of Threatened Species. Version 2020-2 [Internet]. 2021. Available from: https://www.iucnredlist.org. [Accessed: July 15, 2021]
  321. 321. Culp TW, Harlow RD, Litchfield C, Reiser R. Analysis of triglycerides by consecutive chromatographic techniques. II. Ucuhuba kernel fat. Journal of the American Oil Chemists' Society. 1965;42:974-978. DOI: 10.1007/BF02632458
  322. 322. Funasakia M, dos Santos BH, Fernandes VLA, Menezes IS. Amazon rainforest cosmetics: Chemical approach for quality control. Química Nova. 2016;39:194-209. DOI: 10.5935/0100-4042.20160008
  323. 323. Plotkin MJ. Ethnobotany and Conservation of the Tropical Forests with Special Reference to the Indians of Southern Suriname [PhD dissertation]. Boston, MA, USA: Tufts University; 1986
  324. 324. Lopes NP, Kato MJ, Andrade EHA, Maia JGS, Yoshida M, Planchart AR, et al. Antimalarial use of volatile oil from leaves of Virola surinamensis (Rol.) Warb. by Waiãpi Amazon Indians. Journal of Ethnopharmacology. 1999;67:313-319. DOI: 10.1016/s0378-8741(99)00072-0
  325. 325. Schultes RE. Evolution of the identification of the myristicaceous hallucinogens of South America. Journal of Ethnopharmacology. 1979;1:211-239. DOI: 10.1016/s0378-8741(79)80013-6
  326. 326. Barata LES, Baker PM, Gottlieb OR, Rúveda EA. Neolignans of Virola surinamensis. Phytochemistry. 1978;17:783-785
  327. 327. Lopes NP, Blumenthal EEA, Cavalheiro AJ, Kato MJ, Yoshida M. Lignans, g-lactones and propiophenones of Virola surinamensis. Phytochemistry. 1996;43:1089-1092
  328. 328. Lopes NP, Chicaro P, Kato MJ, Albuquerque S, Yoshida M. Flavonoids and lignans from Virola surinamensis twigs and their in vitro activity against Trypanosoma cruzi. Planta Medica. 1998;64:667-669
  329. 329. Barata LE, Santos LS, Ferri PH, Phillipson JD, Paine A, Croft SL. Anti-leishmanial activity of neolignans from Virola species and synthetic analogues. Phytochemistry. 2000;55:589-595. DOI: 10.1016/s0031-9422(00)00240-5
  330. 330. Veiga A, Albuquerque K, Corrêa ME, Brigido H, Silva E, Silva J, et al. Leishmania amazonensis and Leishmania chagasi: in vitro leishmanicide activity of Virola surinamensis (Rol.) Warb. Experimental Parasitology. 2017;175:68-73. DOI: 10.1016/j.exppara.2017.02.005
  331. 331. Cabral MMO, Alencar JA, Guimarães AE, Kato MJ. Larvicidal activity of grandisin against Aedes aegypti. Journal of the American Mosquito Control Association. 2009;25:103-105. DOI: 10.2987/08-5828.1
  332. 332. Nogueira CD, de Mello RP, Kato MJ, Cabral MM. Disruption of Chrysomya megacephala growth caused by lignin grandisin. Journal of Medical Entomology. 2009;46:281-283. DOI: 10.1603/033.046.0212
  333. 333. Carvalho AAV, Galdino PM, Nascimento MVM, Kato MJ, Valadares MC, Cunha LC, et al. Antinociceptive and antiinflammatory activities of grandisin extracted from Virola surinamensis. Phytotherapy Research. 2010;24:113-118. DOI: 10.1002/ptr.2882
  334. 334. Hiruma-Lima CA, Batista LM, de Almeida ABA, de Pietro ML, Campaner dos Santos L, Vilegas W, et al. Antiulcerogenic action of ethanolic extract of the resin from Virola surinamensis Warb. (Myristicaceae). Journal of Ethnopharmacology. 2009;122:406-409. DOI: 10.1016/j.jep.2008.12.023
  335. 335. Lima EO, Maia RF, Barbosa RCSBC, Xavier-Filho L, Paulo MQ, Santos LS, et al. Atividade anti microbiana de neolignanas 8.O.40 e derivados sinteticos-l. Ciência, Cultura e Saúde. 1987;9:55-57
  336. 336. Zacchino S, Rodríguez G, Santecchia C, Pezzenati G, Giannini F, Enriz R. In vitro studies on mode of action of antifungal 8.O.4'-neolignans occurring in certain species of Virola and related genera of Myristicaceae. Journal of Ethnopharmacology. 1998;62:35-41. DOI: 10.1016/s0378-8741(98)00056-7
  337. 337. Sherazi ST, Mahesar S, Arain A, Uddin S. Guava (Psidium guajava) oil (Chapter 27). In: Ramadan MF, editor. Fruit Oils: Chemistry and Functionality. Cham, Switzerland: Springer Nature Switzerland AG; 2019. pp. 541-559. DOI: 10.1007/978-3-030-12473-1_27
  338. 338. Joshi N, Patidar K, Solanki R, Mahawar V. Preparation and evaluation of herbal hair growth promoting shampoo formulation containing Piper betle and Psidium guajava leaves extract. International Journal of Green Pharmacy. 2018;12(Suppl):S835-S839. DOI: 10.22377/IJGP.V12I04.2263
  339. 339. Gutiérrez RM, Mitchell S, Solis RV. Psidium guajava: A review of its traditional uses, phytochemistry and pharmacology. Journal of Ethnopharmacology. 2008;117:1-27. DOI: 10.1016/j.jep.2008.01.025
  340. 340. Shruthi SD, Roshan A, Timilsina SS, Sunita S. A review on the medicinal plant Psidium guajava Linn. (Myrtaceae). Journal of Drug Delivery and Therapeutics. 2013;3:162-168
  341. 341. Naseer S, Hussain S, Naeem N, Pervaiz M, Rahman M. The phytochemistry and medicinal value of Psidium guajava (guava). Clinical Phytoscience. 2018;4:32. DOI: 10.1186/s40816-018-0093-8
  342. 342. Ngbolua K, Lufuluabo LG, Moke LE, Bongo GN, Liyongo CI, Ashande CM, et al. A review on the phytochemistry and pharmacology of Psidium guajava L. (Myrtaceae) and future direction. Discovery Phytomedicine. 2018;5:7-13. DOI: 10.15562/phytomedicine.2018.58
  343. 343. Shah AJ, Begum S, Hassan SI, Ali SN, Siddiqui BS, Gilani A-H. Pharmacological basis for the medicinal use of Psidium guajava leaf in hyperactive gut disorders. Bangladesh Journal of Pharmacology. 2011;6:100-105. DOI: 10.3329/bjp.v6i2.8692
  344. 344. Wei L, Li Z, Chen B. Clinical study on treatment of infantile rotaviral enteritis with Psidium guajava L. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2000;20:893-895
  345. 345. Tona L, Kambu K, Ngimbi N, Mesia K, Penge O, Lusakibanza M, et al. Antiamoebic and spasmolytic activities of extracts from some antidiarrheal traditional preparations used in Kinshasa, Congo. Phytomedicine. 2000;7:31-38. DOI: 10.1016/S0944-7113(00)80019-7
  346. 346. Lozoya X, Reyes-Morales H, Chávez-Soto MA, Martínez-García MC, Soto-González Y, Doubova SV. Intestinal antispasmodic effect of a phytodrug of Psidium guajava folia in the treatment of acute diarrhoeal diseases. Journal of Ethnophamacology. 2002;83:19-24. DOI: 10.1016/s0378-8741(02)00185-x
  347. 347. Jaiarj P, Khoohaswan P, Wongkrajang Y, Peungvicha P, Suriyawong P, Saraya ML, et al. Anticough and antimicrobial activities of Psidium guajava Linn. leaf extract. Journal of Ethnopharmacology. 1999;67:203-212. DOI: 10.1016/s0378-8741(99)00022-7
  348. 348. Ojewole JA. Antiinflammatory and analgesic effects of Psidium guajava Linn. (Myrtaceae) leaf aqueous extract in rats and mice. Methods and Findings in Experimental and Clinical Pharmacology. 2006;7:441-446. DOI: 10.1358/mf.2006.28.7.1003578
  349. 349. Lutterodt GD, Maleque A. Effects on mice locomotor activity of a narcotic-like principle from Psidium guajava leaves. Journal of Ethnopharmacology. 1988;24:219-231. DOI: 10.1016/0378-8741(88)90155-9
  350. 350. Shaheen HM, Ali BH, Alqarawi AA, Bashir AK. Effect of Psidium guajava leaves on some aspects of the central nervous system in mice. Phytotherapy Research. 2000;14:107-111. DOI: 10.1002/(sici)1099-1573(200003)14:2<107::aid-ptr602>3.0.co;2-z
  351. 351. Meckes M, Calzada F, Tortoriello J, Gonzalez JL, Martinez M. Terpenoids isolated from Psidium guajava with depressant activity on central nervous system. Phytotherapy Research. 1996;10:600-603. DOI: 10.1002/(SICI)1099-1573(199611)10:7<600::AID-PTR918>3.0.CO;2-7
  352. 352. Chiwororo WD, Ojewole JA. Biphasic effect of Psidium guajava Linn. (Myrtaceae) leaf aqueous extract on rat isolated vascular smooth muscles. Journal of Smooth Muscle Research. 2008;44:217-229. DOI: 10.1540/jsmr.44.217
  353. 353. Singh RB, Rastogi SS, Singh NK, Ghosh S, Niaz MA. Effects of guava intake on serum total and high-density lipoprotein cholesterol levels and on systemic blood pressure. American Journal of Cardiology. 1992;70:1287-1291. DOI: 10.1016/0002-9149(92)90763-o
  354. 354. Singh RB, Rastogi SS, Singh NK, Ghosh S, Gupta S, Niaz MA. Can guava fruit intake decrease blood pressure and blood lipids? Journal of Human Hypertension. 1993;7:33-38
  355. 355. Rai PK, Mehta S, Watal G. Hypolipidaemic and hepatoprotective effects of Psidium guajava raw fruit peel in experimental diabetes. Indian Journal of Medical Research. 2010;131:820-824
  356. 356. Kumari S, Rakavi R, Mangaraj M. Effect of guava in blood glucose and lipid profile in healthy human subjects: A randomized controlled study. Journal of Clinical and Diagnostic Research. 2016;10:BC04-BC07. DOI: 10.7860/JCDR/2016/21291.8425
  357. 357. Nundkumar N, Ojewole JA. Studies on the antiplasmodial properties of some South African medicinal plants used as antimalarial remedies in Zulu folk medicine. Methods and Findings in Experimental and Clinical Pharmacology. 2002;24:397-401. DOI: 10.1358/mf.2002.24.7.696540
  358. 358. Tangpu TV, Yadav AK. Anticestodal efficacy of Psidium guajava against experimental Hymenolepis diminuta infection in rats. Indian Journal of Pharmacology. 2006;38:29-32. DOI: 10.4103/0253-7613.19849
  359. 359. Qadan F, Thewaini AJ, Ali DA, Afifi R, Elkhawad A, Matalka KZ. The antimicrobial activities of Psidium guajava and Juglans regia leaf extracts to acne-developing organisms. American Journal of Chinese Medicine. 2005;33:197-204. DOI: 10.1142/S0192415X05002783
  360. 360. Nwinyi O, Chinedu SN, Ajani OO. Evaluation of antibacterial activity of Psidium guajava and Gongronema latifolium. Journal of Medicinal Plants Research. 2008;2:189-192
  361. 361. Buvaneswari S, Raadha CK, Krishnaveni N, Jayashree S. In vitro antimicrobial activity of Psidium guajava against clinically important strains. E-Journal of Life Sciences. 2011;1:14-22
  362. 362. Anas K, Jayasree PR, Vijayakumar T, Manish Kumar PR. In vitro antibacterial activity of Psidium guajava Linn. leaf extract on clinical isolates of multidrug resistant Staphylococcus aureus. Indian Journal of Experimental Biology. 2008;46:41-46
  363. 363. Rahim N, Gomes DJ, Watanabe H, Rahman SR, Chomvarin C, Endtz HP, et al. Antibacterial activity of Psidium guajava leaf and bark against multidrug-resistant Vibrio cholerae: Implication for cholera control. Japanese Journal of Infectious Diseases. 2010;63:271-274
  364. 364. Rodriguez RC, Cruz PH, Rios HG. Lectins in fruits having gastrointestinal activity: Their participation in hemagglunating property of Escherichia coli O157:H7. Archives of Medical Research. 2001;32:251-257. DOI: 10.1016/s0188-4409(01)00287-9
  365. 365. Al Aboody MS, Mickymaray S. Antifungal efficacy and mechanisms of flavonoids. Antibiotics (Basel). 2020;9:45. DOI: 10.3390/antibiotics9020045
  366. 366. Bashir O, Jan N, Gani G, Naik H, Hussain S, Reshi M, et al. Food applications of Nigella sativa seeds (Chapter 13). In: Ramadan MF, editor. Black Cumin (Nigella sativa) Seeds: Chemistry, Technology, Functionality, and Applications. Food Bioactive Ingredients. Vol. 2021. Switzerland: Springer; 2021. pp. 191-208. DOI: 10.1007/978-3-030-48798-0_13
  367. 367. Yarnell E, Abascal K. Nigella sativa: holy herb of the Middle East. Alternative and Complementary Therapies. 2011;17:99-105
  368. 368. Ahmad A, Husain A, Mujeeb M, Khan SA, Najmi AK, Siddique NA, et al. A review on therapeutic potential of Nigella sativa: A miracle herb. Asian Pacific Journal of Tropical Biomedicine. 2013;3:337-352. DOI: 10.1016/S2221-1691(13)60075-1
  369. 369. Kumar R. Edible oils: Natural healer for fungal infections. Scientific India. 2017;5:11-14. DOI: 10.13140/RG.2.2.28738.89281
  370. 370. Bukhari MH, Khalil J, Qamar S, Qamar Z, Zahid M, Ansari N, et al. Comparative gastroprotective effects of natural honey, Nigella sativa and cimetidine against acetylsalicylic acid induced gastric ulcer in albino rats. Journal of the College of Physicians and Surgeons Pakistan. 2011;21:151-156
  371. 371. Hannan A, Saleem S, Chaudhary S, Barkaat M, Arshad MU. Antibacterial activity of Nigella sativa against clinical isolates of methicillin-resistant Staphylococcus aureus. Journal of Ayub Medical College, Abbottabad. 2008;20:72-74
  372. 372. Hanafy MSM, Hatem ME. Studies on the antimicrobial activity of Nigella sativa seed (black cumin). Journal of Ethnopharmacology. 1991;34:275-278. DOI: 10.1016/0378-8741(91)90047-H
  373. 373. Khan MA, Ashfaq MK, Zuberi HS, Mahmood MS, Gilani AH. The in vivo antifungal activity of the aqueous extract from Nigella sativa seeds. Phytotherapy Research. 2003;17:183-186. DOI: 10.1002/ptr.1146
  374. 374. Bita A, Rosu AF, Calina D, Rosu L, Zlatian O, Dindere C, et al. An alternative treatment for Candida infections with Nigella sativa extracts. European Journal of Hospital Pharmacy. 2012;19:162. DOI: 10.1136/ejhpharm-2012-000074.203
  375. 375. Azeiz AAZ, Saad AH, Darweesh MF. Efficacy of thymoquinone against vaginal candidiasis in prednisolone-induced immunosuppressed mice. The Journal of American Science. 2013;9:155-159
  376. 376. Agarwal R, Kharya MD, Shrivastava R. Antimicrobial and anthelmintic activities of the essential oil of Nigella sativa Linn. Indian Journal of Experimental Biology. 1979;17:1264-1265
  377. 377. Aboul Ela MA, El-Shaer NS, Ghanem NB. Antimicrobial evaluation and chromatographie analysis of some essential and fixed oils. Pharmazie. 1996;51:993-994
  378. 378. Bourrel C, Dargent R, Vilrem G, Gaset A. Chemical analysis and fungistatic properties of some essential oils in a liquid medium. Effects on hyphal morphogenesis. Rivista Italiana EPPOS. 1995;6:31-42
  379. 379. Singh G, Marimuthu P, de Heluani CS, Catalan C. Chemical constituents and antimicrobial and antioxidant potentials of essential oil and acetone extract of Nigella sativa seeds. Journal of the Science of Food and Agriculture. 2005;85:2297-2306. DOI: 10.1002/jsfa.2255
  380. 380. Sunita M, Meenakshi SH. Chemical composition and antidermatophytic activity of Nigella sativa essential oil. African Journal of Pharmacy and Pharmacology. 2013;7:1286-1292. DOI: 10.5897/AJPP12.377
  381. 381. El-Nagerabia SA, Al-Bahryb SN, Elshafieb AE, AlHilalib S. Effect of Hibiscus sabdariffa extract and Nigella sativa oil on the growth and aflatoxin B1 production of Aspergillus flavus and Aspergillus parasiticus strains. Food Control. 2012;25:59-63. DOI: 10.1016/j.foodcont.2011.09.033
  382. 382. Fierro IM, Barja-Fidalgo C, Cunha FQ, Ferreira SH. The involvement of nitric oxide in the anti-Candida albicans activity of rat neutrophils. Immunology. 1996;89:295-300. DOI: 10.1046/j.1365-2567.1996.d01-742.x
  383. 383. Rogozhin EA, Oshchepkova YI, Odintsova TI, Khadeeva NV, Veshkurova ON, Egorov TA, et al. Novel antifungal defensins from Nigella sativa L. seeds. Plant Physiology and Biochemistry. 2011;49:131-137. DOI: 10.1016/j.plaphy.2010.10.008
  384. 384. Avula B, Chittiboyina AG, Wang Y-H, Sagi S, Raman V, Wang M, et al. Simultaneous determination of aegeline and six coumarins from different parts of the plant Aegle marmelos using UHPLC-PDA-MS and chiral separation of aegeline enantiomers using HPLC-ToF-MS. Planta Medica. 2016;82:580-588. DOI: 10.1055/s-0042-103160
  385. 385. Karmase A, Jagtap S, Bhutani KK. Anti adipogenic activity of Aegle marmelos Correa. Phytomedicine. 2013;20:1267-1271
  386. 386. Roytman MM, Pörzgen P, Lee CL, Huddleston L, Kuo TT, Bryant-Greenwood P, et al. Outbreak of severe hepatitis linked to weight-loss supplement OxyELITE Pro. American Journal of Gastroenterology. 2014;109:1296-1298. DOI: 10.1038/ajg.2014.159
  387. 387. Pathirana CK, Madhujith T, Eeswara J. Bael (Aegle marmelos L. Correa), a medicinal tree with immense economic potentials. Advances in Agriculture. 2020:8814018. DOI: 10.1155/2020/8814018
  388. 388. Sarkar T, Salauddin M, Chakraborty R. In-depth pharmacological and nutritional properties of bael (Aegle marmelos): A critical review. Journal of Agriculture and Food Research. 2020;2:100081. DOI: 10.1016/j.jafr.2020.100081
  389. 389. Manandhar B, Paudel KR, Sharma B, Karki R. Phytochemical profile and pharmacological activity of Aegle marmelos Linn. Journal of Integrative Medicine. 2018;16:153-163. DOI: 10.1016/j.joim.2018.04.007
  390. 390. Mazumder R, Bhattacharya S, Mazumder A, Pattnaik AK, Tiwary PM, Chaudhary S. Antidiarrhoeal evaluation of Aegle marmelos (Correa) Linn. root extract. Phytotherapy Research. 2006;20:82-84. DOI: 10.1002/ptr.1804
  391. 391. Arul V, Miyazaki S, Dhananjayan R. Studies on the anti-inflammatory, antipyretic and analgesic properties of the leaves of Aegle marmelos Corr. Journal of Ethnopharmacology. 2005;96:159-163
  392. 392. Sharma B, Satapathi SK, Roy P. Hypoglycemic and hypolipidemic effect of Aegle marmelos (L.) leaf extract on streptozotocin-induced diabetic mice. International Journal of Pharmacology. 2007;3:444-452
  393. 393. Sevugan A, Subramanian K, Balamuthu K, Abdul BAA, Mohammed AA, Mandali VR. Antidiabetic activity of leaf and callus extracts of Aegle marmelos in rabbit. Science Asia. 2008;2008(34):317-321
  394. 394. Patel AR, Garach D, Chakraborty M, Kamath JV. Aegle marmelos (Linn.): A therapeutic boon for human health. Indian Journal of Research in Ayurveda and Pharmacy. 2012;3:159-163
  395. 395. Jaswanth A, Loganathan V, Manimaran S. Wound healing activity of Aegle marmelos. Indian Journal of Pharmaceutical Sciences. 2001;63:41-44
  396. 396. Rani P, Khullar N. Antimicrobial evaluation of some medicinal plants for their anti-enteric potential against multi-drug resistant Salmonella typhi. Phytotherapy Research. 2004;18:670-673. DOI: 10.1002/ptr.1522
  397. 397. Balakumar S, Rajan S, Thirunalasundari T, Jeeva S. Antifungal activity of Aegle marmelos (L.) Correa (Rutaceae) leaf extract on dermatophytes. Asian Pacific Journal of Tropical Biomedicine. 2011;1:309-312. DOI: 10.1016/S2221-1691(11)60049-X
  398. 398. Yogeshwar M, Gade RM, Shitole AV. Evaluation of antifungal activities of extracts of Aegle marmelos, Syzygium cumini and Pongamia pinnata against Pythium debaryanum. Indian Journal of Pharmaceutical Sciences. 2017;79:377-384. DOI: 10.4172/pharmaceutical-sciences.1000240
  399. 399. Singh KV, Bhatt SK, Sthapak JK. Antimicrobial and anthelmintic properties of the seeds of Aegle marmelos. Fitoterapia. 1983;54:261-264
  400. 400. Banerjee AK, Kaul VK, Nigam SS. Chemical, microbial and anthelmintic examination of the seeds of Aegle marmelos Corr. Indian Drugs. 1984;21:217-218
  401. 401. Mishra BB, Kishore N, Tiwari VK, Singh DD, Tripathi V. A novel antifungal anthraquinone from seeds of Aegle marmelos Correa (family Rutaceae). Fitoterapia. 2010;81:104-107. DOI: 10.1016/j.fitote.2009.08.009
  402. 402. Mishra BB, Singh DD, Kishore N, Tiwari VK, Tripathi V. Antifungal constituents isolated from the seeds of Aegle marmelos. Phytochemistry. 2010;71:230-234. DOI: 10.1016/j.phytochem.2009.10.013
  403. 403. Pitre S, Srivastava SK. Pharmacological, microbiological and phytochemical studies on the roots of Aegle marmelos. Fitoterapia. 1987;58:194-197
  404. 404. Sliemandagger TA, Nicholson WL. Role of dipicolinic acid in survival of Bacillus subtilis spores exposed to artificial and solar UV radiation. Applied and Environmental Microbiology. 2001;2001(67):1274-1279. DOI: 10.1128/aem.67.3.1274-1279.2001
  405. 405. Mans DRA. From forest to pharmacy: Plant-based traditional medicines as sources for novel therapeutics. Academia Journal of Medicinal Plants. 2013;1:101-110
  406. 406. Van Der Heijden R, Jacobs DI, Snoeijer W, Hallard D, Verpoorte R. The Catharanthus alkaloids: Pharmacognosy and biotechnology. Current Medical Chemistry. 2004;11:607-628. DOI: 10.2174/0929867043455846
  407. 407. Kingston DG, Newman DJ. Taxoids: Cancer-fighting compounds from nature. Current Opinion in Drug Discovery and Development. 2007;10:130-144
  408. 408. Vane JR. The history of inhibitors of angiotensin-converting enzyme. Journal of Physiology and Pharmacology. 1999;50:489-498
  409. 409. Furman B. The development of Byetta (exenatide) from the venom of the Gila monster as an anti-diabetic agent. Toxicon. 2012;59:464-471. DOI: 10.1016/j.toxicon.2010.12.016
  410. 410. Shafia S, Chandluri P, Ganpisetti R, Lakshmi BVS, Swami PA. Erythromycin use as broad spectrum antibiotic. World Journal of Pharmaceutical and Medical Research. 2016;2:23-26
  411. 411. Cuddihy G, Wasan EK, Di Y, Wasan KM. The development of oral amphotericin B to treat systemic fungal and parasitic infections: Has the myth been finally realized? Pharmaceutics. 2019;11:99. DOI: 10.3390/pharmaceutics11030099
  412. 412. Perlin DS, Shor E, Zhao Y. Update on antifungal drug resistance. Current Clinical Microbiology Reports. 2015;2:84-95. DOI: 10.1007/s40588-015-0015-1
  413. 413. Bodeker C, Bodeker G, Ong CK, Grundy CK, Burford G, Shein K. WHO Global Atlas of Traditional, Complementary and Alternative Medicine. Geneva, Switzerland: World Health Organization; 2005

Written By

Dennis R.A. Mans

Reviewed: 17 January 2022 Published: 23 February 2022