Next Article in Journal
Enteric Pathogens Detected in Children under Five Years Old Admitted with Diarrhea in Moshi, Kilimanjaro, Tanzania
Previous Article in Journal
Tsunami-Driven Spread of Toxoplasma gondii and Other Microbial Pathogens: Implications for Cetacean Health and Conservation
Previous Article in Special Issue
Exploring Possible Ways to Enhance the Potential and Use of Natural Products through Nanotechnology in the Battle against Biofilms of Foodborne Bacterial Pathogens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antifungal Compounds from Microbial Symbionts Associated with Aquatic Animals and Cellular Targets: A Review

1
Institute of Fisheries and Aquatic Science, University of Douala, Douala P.O. Box 2701, Cameroon
2
Department of Biomedical Sciences, University of Buea, Buea P.O. Box 63, Cameroon
3
Department of Biological Sciences, University of Cape Town, Private Bag X3, Rondebosch, Cape Town 7701, South Africa
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(4), 617; https://doi.org/10.3390/pathogens12040617
Submission received: 18 March 2023 / Revised: 11 April 2023 / Accepted: 13 April 2023 / Published: 18 April 2023

Abstract

:
Fungal infections continue to be a serious public health problem, leading to an estimated 1.6 million deaths annually. It remains a major cause of mortality for people with a weak or affected immune system, such as those suffering from cancer under aggressive chemotherapies. On the other hand, pathogenic fungi are counted among the most destructive factors affecting crops, causing a third of all food crop losses annually and critically affecting the worldwide economy and food security. However, the limited number currently available and the cytotoxicity of the conventional antifungal drugs, which are not yet properly diversified in terms of mode of action, in addition to resistance phenomena, make the search for new antifungals imperative to improve both human health and food protection. Symbiosis has been a crucial alternative for drug discovery, through which many antimicrobials have been discovered. This review highlights some antifungal models of a defensive symbiosis of microbial symbiont natural products derived from interacting with aquatic animals as one of the best opportunities. Some recorded compounds with supposed novel cell targets such as apoptosis could lead to the development of a multitherapy involving the mutual treatment of fungal infections and other metabolic diseases involving apoptosis in their pathogenesis pathways.

1. Introduction

Fungal infections are caused by pathogenic fungi that can be superficial at the level of the skin or biological mucosa, or they can be systemic, disseminated inside the whole body or on a particular inner organ, such as the lung, brain, heart, liver, and blood. These infections continue to be a serious public health problem, leading to an estimated 1.6 million deaths annually [1]. Fungal infections are dangerous and remain a major cause of mortality for people with a weak or affected immune system, such as those suffering from cancer under aggressive chemotherapies. Besides, the number of immunocompromised is growing due to an increase in the availability of hospital care, the introduction of suppressive immunotherapy, and the emerging immune diseases such as immunodeficiency viral infections, cancers, and organ transplantation that make these vulnerable populations exposed to such opportunistic fungal infections. On the other hand, pathogenic fungi are counted among the most destructive factors affecting crops, causing a third of all food crop losses annually, which critically affects the worldwide economy and food security. Aquaculture is also impaired by pathogenic fungi, which reduce fish production yield. According to the FAO’s report from 2009 to 2010 (https://www.fao.org accessed on 2 February 2023), fungal infections affect the most important crops in the world in order of occurrence: maize, potatoes, soybean, rice, and wheat. Because of these crop losses, 61% of the worldwide population will lack food [2,3]. However, the limited number currently available and the cytotoxicity of the conventional antifungal drugs, which are not yet properly diversified in terms of mode of action, in addition to resistance phenomena, make the research for new antifungals imperative to improve both human health and crop protection [4,5,6]. For many years, research carried out for drug discovery has involved a lot of natural and artificial sources. Among them, natural sources seemed to represent the most abundant and diverse place to find new bioactive molecules. In that way, symbiosis has been a crucial alternative for drug discovery, through which many antimicrobials have been discovered. Symbiosis can be mutualistic, commensal, or parasitic. Further, it can be optional when both organisms live independently or obligatory when these organisms need each other to survive. During that biological collaboration, a set of secondary metabolites is produced, playing an important role in the management of the interactions between the concerned organisms [7]. These metabolites could serve as a starting point for drug discovery and production.
The discovery of antimicrobials derived from microorganisms involved in symbiosis constitutes a field of research that is not widely explored. However, some bioactive molecules against pathogenic fungi have been isolated from bacteria on Acromyrmex octospinosus ants. Nystatin P1 is an antifungal compound produced by Pseudonocardia sp. through a nystatin-like biosynthetic gene cluster identified in the bacteria as symbionts on A. octospinosus [8]. Moreover, it was recently shown that candicidin and antimycins are also produced by Streptomyces sp. symbionts and exhibit antifungal activity against Candida albicans (C. albicans) and Escovopsis sp. [9,10]. Griseofulvin too (7-chloro-4,6,2′-trimethoxy-6′-methylgris-2′-en-3,4′-dione) is an antifungal mycotoxin isolated from Penicillium species. It has been widely used for the treatment of superficial dermatomycoses [11]. The mycotoxin was first isolated from a terrestrial fungus, the mold Penicillium griseofulvum [12]. The bioactive compound was also detected in the aquatic animal sponge Axinella verrucosa (A. verrucosa) through its fungal symbiont from the Penicillium genus. The ethyl acetate extract of the Penicillium yielded the production of griseofulvin [13].
This review highlights the involvement of microbial symbiont interactions with aquatic animals and the possibility of biomimicry of these interactions for the discovery of new bio antifungals with new cellular targets to better fight antimicrobial resistance and provides recommendations for future research concerning antifungal discovery. The next sections of this work will be focused on the explanations of why attention is being turned to microbial symbiosis with aquatic animals, in particular, some antifungal compounds isolated from such symbiosis together with their cellular targets and recommendations concerning future research studies.

2. A Microbial Symbiosis Approach Associated with Aquatic Animals for Antimicrobial Discovery

The current available antifungal drugs exhibit toxicity and resistance phenomena. Pathogenic fungi are eukaryotic cells, just as are human cells. So, they share the same targets, which do not ease the development of new drugs and lead to cytotoxicity. On the other hand, the limited available classes or cell targets of these antifungals promote the development of antibiotic resistance. Regarding the current and persistent problem in that research field, new antifungal molecules are now in the development process. However, a lot of these new molecules still target the same fungal cell sites [6], which predicts future resistance. So, to contribute to expanding the research on new compounds with new cell targets, aquatic environments with an emphasis on microbial symbiosis in aquatic animals are proposed here as potent bioresources.
Water as oceans is the most abundant molecule on the earth’s surface, occupying more than 70% of the surface of the planet with the highest worldwide biodiversity [14]. About 1.5 million species have already been discovered worldwide. Of those, 1.5 million, or 80%, are of terrestrial origin, whereas aquatic environments only account for 20% [15]. However, some scientists affirmed that the more complex or larger an ecosystem is, the greater its biodiversity. According to this approach, aquatic ecosystems, which are more abundant on the planet, may contain more new species than terrestrial surfaces. But the lowest percentage of the identified species in water environments could be justified by the fact that water ecosystems are underexplored, which opens new perspectives for the discovery of new animal organisms with potent microbial symbionts.
The investigation was focused on symbiosis associated with animals because Homo sapiens and animals are biologically closed [16,17]. Moreover, for in vivo tests using animal models, the human response is closely compared to one of those animals. It does not mean that they are identical. However, their defense systems against a toxic or a microbial aggressor sometimes involve the same biochemical pathways. Animal biology remains the focal point for studying humanity’s survival strategies [18]. Furthermore, this approach could well be applied to the protection of plants and foodstuffs because some pathogenic fungi affecting animals also cause diseases on plants and various foodstuffs, but above all, they all often share similar cell structures, which could represent common antifungal targets. This broadens the scope of research for antifungals against human, plant, and other food infections starting from the same bioresource represented by microbial symbioses of aquatic origin.
Aquatic animals use many features to defend themselves against a plethora of infectious diseases. These features include mucus, skin, and gills, which constitute protection toward their natural niches and against pathogenic agents. Mucus is found on the skin, gills, and intestines of organisms and contains antimicrobial agents such as proteins, lysozyme, immunoglobulin, enzymes, and lectins that could benefit human health research in clinical studies [19]. In that way, some experimental activities have shown the antimicrobial activities of the mucus extracts of Maculabatis gerrardi and Pastinachus sephen against Candida tropicalis (C. tropicalis), Aspergillus niger (A. niger), Penicillium sp., Trichophyton mentagrophytes, Alternaria alternata (A. alternata), C. albicans, Rhizopus sp., Mucor sp., and Trichophyton rubrum [20]. A methanol extract of Perna viridis gills also exhibited potent antifungal activity against A. niger and Mucor sp. Moreover, in amphibians, some symbiotic skin bacteria located on the frog skin produce antifungal metabolites towards the cutaneous pathogen Batrachochytrium dendrobatidis (B. dendrobatidis), which has caused many amphibian population declines and extinctions [21]. On the other hand, some studies suggest that the population of microbial symbionts existing in mucus is responsible for the production of specialized metabolites involved in the management of host pathogen proliferation. In addition to mucus, other organs such as gills, skin, gut, and olfactory organs display a wide diversity of microorganisms living in symbiosis with aquatic animals, which are responsible for anti-infectious activity [22,23].
So, in the context of natural product chemistry, defensive symbioses can produce novel bioactive compounds to protect the host, leading to an interest in human health and crop protection [24]. Symbiosis interactions offer a biological platform for the exchange of bioactive compounds between the host and the microbial community. The naturally produced products act through different mechanisms. They could function as deterrents to predation, nutritional sources, immunomodulatory factors, genes transferred to the host, or antibiotics [25]. Our field of interest resides in their antibiotic function.
One of the best opportunities to exploit the chemical-mediated defensive symbiosis interaction between microorganisms and aquatic animals is to biomimic the mechanism of that defense against infectious agents. That is why investigating animal defense systems against fungal aggressors through biomimicry could shed light on new effective antifungal agents to treat fungal infections in humans, other animal species, and even crops.

3. Aquatic Animal-Microbial Symbionts Derived Antifungal Compounds and Their Targets

Some models of defensive symbiosis between microbial symbionts and natural products derived from their interactions with aquatic animals have been defined here. The natural products are grouped according to 7 cell targets (cellular enzymes, resistance factors, cell wall, cell multiplication and differentiation, plasma membrane, immunomodulation-apoptosis, and multicellular targets). Human fungal pathogenic fungi, Aspergillus sp., Candida albicans, and Trichophyton sp., were susceptible to 1–3 µg/L concentrations of the bioactive compound. Concerning plant protection, inhibitory values were recorded up to 0.5 µg/L and 0.39 μmol/L, 25-fold stronger than that of the positive control, ketoconazole, for the inhibition of Phytophthora capsici (P. capsici) zoospore motility and Pestalotia calabae (P. calabae), respectively. The following Table 1 highlights the details of the bioactive compounds, their producing sources, their antifungal cellular targets, and the available chemical structure.

3.1. Compounds Acting on the Plasma Membrane

A total of five organic compounds were identified as acting on the plasma membrane: iturin, 3,5-dibromo-2-(3,5-dibromo-2-methoxyphenoxy) phenol, YM-202204, theonellamide F, and theopalauamide.

3.1.1. Iturin

A. aerophoba is a sponge that contains a large number of bacteria. Antimicrobial activities of bacterial isolates from A. aerophoba were tested against many microbial pathogens. The results showed that B. subtilis strains A184, A190, and A202 exhibited strong activity against the fungus C. albicans [13,90]. According to some research results, the fungicidal activity in Bacillus is an indicator of the presence of lipopeptide from the iturin (compound 9) class [90]. Compound 9 is included in the lipopeptide group, which acts as an immune stimulator in plants. Its main structure is heptapeptides linked to a β-amino fatty acid chain with a length of 14 to 17 carbons and a molecular mass of ~1.1 kDa [91]. The differences in heptapeptides show derivative compounds of iturin (e.g., bacillomycin, mycosubtilin) [44]. Their action mechanism involves the disturbance of the plasma membrane of the fungal target. The compound affects the morphology and membrane structure of yeast cells by increasing the electrical conductance of bimolecular lipid membranes and acting as a nontoxic and nonpyrogenic immunological adjuvant [43,46].

3.1.2. 3,5-Dibromo-2-(3,5-dibromo-2-methoxyphenoxy)phenol

It has been proven that Vibrio sp. bacteria isolated from the sponge Dysidea sp. were able to biosynthesize 3,5-dibromo-2-(3′,5′-dibromo-2′-methoxyphenoxy) phenol (compound 10). The compound was isolated from ethanol extracts of the sponge Dysidea sp. It belongs to the group of brominated diphenyl ethers, with a molecular mass of 531.82 g/mol [47]. Inhibitory parameters of compound 10 were determined against pathogenic fungi. A. fumigatus, A. fumigatus, C. albicans, and C. tropicalis were inhibited at 7.8 mg/mL. A. flavus and C. glabrata have shown susceptibility concentrations of 1.95 mg/mL and 15.2 mg/mL, respectively. Moreover, the compound exhibited fungicidal activity against A. fumigatus at 15.62 mg/mL and against C. albicans at 7.81 mg/mL. The investigation of a possible target revealed the disruption of the fungal cell membrane, expressed primarily in the leakage of potassium ions [48].

3.1.3. YM-202204

YM-202204 (compound 11) is an antifungal that also exhibits antibacterial properties. It was discovered in the culture broth of the marine fungus Phoma sp. Q60596. The structure is a pyrone determined by several spectroscopic experiments as a lactone compound appearing as a yellow syrup with a molecular mass of 646 g/mol [49]. Some researchers have shown that pyrenes could also be isolated from other fungi, such as Fusarium species [49,92]. C. albicans, C. neoformans, and A. fumigatus were highly susceptible to compound 11. The latter inhibited glycosyl-phosphatidyl-inositol (GPI)-anchoring in yeast cell membranes [49].

3.1.4. Theonellamide F

Marine sponges from the Theonella and Discodermia genera contain a reserve of bioactive metabolites. Theonellamide F (compound 12) was isolated from alcohol and aqueous extracts of the marine sponge, genus Theonella. It is a dodecapeptide composed of L-Asn, L-aThr, two residues of L-Ser, L-Phe, bAla, (2S,3R)-3-hydroxyasparagine, (2S,4R)-Z-amino-4-hydroxyadipic acid, r-L-histidine-D-alanine, L-p-bromophenylalanine, and (3S,4S,5E,7E)-3-amino-4-hydroxy-6-methyl-8-p-bromo- phenyl)-5,7-octadienoic acid, with an unprecedented histidinoalanine bridge [50,93]. Compound 12 showed activity against several fungi, including Candida spp., Trichophyton spp., and Aspergillus spp., at active concentrations of 3.2–12 µg/mL. More recent biological studies using mutations/deletions of genes in the ergosterol biosynthetic pathway and calcein as a fluorescent dye have respectively shown that the yeast Cerevisiae pombe displayed high tolerances or slight resistances to compound 12 and a loss of membrane integrity when treated with the dye [52]. Theonellamides represent a new class of sterol-binding agents [51].

3.1.5. Theopalauamide

T. swinhoei is a Palauan sponge that contains bacterial symbionts that produce theopalauamide (compound 13). These bacterial symbionts are composed of C. Entotheonella palauensis from the d-subdivision of proteobacteria. Compound 13 is an antifungal, bicyclic glycopeptide presented in the form of a white powder with a molecular mass of 1746.9 g/mol [57]. Concerning its biological activity, yeast chemical-genomic approaches were used to determine the mode of action. The results of several research approaches have shown that theopalauamide represents a new class of sterol-binding compound. Biochemical experiments have specifically identified ergosterol as the primary target of theopalauamide [55,56].

3.2. Immunomodulation and Apoptosis

Mammals share with some pathogenic fungi two orthologous apoptosis genes: apoptosis-inducing factor (Aif) and endonuclease G (NuGl) that are responsible for programmed cell death (PCD), which represent challenging antifungal targets [94,95]. Two compounds (indole-3-carboxaldehyde and isatin) can modulate the immune system in order to stop the fungal infection and activate a cell-programmed death in the pathogen.

3.2.1. Indole 3-carboxaldehyde

Microbial symbiosis relationships with animals are not strange. Surface bacteria from amphibians’ skin are beneficial to the host amphibian [96]. The fungus J. lividum, living at the skin surface of the red-backed salamander P. cinereus (characterized by its ability to exploit both aquatic and terrestrial habitats), synthesizes secondary metabolites that inhibit the pathogenic fungus B. dendrobatidis, which is a chytrid causing Chytridiomycosis. Indole-3-carboxaldehyde (compound 14) is the heteroarenecarbaldehyde produced. The indole is a heteroarenecarbaldehyde, an indole alkaloid with 145.16 g/mol of molecular mass. The latter inhibits the pathogen’s growth at 68.9 μM. These results suggest that cutaneous symbiosis bacteria lead to amphibian resistance to fungal diseases. In addition, compound 14 also interferes with the growth of C. albicans by reducing vulvovagival candidiasis, as shown in an in vivo model study. The presence of compound 14 in the animal organism acts as immunomodulatory by stimulating the production of IL-22 via the aryl hydrocarbon receptor (AhR), promoting IL-18 expression, and providing protection against Candida infection [59].

3.2.2. Isatin

P. macrodactylus is a shrimp that is highly resistant to a fungal infection at its embryonic stage. The pathogenic fungus that causes the infection is L. callinectes, which also affects many other crustaceans. A particular bacterium, Alteromonas sp., spreads at the external envelope of the crustacean embryos and produces 2,3-indolinedione, commonly called isatin (compound 15). The compound inhibits the pathogenic fungus. Compound 15, which is also found in plants and other animals, possesses an indole ring structure, which is common to many pharmaceuticals and heterocyclic natural products of biological interest [97]. If exposed to the fungus, bacteria-free embryos quickly die, whereas similar embryos reinoculated with the bacteria or treated only with compound 15 survive. So, the symbiotic Alteromonas sp. bacteria protect P. macrodactylus embryos against L. callinectes infection by releasing the antifungal compound 15 [62]. In addition, other scientists have revealed the susceptibility of C. albicans, C. monosa, C. glabrata, T. longifusus, M. canis, and A. flavus to indole (0.25–1 mg/mL) [60,61]. While studying the mechanism of action of compound 15, closely related compounds have shown apoptotic effects on C. albicans at sub-inhibitory concentrations, suggesting targeting apoptosis could be considered as an alternative to be explored for antifungal drug discovery [63].

3.3. Cell Differentiation and Multiplication

Griseofulvine, majusculamide C, and surfactin are the three organic compounds that act as chemical interference in the multiplication and differentiation of pathogenic fungi by blocking mitosis and the genetic material of the pathogens.

3.3.1. Griseofulvin

The Penicillium fungus was isolated from the sponge A. verrucosa in the Mediterranean Sea. The microorganism Penicillium sp. has been submitted to an ethyl acetate maceration to obtain solvent extracts containing griseofulvin (compound 6) [13]. Compound 6 or 7-chloro-4,6,2′-trimethoxy-6′- methylgris-2′-en-3,4′-dione (352.8 g/mol) is a mycotoxin produced by various species of Penicillium [98]. Compound 6 disrupts the mitotic spindle by interacting with the polymerized microtubules, leading to the production of multinucleate fungal cells and stopping cell division at metaphase. The inhibition of nucleic acid synthesis and the formation of hyphal cell wall material also may be involved. The result is distortion, irregular swelling, and spiral curling of the hyphae. The bioactive molecule is fungistatic in mature dermatophytic cells and fungicidal in immature ones [99]. The isolated compound is a widely used antifungal drug for the treatment of superficial dermatomycoses. However, because it is carcinogenic and teratogenic in animal models, there is considerable concern regarding its clinical application. Further, it produces numerical chromosome aberrations in human lymphocytes and cell lines. There are conflicting reports on the ability of compound 6 to induce structural chromosomal aberrations. However, compound 6 induces micronucleus formation both in isolated peripheral lymphocytes and lymphocytes from whole blood cultures [11].

3.3.2. Majusculamide C

A cyclic depsipeptide, majusculamide C (compound 7), was isolated from the sponge P. trachys collected at the Enewetak Atoll (Marshall Island, Pacific Ocean). It was originally isolated from the toxic blue-green alga Lyngbya majuscula obtained from the same site. Compound 7 exhibited antifungal activity against pathogens of commercially important plants [13,38]. The compound is active against several plant pathogenic fungi such as R. solani, P. aphanidermatum, A. euteiches, and P. infestans at concentrations of, respectively, 4 µM, <1 µM, 2 µM, and 1 µM [40]. The cyclic depsipeptide acts as a microfilament-depolymerizing agent that shows potent fungicidal activity and may have been used in the treatment of resistant fungi-inducing diseases of domestic plants and crops [41].

3.3.3. Surfactin

The microflora of A. aerophoba constituted by bacteria B. subtilis A190 B. subtilis A184 was responsible for surfactin class secondary metabolites production [13]. Surfactin (compound 8), with a molecular weight of 1036.3 g/mol, was thus characterized as a lipopeptide composed of a heptapeptide with the following sequence: L-Glu1-L-Leu2-D-Leu3-L-Val4-L-Asp5-D-Leu6-L-Leu7, forming a lactone ring structure with a β-hydroxy fatty acid chain. Bearing both a hydrophilic peptide portion and a lipophilic fatty acid chain, compound 8 is amphiphilic, leading to exceptional biosurfactant activities and diverse biological activities [100]. The surfactin group has two polar amino acid residues, such as Glu and Asp, and it has been concluded that they bind with DNA via hydrogen bonds. The compound affected F. moniliforme growth by causing morphological changes in hyphae, suggesting that it markedly contributed to inhibiting fungal growth. DNA binding results indicated that the lipopeptide negatively influenced the maintenance of DNA integrity by binding to F. moniliforme DNA, which might in turn genetically affect DNA function for F. moniliforme growth [42].

3.4. Cellular Enzymes

Brefeldin A and roridin A were found to be cellular enzyme inhibitors. They inhibit acid phosphatase and dehydrogenase activity, applying their functions to the secretory pathways and the respiratory chain, respectively.

3.4.1. Brefeldin A

Penicillium species isolated from Annella sea fan yielded Brefeldin A (compound 1). The compound (C16H24O4) is a macrolide with a molecular mass of 280.36 g/mol and possesses antibiotic properties [26,27]. The pathogenic fungus M. gypseum SH-MU-4 has shown a high susceptibility to compound 1, exhibiting a minimum inhibitory concentration value of 228.57 mM [101]. The study of the mode of action has revealed a dose-dependent inhibition of the cell-surface enzyme acid phosphatase (APase) in the periplasm of C. albicans, leading to intracellular accumulations of enzyme protein. Cells grown in the presence of compound 1 became denser than those grown in the absence of the active compound. It has been concluded that fungal cell-surface growth was also blocked by treatments containing compound 18. The APase that was accumulated intracellularly migrated faster on SDS-PAGE, suggesting less N-linked glycosylation compared with the mature periplasmic APase produced in the absence of BFA. Pulse-chase experiments and gel-filtration of oligosaccharides released by Endo H treatment suggested that the core-glycosylated precursor form of APase accumulated in the presence of BFA [27].

3.4.2. Roridin A

Roridin A (compound 2) from Myrotheeium roridum and Fusarium sp. is a sesquiterpene mycotoxin (12, 13-epoxy-trichothec-9-ene moiety) developed by these microorganisms with a molecular mass of 532.6 g/mol [102,103]. To obtain the compound, organic solvent extraction is used by macerating Myrothecium sp. inside ethyl acetate-ethanol (EtOAc). The producing symbiont is isolated from the marine sponge Axinella sp. The EtOAc symbiont extract has inhibited the growth of S. sclerotiorum, S. cerevisiae, and M. grisea. After separation by silica gel column fractionation and high-performance liquid chromatography (HLPC), roridin A and trichothecenes have been purified. The inhibitory concentrations of the compounds revealed MIC values close to those of fluconazole [29]. The bioactive agent showed antifungal activity against S. cerevisiae, M. grisea, and S. sclerotiorum with minimum inhibitory concentrations (MIC) of 31.25, 125, and 31.25 μg/mL, respectively. In vitro antifungal tests showed that the purified fractions were active against A. niger, T. rubrum, and C. albicans with MICs of 31.25, 62.5, and 125 μg/mL, respectively [30]. Compound 2 was able to inhibit the dehydrogenase activity of S. cerevisiae by up to 1 µg/mL. Furthermore, preliminary attention to the structure–fungitoxicity relationship of roridin A, whose MICs are comparable to ketoconazole, allowed a temporary observation that the presence of a hydroxyl group at C-13, as well as the ether bondage between C-5 and C-13, could increase almost equally the fungicidal action against A. niger and T. rubrum [30].

3.5. Resistance Factors

Multidrug resistance factors evolve from drug efficacy failure, which is characterized by the development of tolerance to a range of drugs. There are also natural resistance factors, such as the development of spores when the fungus enters the nutritive restriction stage of its life. Modiolide A from Paraphaeosphaeria sp. and Fumiquinazoline A (compound 4) from A. fumigatus were able to thwart fungal resistance.

3.5.1. Modiolide A

Modiolide A (compound 3) is produced by a fungus, Paraphaeosphaeria sp. (N-119), isolated from a marine horse mussel. The organic natural product appears as a colorless oil with a molecular mass of 198.22 g/mol and a chemical structure determined using spectroscopy. Compound 3 exhibited a wide antimicrobial potential against N. crassa, a pathogenic fungus, with a MIC value of 3 µg/mL, and against the bacteria Micrococcus luteus with an inhibition concentration of up to 16.7 µg/mL as MIC [32]. Moreover, it impairs P. capsici by destroying 100% of its zoospore motility within 30 min at a concentration of 0.5 μg/mL. Its zoosporicidal potency is probably due to the presence of epoxide-containing cyclic lactones, which may be responsible for the inhibitory and zoosporicidal activities [104]. These spores, which can locate the host using their infectious propagules and accumulate at the sites of the infection through particular chemical signals, highly contribute to the pathogenic success of the Phytophthora genus. Moreover, any impairment of the spore motility greatly reduces its pathogenesis [105,106].

3.5.2. Fumiquinazoline

Fumiquinazoline A (compound 21; 445.5 g/mol) originated from the pathogenic fungus A. fumigatus, located at the gastrointestinal tract as endosymbionts of the fish Pseudolabrus japonicas. The mycelia of the fungus were cultured, and the compound was extracted. It was shown that compound 21 possesses cytotoxic activity against P388 lymphocytic leukemia cells [35]. The natural product was also capable of expressing antifungal activities against phytopathogenic fungi such as B. cinerea, A. solani, A. alternata (Fries) Keissler, C. gloeosporioides, F. solani, F. oxysporum f. sp. Niveum, F. oxysporum f. sp. Vasinfectum, and Gibberella saubinettii with MIC values ranging from 12.5–50 μg/mL. It is worth pointing out that the activity was more pronounced toward B. cinerea, A. solani, A. alternata, and G. saubinettii than toward Fusarium species. In addition, compound 21 highlighted an antifeedant activity with a moderate antifeedant index (AFI) of 45%. Moreover, fumiquinazoline A showed moderate activity with an AFI of 45.0%. However, when structural modifications occur on the active compound by inserting one hydroxyl group into C-3 to produce 3-hydroxyfumiquinazoline A, the antifeedant activity has considerably decreased, up to an AFI of 7.5 [34]. The structures of fumiquinazolines are quinazolinones fused with a simple piperazine ring system or more complex spiro moieties. The fused rings, the basic amine, and the rich stereochemistry are structural features useful for structure-activity relationship studies [36].

3.6. Cell Wall

The fungal cell wall is the first point of contact that initiates infection. The synthesis of cellular fungal components and the surface proteins represent capital targets to develop more efficient therapeutics or vaccines [107].

Chitinase 34 kDa

Chitinase 34 kDa (compound 5) is an enzyme extracted from the bacterial strain DA11 identified as Streptomyces sp. by 16S rDNA sequencing (GenBank accession no. DQ180128). The bacterial strain grows as a symbiont on the sponge C. austrialiensis [37]. A volume of 20 μL of the compound has been tested for its eventual antifungal activity against A. niger and C. albicans. After 120 h of incubation, an inhibition zone as a circle around compound 5 containing discs was identified, characterizing the inhibition surface of the fungus growth. The antifungal inhibition diameters were 10.98 ± 0.49 mm for A. niger and 10.48 ± 0.45 mm for C. albicans. The obtained results demonstrate the antifungal activity of the purified chitinase, which acts by degrading chitin from the cell wall. Potential uses of chitinases as biocontrol agents have been reported [108]. Chitinases from S. griseus have also shown antifungal activity against Aspergillus sp., Phycomyces blakesleeanus, Trichoderma reesei, A. nidulans, B. cinerea, Fusarium culmorum, Gulgnardia bidwellii, and Sclerotia sclerotiorum. The enzyme remains active at 85% within temperatures ranging from 30 °C to 45 °C. These results were comparable to those obtained from a chitinase isolated from a novel marine sediment Streptomyces strain, which exhibited activity at an interval temperature of 20 °C to 65 °C. For this last example, the best activity was noticed between 45 °C and 50 °C [109]. Moreover, it is also known that pH influences ionizations at the catalytic site. In that way, for some other Streptomyces species, the optimum chitinase activity was found to be included within a pH interval of 3.3 to 7.5 [110]. So, enzymatic functional features show that the extracted marine chitinase can be active under basic conditions because of its high pH tolerance and a maximum salinity tolerance of 45 g‰ psu. The latter justifies one of the characteristics of marine microorganisms, which is to tolerate salt in their living environment [111].

3.7. Compounds with Unknown Targets

The remaining compounds 1848, tambjamine C, tambjamine E, roridin D, violacein, tambjamine F, theonegramide G, theonegramide, tambjamine H, tambjamine I, tambjamine J, LL-Z1640-2, 1-hydroxy-6-methyl-8-(hydroxymethyl)xanthone, xestodecalactone B, peniciadametizine A, caerulomycin A, ND, 87.12 kDa active protein, 1-hydroxy-6-methyl-8-(hydroxylmethyl)xanthone, modiolide B, (3S)-(3,5-dihydroxyphenyl)butan-2-one, (3R,4S)-hydroxymellein, 3R)-6-methoxy-7-chloromellein, (3R)-6-methoxymellein, 4,8-dihydroxy-3,4-dihydro-2H-naphthalen-1-one, (3R,4R)-hydroxymellein, (R)-mellein, seragikinone A, microsphaeropsin, (3S)-(3,5-dihydroxyphenyl)butan-2-one, xestolactone B, and resorcylic acid lactones “zeaenol”, were recorded as derivating from microbial symbionts associated with aquatic animals. However, their antifungal targets were still not properly determined. Among these compounds, tambjamines (F, G, H–J) (compounds 22, 23, 2527) were obtained from P. tunicata on bryozoans and some other marine animals, showing antifungal activity against M. furfur (0.1–1 mg/mL) and C. albicans (1 mg/mL). LL-Z1640-2 (compound 28) has exhibited an inhibiting potential at a MIC = 0.39 μmol/L, 25-fold stronger than that of the positive control ketoconazole against the fungal pathogen P. calabae. The compound also impaired the growth of P. viticola and P. infestans. Peniciadametizine A (compound 31), which originated from the P. adametzioides AS-53a fungus found on an unidentified marine sponge, was able to inhibit A. brassicae (MIC: 4.0 µg/mL). In general, the antifungal mechanism of these compounds remains undefined, opening new investigative opportunities for the identification of their cell targets to eventually find new antifungal mechanisms of action [13,24,32,71,74,75,76,77,79,80,81,82,83,84,85,86,87,88,89,112,113].

4. Relevant Information for Future Research

Six main classes of antifungal drugs are currently available for the treatment of fungal infections. They include polyenes that act on membranes by binding ergosterol and azoles that lead to the accumulation of 14-methyl-sterol. Echinochandines target β-1,3-glucane on the cell wall, allylamines inhibit epoxydase, and griseofulvine acts on microtubule polymerization [114]. Some of the following identified compounds in the study highlighted novel insights into antifungal activity targets. In the study, three new targets were suggested. Fungal cellular enzymes such as acid phosphatase, GTPase, and dehydrogenase are targeted by Brefeldin A and Roridin A (compounds 1 and 2). Moreover, modiolide A (compound 3) acting on resistance factors, such as the fungal spore motility of P. capsici zoospores [32], constitutes a leading opportunity for developing therapeutics to specifically inhibit resistance factors. Finally, immunomodulation and apoptosis pathways were impaired by indole-3-carboxaldehyde and isatin (compounds 14 and 15), which targeted immune cells as an immunomodulator in the vulvocandidiasis model and as an apoptosis enhancer on Candida and Aspergillus microorganisms. To the best of our knowledge, there are not yet conventional antifungal drugs targeting these three targets: cellular enzymes, resistance factors, and the immune/apoptotic pathway. However, the challenge behind this resides in the fact that these fungal eukaryotic cells sometimes share the same cellular features as human eukaryotic cells, such as cellular enzymes, the immune system, and apoptosis. For example, apoptosis biochemical mechanisms are also integrated into human cells’ metabolic pathways [95,96,115]. Unless a specific antifungal therapy is only for cancer, people acting on apoptosis should be considered. So, deep investigations through preclinical and clinical trials involving toxicity studies may produce effective results in the development of new antifungal drugs with novel cell targets from aquatic sources. Actually, from these reports, spore motility seems to represent the best suggested novel antifungal target to investigate. Even the combination of these compounds with different and new cell targets may lead to promising effects.
The multiple advantages of the combination approach have already been shown [116,117,118]. This approach exploits various beneficial characteristics associated with each combination product for the improvement of the effectiveness, the reduction of toxicity, the reduction of the doses to be administered, and the reduction of the features associated with the development of drug resistance [119]. In addition, this approach would involve a greater diversity of secondary metabolites in antimicrobial action. Combinations could therefore possibly target several metabolic pathways at once, thereby enhancing the desired therapeutic effect, most likely through synergistic interactions [120]. That is also the case with diacetylphloroglucinol (compound 16), which acts at the same time on the mitochondrial membrane and causes hyphal disorganization. In addition, tyrosol (compound 17) affects cell membrane permeability and inhibits the biofilm on Candida at the same time to produce the inhibition activity.

5. Conclusions

The present study gives insights into how to learn from the microbial symbiosis associated with aquatic animals and develop mimicry strategies for the development of infectious disease treatments, particularly in the field of antifungals. Natural products derived from such symbiotic interactions may be mimicked, or the physical behavior of some fungi to protect the host could also be integrated into the antimicrobial development strategies. One example of chemically mediated defense from a microbial symbiont to protect its host is the case of P. macrodactylus embryos (crustacean), which defend themselves through the action of a symbiont bacteria, Alteromonas sp., to fight the fungal infection caused by L. callinectes on its embryos [62] by producing isatin, an antimicrobial agent to impair the progression of the infection, where close derivative isatin compounds act by activating the pathogen apoptosis pathway. Thus, three main targets to investigate in preclinical and clinical trials have been identified as potent antifungal cell targets as well as antifungal agents in cancer therapies. Besides, it is worth noting the presence of the identified antifungal targets here in this study as acid phosphatases, GTPases, and apoptosis, which are also involved in some cancer pathologies. Some of the mentioned compounds with apoptotic effects could also be involved in the drug discovery process of metabolic diseases because the same pathogenesis pathway as apoptosis activation is highlighted in cancer (compounds 1, 2, 7, 15, 21, 26, and 27).

Author Contributions

Conceptualization, M.N.L.N.-M.; supervision; H.G.M., writing—review and editing, E.Z.M., M.N.L.N.-M. and H.G.M.; writing—original draft, M.N.L.N.-M.; visualization, E.Z.M., M.N.L.N.-M. and H.G.M. All authors have read and agreed to the published version of the manuscript.

Funding

Partial financial support for M.N.L.N.-M. is acknowledged: funding from The African-German Network of Excellence in Science (AGNES). Partial financial support for H.G.M. is acknowledged: funding from the National Research Foundation of South Africa grant number 109204 [IFR170221223270] and the University of Cape Town (URC grant).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All studies from which data are presented are cited and referenced in the manuscript.

Acknowledgments

The authors gratefully acknowledge the support from The Africa German Network of Excellence in Science (AGNES) for the realization of this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brown, G.D.; Denning, D.W.; Gow, N.A.R.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden killers: Human fungal infections. Sci. Transl. Med. 2012, 4, 165rv13. [Google Scholar] [CrossRef]
  2. Fausto, A.; Rodrigues, M.L.; Coelho, C. The still underestimated problem of fungal diseases worldwide. Front. Microbiol. 2019, 10, 214. [Google Scholar]
  3. Fisher, M.C.; Henk, D.A.; Briggs, C.J.; Brownstein, J.S.; Madoff, L.C.; McCraw, S.L.; Gurr, S.J. Emerging fungal threats to animal, plant and ecosystem health. Nature 2012, 484, 186–194. [Google Scholar] [CrossRef] [PubMed]
  4. Seipke, R.F.; Grüschow, S.; Goss, R.J.M.; Hutchings, M.I. Isolating antifungals from fungus-growing ant symbionts using a genome-guided chemistry approach. Methods Enzymol. 2012, 517, 47–70. [Google Scholar] [PubMed]
  5. Chemaly, R.F. The ever-growing world of infections in immunocompromised patients: Major headways with numerous shortcomings. Clin. Microbiol. Infect. 2021, 27, 1379–1380. [Google Scholar] [CrossRef]
  6. Wall, G.; Lopez-Ribot, J.L. Current Antimycotics, New Prospects, and Future Approaches to Antifungal Therapy. Antibiotics 2020, 9, 445. [Google Scholar] [CrossRef]
  7. Gogineni, V.; Chen, X.; Hanna, G.; Mayasari, D.; Hamann, M.T. Role of symbiosis in the discovery of novel antibiotics. J. Antibiot. 2020, 73, 490–503. [Google Scholar] [CrossRef]
  8. Barke, J.; Seipke, R.F.; Grüschow, S.; Heavens, D.; Drou, N.; Bibb, M.J.; Goss, R.J.M.; Yu, D.W.; Hutchings, M.I. A mixed community of actinomycetes produce multiple antibiotics for the fungus farming ant Acromyrmex octospinosus. BMC Biol. 2010, 8, 109. [Google Scholar] [CrossRef]
  9. Haeder, S.; Wirth, R.; Herz, H.; Spiteller, D. Candicidin-producing Streptomyces support leaf-cutting ants to protect their fungus garden against the pathogenic fungus Escovopsis. Proc. Natl. Acad. Sci. USA 2009, 106, 4742. [Google Scholar] [CrossRef]
  10. Seipke, R.F.; Barke, J.; Brearley, C.; Hill, L.; Yu, D.W.; Goss, R.J.M.; Hutchings, M.I. A single Streptomyces symbiont makes multiple antifungals to support the fungus farming ant acromyrmex octospinosus. PLoS ONE 2011, 6, e22028. [Google Scholar] [CrossRef]
  11. Kolachana, P.; Smith, M.T. Induction of kinetochore-positive micronuclei in human lymphocytes by the anti-fungal drug griseofulvin. Mutat. Res. Toxicol. 1994, 322, 151–159. [Google Scholar] [CrossRef] [PubMed]
  12. Goldman, L.; Schwarz, J.; Preston, R.H.; Beyer, A.; Loutzenhiser, J. Current Status of Griseofulvin: Report on One Hundred Seventy-Five Cases. J. Am. Med. Assoc. 1960, 172, 532–538. [Google Scholar] [CrossRef]
  13. Thomas, T.R.A.; Kavlekar, D.P.; LokaBharathi, P.A. Marine drugs from sponge-microbe association—A review. Mar. Drugs 2010, 8, 1417–1468. [Google Scholar] [CrossRef] [PubMed]
  14. Bollmann, M.; Bosch, T.; Cau, K.; Colijn, F.; Cau, K.; Ebinghaus, R.; Gkss, R.C.; Körtzinger, A.; Leibniz, I. Living With the Oceans—World ocean review. World Ocean Rev. 2010, 1, 236. [Google Scholar]
  15. Grosberg, R.K.; Vermeij, G.J.; Wainwright, P.C. Biodiversity in water and on land. Curr. Biol. 2012, 22, R900–R903. [Google Scholar] [CrossRef]
  16. Vaughan, T.A.; Ryan, J.M.; Czaplewski, N.J. Mammalogy, 6th ed.; Jones & Bartlett Learning: Burlington, VT, USA, 2013; p. 755. [Google Scholar]
  17. Tarazona, A.M.; Ceballos, M.C.; Broom, D.M. Animals: One Health, One Welfare, One Biology. Animals 2020, 10, 1516. [Google Scholar]
  18. Hart, B.L. Behavioural defences in animals against pathogens and parasites: Parallels with the pillars of medicine in humans. Philos. Trans. R. Soc. B Biol. Sci. 2011, 366, 3406. [Google Scholar] [CrossRef]
  19. Tiralongo, F.; Messina, G.; Lombardo, B.M.; Longhitano, L.; Li Volti, G.; Tibullo, D. Skin Mucus of Marine Fish as a Source for the Development of Antimicrobial Agents. Front. Mar. Sci. 2020, 7, 541853. [Google Scholar] [CrossRef]
  20. Vennila, R.; Rajesh Kumar, K.; Kanchana, S.; Arumugam, M.; Vijayalakshmi, S.; Balasubramaniam, T. Preliminary investigation on antimicrobial and proteolytic property of the epidermal mucus secretion of marine stingrays. Asian Pac. J. Trop. Biomed. 2011, 1, S239–S243. [Google Scholar] [CrossRef]
  21. Becker, M.H.; Walke, J.B.; Cikanek, S.; Savage, A.E.; Mattheus, N.; Santiago, C.N.; Minbiole, K.P.C.; Harris, R.N.; Belden, L.K.; Gratwicke, B. Composition of symbiotic bacteria predicts survival in Panamanian golden frogs infected with a lethal fungus. Proc. R. Soc. B Biol. Sci. 2015, 282, 20142881. [Google Scholar] [CrossRef]
  22. Lowrey, L.; Woodhams, D.C.; Tacchi, L.; Salinas, I. Topographical mapping of the rainbow trout (Oncorhynchus mykiss) microbiome reveals a diverse bacterial community with antifungal properties in the skin. Appl. Environ. Microbiol. 2015, 81, 6915–6925. [Google Scholar] [CrossRef] [PubMed]
  23. Reverter, M.; Tapissier-Bontemps, N.; Lecchini, D.; Banaigs, B.; Sasal, P. Biological and ecological roles of external fish mucus: A review. Fishes 2018, 3, 41. [Google Scholar] [CrossRef]
  24. Florez, L.V.; Biedermann, P.H.W.; Engl, T.; Kaltenpoth, M. Defensive symbioses of animals with prokaryotic and eukaryotic microorganisms. Nat. Prod. Rep. 2015, 32, 879–1156. [Google Scholar] [CrossRef] [PubMed]
  25. Schmidt, E.W. Trading molecules and tracking targets in symbiotic interactions. Nat. Chem. Biol. 2008, 4, 466–473. [Google Scholar] [CrossRef] [PubMed]
  26. Pandey, A. Pharmacological significance of marine microbial bioactive compounds. Environ. Chem. Lett. 2019, 17, 1741–1751. [Google Scholar] [CrossRef]
  27. Arioka, M.; Hirata, A.; Takatsuki, A.; Yamasaki, M. Brefeldin A blocks an early stage of protein transport in Candida albicans. J. Gen. Microbiol. 1991, 137, 1253–1262. [Google Scholar] [CrossRef]
  28. Johns, A. Towards the Development of Novel Aspergillus Fumigatus Targeted Antifungals with an In-Depth Analysis of Sfp-PPTase, PptA [Internet]. 2015. Available online: http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.684773 (accessed on 31 May 2022).
  29. Xie, L.W.; Jiang, S.M.; Zhu, H.H.; Sun, W.; Ouyang, Y.C.; Dai, S.K.; Li, X. Potential inhibitors against Sclerotinia sclerotiorum, produced by the fungus Myrothecium sp. associated with the marine sponge Axinella sp. Eur. J. Plant Pathol. 2008, 122, 571–578. [Google Scholar] [CrossRef]
  30. Liu, J.Y.; Huang, L.L.; Ye, Y.H.; Zou, W.X.; Guo, Z.J.; Tan, R.X. Antifungal and new metabolites of Myrothecium sp. Z16, a fungus associated with white croaker Argyrosomus argentatus. J. Appl. Microbiol. 2006, 100, 195–202. [Google Scholar] [CrossRef]
  31. Reiss, J. Influence of Fusarium and Myrothecium mycotoxins on dehydrogenase activity of Saccharomyces cerevisiae. Mycopathologia 1983, 81, 187–189. [Google Scholar] [CrossRef]
  32. Tsuda, M.; Mugishima, T.; Komatsu, K.; Sone, T.; Tanaka, M.; Mikami, Y.; Kobayashi, J. Modiolides A and B, two new 10-membered macrolides from a marine-derived fungus. J. Nat. Prod. 2003, 66, 412–415. [Google Scholar] [CrossRef]
  33. Bajpai, V.K. Antimicrobial secondary metabolites from marine fungi: A mini review. Indian J. Mar. Sci. 2016, 45, 1067–1075. [Google Scholar]
  34. Li, X.; Zhang, Q.; Zhang, A.; Gao, J. Metabolites from Aspergillus an endophytic fungus associated with Melia azedarach, and Their Antifungal, Antifeedant and Toxic Activities. J. Agric. Food Chem. 2012, 60, 3424–3431. [Google Scholar] [CrossRef] [PubMed]
  35. Kobayashi, J.; Ishibashi, M. Bioactive Metabolites of Symbiotic Marine Microorganisms. Chem. Rev. 1993, 93, 1753–1769. [Google Scholar] [CrossRef]
  36. Resende, D.I.S.P.; Boonpothong, P.; Sousa, E.; Kijjoa, A.; Pinto, M.M.M. Chemistry of the fumiquinazolines and structurally related alkaloids. Nat. Prod. Rep. 2019, 36, 7–34. [Google Scholar] [CrossRef]
  37. Han, Y.; Yang, B.; Zhang, F.; Miao, X.; Li, Z. Characterization of antifungal chitinase from marine Streptomyces sp. DA11 associated with South China sea sponge Craniella australiensis. Mar. Biotechnol. 2009, 11, 132–140. [Google Scholar] [CrossRef]
  38. Dunlap, W.C.; Battershill, C.N.; Liptrot, C.H.; Cobb, R.E.; Bourne, D.G.; Jaspars, M.; Long, P.F.; Newman, D.J. Biomedicinals from the phytosymbionts of marine invertebrates: A molecular approach. Methods 2007, 42, 358–376. [Google Scholar] [CrossRef]
  39. Williams, D.E.; Burgoyne, D.L.; Rettig, S.J. The Isolation of Majusculamide C from the Sponge of the 2-Methyl-3-Aminopentanoic. J. Nat. Prod. 1993, 56, 545–551. [Google Scholar] [CrossRef]
  40. Niedermeyer, T.H.J. Anti-infective Natural Products from Cyanobacteria. Planta Med. 2015, 81, 1309–1325. [Google Scholar] [CrossRef]
  41. Moore, R.E. Cyclic peptides and depsipeptides from cyanobacteria: A review. J. Ind. Microbiol. 1996, 16, 134–143. [Google Scholar] [CrossRef]
  42. Jiang, J.; Gao, L.; Bie, X.; Lu, Z.; Liu, H.; Zhang, C.; Lu, F.; Zhao, H. Identification of novel surfactin derivatives from NRPS modification of Bacillus subtilis and its antifungal activity against Fusarium moniliforme. BMC Microbiol. 2016, 16, 31. [Google Scholar] [CrossRef]
  43. Rodrigues, L.; Banat, I.M.; Teixeira, J.; Oliveira, R. Biosurfactants: Potential applications in medicine. J. Antimicrob. Chemother. 2006, 57, 609–618. [Google Scholar] [CrossRef] [PubMed]
  44. Mongkolthanaruk, W. Classification of bacillus beneficial substances related to plants, humans and animals. J. Microbiol. Biotechnol. 2012, 22, 1597–1604. [Google Scholar] [CrossRef] [PubMed]
  45. Niekawa, E.T.G.; Simionato, A.S.; Barazetti, A.R.; Cano, B.G.; Emiliano, J.; Afonso, L.; de Lima Andreata, M.F.; Dealis, M.L.; Chryssafidis, A.L.; Andrade, G. The microbial role in the control of phytopathogens—An alternative to agrochemicals. In Microbiome Stimulants for Crops; Woodhead Publishing: Cambridge, UK, 2021; pp. 159–177. [Google Scholar] [CrossRef]
  46. Parray, J.A.; Shameem, N. Metabolomics and microbial biocontrol agents. In Sustainable Agriculture; Academic Press: Cambridge, MA, USA, 2020; pp. 181–229. [Google Scholar] [CrossRef]
  47. Elyakov, G.B.; Kuznetsova, T.; Mikhailov, V.V.; Maltsev, I.I.; Voinov, V.G.; Fedoreyev, S.A. Brominated diphenyl ethers from a marine bacterium associated with the sponge Dysidea sp. Experientia 1991, 47, 632–633. [Google Scholar] [CrossRef]
  48. Sionov, E.; Roth, D.; Sandovsky-Losica, H.; Kashman, Y.; Rudi, A.; Chill, L.; Berdicevsky, I.; Segal, E. Antifungal effect and possible mode of activity of a compound from the marine sponge Dysidea herbacea. J. Infect. 2005, 50, 453–460. [Google Scholar] [CrossRef] [PubMed]
  49. Nagai, K.; Kamigiri, K.; Matsumoto, H.; Kawano, Y.; Yamaoka, M.; Shimoi, H.; Watanabe, M.; Suzuki, K. YM-202204, a new antifungal antibiotic produced by marine fungus Phoma sp. J. Antibiot. 2002, 55, 1036–1041. [Google Scholar] [CrossRef] [PubMed]
  50. Matsunaga, S.; Fusetani, N.; Hashimoto, V.K.; Walchlit, M.; Theonellamide, F. A Novel Antifungal Bicyclic Peptide from a Marine Sponge Theonella sp. J. Am. Chem. Soc. 1989, 2, 2582–2588. [Google Scholar] [CrossRef]
  51. Nishimura, S.; Arita, Y.; Honda, M.; Iwamoto, K.; Matsuyama, A.; Shirai, A.; Kawasaki, H.; Kakeya, H.; Kobayashi, T.; Matsunaga, S.; et al. Marine antifungal theonellamides target 3β 2-hydroxysterol to activate Rho1 signaling. Nat. Chem. Biol. 2010, 6, 519–526. [Google Scholar] [CrossRef] [PubMed]
  52. Yadav, S. Synthesis of the Western Hemisphere of Theonellamide C. The Graduate School at LSU Digital Commons, Editor. 2013. Available online: https://digitalcommons.lsu.edu/gradschool_dissertations/2836 (accessed on 3 February 2023).
  53. Schmidt, E.W.; Obraztsova, A.Y.; Davidson, S.K.; Faulkner, D.J.; Haygood, M.G. Identification of the antifungal peptide-containing symbiont of the marine sponge Theonella swinhoei as a novel δ-proteobacterium, “Candidatus Entotheonella palauensis”. Mar. Biol. 2000, 136, 969–977. [Google Scholar] [CrossRef]
  54. Piel, J. Metabolites from symbiotic bacteria. Nat. Prod. Rep. 2004, 26, 338–362. [Google Scholar] [CrossRef]
  55. Ho, C.H.; Magtanong, L.; Barker, S.L.; Gresham, D.; Nishimura, S.; Natarajan, P.; Koh, J.L.Y.; Porter, J.; Gray, C.A.; Andersen, R.J.; et al. A molecular barcoded yeast ORF library enables mode-of-action analysis of bioactive compounds. Nat. Biotechnol. 2009, 27, 369–377. [Google Scholar] [CrossRef]
  56. Ho, C.H.; Piotrowski, J.; Dixon, S.J.; Baryshnikova, A.; Costanzo, M.; Boone, C. Combining functional genomics and chemical biology to identify targets of bioactive compounds. Curr. Opin. Chem. Biol. 2011, 15, 66–78. [Google Scholar] [CrossRef] [PubMed]
  57. Schmidt, E.W.; Bewley, C.A.; Faulkner, D.J. Theopalauamide, a Bicyclic Glycopeptide from Filamentous Bacterial Symbionts of the Lithistid Sponge Theonella swinhoei from Palau and Mozambique. J. Org. Chem. 1998, 63, 1254–1258. [Google Scholar] [CrossRef]
  58. Brucker, R.M.; Harris, R.N.; Schwantes, C.R.; Gallaher, T.N.; Flaherty, D.C.; Lam, B.A.; Minbiole, K.P.C. Amphibian Chemical Defense: Antifungal Metabolites of the Microsymbiont Janthinobacterium lividum on the Salamander Plethodon cinereus. J. Chem. Ecol. 2008, 34, 1422–1429. [Google Scholar] [CrossRef] [PubMed]
  59. Borghi, M.; Pariano, M.; Solito, V.; Puccetti, M.; Bellet, M.M.; Stincardini, C.; Renga, G.; Vacca, C.; Sellitto, F.; Mosci, P.; et al. Targeting the Aryl Hydrocarbon Receptor With Indole-3-Aldehyde Protects From Vulvovaginal Candidiasis via the IL-22-IL-18 Cross-Talk. Front. Immunol. 2019, 10, 02364. [Google Scholar] [CrossRef] [PubMed]
  60. Mirjana, K.; Marica, M.-S.; Marija, M.; Dusica, M. Antiviral, antibacterial, and antifungal activities of isatin N-mannich bases. J. Pharm. Sci. 1975, 64, 881–882. [Google Scholar]
  61. Chohan, Z.H.; Rauf, A.; Khan, K.M.; Supuran, C.T. Isatin-derived Antibacterial and Antifungal Compounds and their Transition Isatin-derived Antibacterial and Antifungal Compounds and their Transition Metal Complexes. J. Enzym. Inhib. Med. Chem. 2004, 417–423. [Google Scholar] [CrossRef]
  62. Gil-Turnes, M.S.; Hay, M.E.; Fenical, W. Symbiotic marine bacteria chemically defend crustacean embryos from a pathogenic fungus. Science 1989, 246, 116–118. [Google Scholar] [CrossRef]
  63. Dar, O.A.; Lone, S.A.; Malik, M.A.; Aqlan, F.M.; Wani, M.Y.; Hashmi, A.A.; Ahmad, A. Synthesis and synergistic studies of isatin based mixed ligand complexes as potential antifungal therapeutic agents. Heliyon 2019, 5, 02055. [Google Scholar] [CrossRef]
  64. Brucker, R.M.; Baylor, C.M.; Walters, R.L.; Lauer, A.; Harris, R.N.; Minbiole, K.P.C. The Identification of 2, 4-diacetylphloroglucinol as an Antifungal Metabolite Produced by Cutaneous Bacteria of the Salamander Plethodon cinereus. J. Chem. Ecol. 2008, 34, 39–43. [Google Scholar] [CrossRef]
  65. De Souza, J.T.; Arnould, C.; Deulvot, C.; Lemanceau, P.; Gianinazzi-Pearson, V.; Raaijmakers, J.M. Effect of 2,4-diacetylphloroglucinol on Pythium: Cellular responses and variation in sensitivity among propagules and species. Phytopathology 2003, 93, 966–975. [Google Scholar] [CrossRef]
  66. Troppens, D.M.; Dmitriev, R.I.; Papkovsky, D.B.; O’Gara, F.; Morrissey, J.P. Genome-wide investigation of cellular targets and mode of action of the antifungal bacterial metabolite 2,4-diacetylphloroglucinol in Saccharomyces cerevisiae. FEMS Yeast Res. 2013, 13, 322–334. [Google Scholar] [CrossRef] [PubMed]
  67. Gil-Turnes, M.S.; Fenical, W. Embryos of Homarus americanus are protected by epibiotic bacteria. Biol. Bull. 1992, 182, 105–108. [Google Scholar] [CrossRef] [PubMed]
  68. Monteiro, D.R.; Arias, L.S.; Fernades, A.R.; Da Silva, L.F.D.; De Castilho, M.O.V.F.; Da Rosa, T.O.; Vieira, A.P.M.; Straioto, F.G.; Barbosa, D.B.; Delbem, A.C.B. Antifungal activity of tyrosol and farnesol used in combination against Candida species in the planktonic state or forming biofilms. J. Appl. Microbiol. 2017, 123, 392–400. [Google Scholar] [CrossRef] [PubMed]
  69. Brilhante, R.S.N.; Caetano, É.P.; de Lima, R.A.C.; Marques, F.J.d.F.; Castelo-Branco, D.d.S.C.M.; Melo, C.V.S.d.; Guedes, G.M.d.M.; Oliveira, J.S.d.; Camargo, Z.P.d.; Moreira, J.L.B.; et al. Terpinen-4-ol, tyrosol, and β-lapachone as potential antifungals against dimorphic fungi. Braz. J. Microbiol. 2016, 47, 917–924. [Google Scholar] [CrossRef] [PubMed]
  70. Sebaa, S.; Boucherit-Otmani, Z.; Courtois, P. Effects of tyrosol and farnesol on Candida albicans biofilm. Mol. Med. Rep. 2019, 19, 3201–3209. [Google Scholar] [CrossRef]
  71. Franks, A.; Haywood, P.; Egan, S.; Kjelleberg, S.; Kumar, N.; Sciences, B.; Wales, S. Isolation and Structure Elucidation of a Novel Yellow Pigment from the Marine Bacterium. Molecules 2005, 10, 1286–1291. [Google Scholar] [CrossRef]
  72. Sasidharan, A.; Sasidharan, N.K.; Amma, D.B.N.S.; Vasu, R.K.; Nataraja, A.V.; Bhaskaran, K. Antifungal activity of violacein purified from a novel strain of Chromobacterium sp. NIIST (MTCC 5522). J. Microbiol. 2015, 53, 694–701. [Google Scholar] [CrossRef]
  73. Choi, S.Y.; Yoon, K.H.; Lee, J.I.; Mitchell, R.J. Violacein: Properties and production of a versatile bacterial pigment. BioMed Res. Int. 2015, 2015, 465056. [Google Scholar] [CrossRef]
  74. Bewley, C.A.; Holland, N.D.; Faulker, D.J. Two classes of metabolites from Theonella swinhoei are localized in distinct populations of bacterial symbionts. Experientia 1996, 52, 716–722. [Google Scholar] [CrossRef]
  75. Fernández de Ullivarri, M.; Arbulu, S.; Garcia-Gutierrez, E.; Cotter, P.D. Antifungal Peptides as Therapeutic Agents. Front. Cell. Infect. Microbiol. 2020, 10, 105. [Google Scholar] [CrossRef]
  76. Liu, Q.; Shao, C.; Gu, Y.; Blum, M.; Gan, L.; Wang, K.; Chen, M.; Wang, C. Antifouling and Fungicidal Resorcylic Acid Lactones from the Sea Anemone-Derived Fungus Cochliobolus lunatus. J. Agric. Food Chem. 2014, 62, 3183–3191. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, L.; Zheng, Y.Y.; Shao, C.L.; Wang, C.Y. Metabolites from marine invertebrates and their symbiotic microorganisms: Molecular diversity discovery, mining, and application. Mar. Life Sci. Technol. 2019, 1, 60–94. [Google Scholar] [CrossRef]
  78. Höller, U.; König, G.M.; Wright, A.D. Three new metabolites from marine-derived fungi of the genera coniothyrium and microsphaeropsis. J. Nat. Prod. 1999, 62, 114–118. [Google Scholar] [CrossRef] [PubMed]
  79. König, G.M.; Kehraus, S.; Seibert, S.F.; Abdel-Lateff, A.; Müller, D. Natural products from marine organisms and their associated microbes. ChemBioChem 2006, 7, 229–238. [Google Scholar] [CrossRef] [PubMed]
  80. Edrada, R.A.; Heubes, M.; Brauers, G.; Wray, V.; Berg, A.; Gra, U.; Wohlfarth, M.; Schaumann, K.; Bringmann, G.; Proksch, P.; et al. Online Analysis of Xestodecalactones A–C, Novel Bioactive Metabolites from the Fungus Penicillium cf. montanense and Their Subsequent Isolation from the Sponge Xestospongia exigua. J. Nat. Prod. 2002, 2, 1598–1604. [Google Scholar] [CrossRef]
  81. El-Hossary, E.M.; Cheng, C.; Hamed, M.M.; El-Sayed Hamed, A.N.; Ohlsen, K.; Hentschel, U.; Abdelmohsen, U.R. Antifungal potential of marine natural products. Eur. J. Med. Chem. 2017, 126, 631. [Google Scholar] [CrossRef]
  82. Liu, Y.; Mándi, A.; Li, X.M.; Meng, L.H.; Kurtán, T.; Wang, B.G. Peniciadametizine A, a dithiodiketopiperazine with a unique spiro[furan-2,7′-pyrazino[1,2-b][1,2]oxazine] skeleton, and a related analogue, peniciadametizine B, from the marine sponge-derived fungus Penicillium adametzioides. Mar. Drugs 2015, 13, 3640–3652. [Google Scholar] [CrossRef]
  83. Ambavane, V.; Tokdar, P.; Parab, R.; Sreekumar, E.S.; Mahajan, G.; Mishra, P.D.; D’Souza, L.; Ranadive, P. Caerulomycin A—An Antifungal Compound Isolated from Marine Actinomycetes. Adv. Microbiol. 2014, 04, 567–578. [Google Scholar] [CrossRef]
  84. Biabani, M.A.F.; Laatsch, H. Advances in chemical studies on low-molecular weight metabolites of marine fungi. J. Prakt. Chem. Chem. Ztg. 1998, 340, 589–607. [Google Scholar] [CrossRef]
  85. Selvin, J.; Shanmughapriya, S.; Gandhimathi, R.; Seghal Kiran, G.; Rajeetha Ravji, T.; Natarajaseenivasan, K.; Hema, T.A. Optimization and production of novel antimicrobial agents from sponge associated marine actinomycetes Nocardiopsis dassonvillei MAD08. Appl. Microbiol. Biotechnol. 2009, 83, 435–445. [Google Scholar] [CrossRef]
  86. Elsebai, M.F.; Ghabbour, H.A. Isocoumarin derivatives from the marine-derived fungus Phoma sp. 135. Tetrahedron Lett. 2016, 57, 354–356. [Google Scholar] [CrossRef]
  87. Bhadury, P.; Mohammad, B.T.; Wright, P.C. The current status of natural products from marine fungi and their potential as anti-infective agents. J. Ind. Microbiol. Biotechnol. 2006, 33, 325–337. [Google Scholar] [CrossRef] [PubMed]
  88. Swathi, J.; Narendra, K.; Sowjanya, M.; Satya, K. Marine fungal metabolites as a rich source of bioactive compounds. Afr. J. Biochem. Res. 2013, 7, 184–196. [Google Scholar] [CrossRef]
  89. Blockley, A.; Elliott, D.R.; Roberts, A.P.; Sweet, M. Symbiotic microbes from marine invertebrates: Driving a new era of natural product drug discovery. Diversity 2017, 9, 49. [Google Scholar] [CrossRef]
  90. Klich, M.A.; Lax, A.R.; Bland, J.M. Inhibition of some mycotoxigenic fungi by iturin A, a peptidolipid produced by Bacillus subtilis. Mycopathologia 1991, 116, 77–80. [Google Scholar] [CrossRef] [PubMed]
  91. Meena, K.R.; Kanwar, S.S. Lipopeptides as the antifungal and antibacterial agents: Applications in food safety and therapeutics. BioMed. Res. Int. 2015, 2015, 473050. [Google Scholar] [CrossRef]
  92. Hiramatsu, F.; Miyajima, T.; Murayama, T.; Takahashi, K.; Koseki, T.; Shiono, Y. Isolation and structure elucidation of neofusapyrone from a marine-derived fusarium species, and structural revision of fusapyrone and deoxyfusapyrone. J. Antibiot. 2006, 59, 704–709. [Google Scholar] [CrossRef]
  93. Fusetani, N.; Matsunaga, S. Bioactive Sponge Peptides. Chem. Rev. 1993, 93, 1793–1806. [Google Scholar] [CrossRef]
  94. Falcone, C.; Mazzoni, C. External and internal triggers of cell death in yeast. Cell. Mol. Life Sci. 2016, 73, 2237–2250. [Google Scholar] [CrossRef] [PubMed]
  95. Wissing, S.; Ludovico, P.; Herker, E.; Büttner, S.; Engelhardt, S.M.; Decker, T.; Link, A.; Proksch, A.; Rodrigues, F.; Corte-Real, M.; et al. An AIF orthologue regulates apoptosis in yeast. J. Cell Biol. 2004, 166, 969–974. [Google Scholar] [CrossRef]
  96. Mcfall-ngai, M.J. Consequences of evolving with bacterial symbionts: Insights from the Squid-Vibrio associations. Annu. Rev. Ecol. Syst. 1999, 30, 235–256. [Google Scholar]
  97. Singh, G.S.; Desta, Z.Y. Isatins as privileged molecules in design and synthesis of spiro-fused cyclic frameworks. Chem. Rev. 2012, 112, 6104–6155. [Google Scholar] [CrossRef] [PubMed]
  98. Oxford, A.E.; Raistrick, H.; Simonart, P. Studies in the biochemistry of micro-organisms. Biochem. J. 1939, 33, 240–248. [Google Scholar] [CrossRef] [PubMed]
  99. Boothe, D.M.; Auburn University College of Veterinary Medicine. Griseofulvin—Pharmacology—MSD Veterinary Manual. 2016. Available online: https://www.msdvetmanual.com/pharmacology/antifungal-agents/griseofulvin (accessed on 14 January 2022).
  100. Théatre, A.; Cano-Prieto, C.; Bartolini, M.; Laurin, Y.; Deleu, M.; Niehren, J.; Fida, T.; Gerbinet, S.; Alanjary, M.; Medema, M.H.; et al. The Surfactin-Like Lipopeptides from Bacillus spp.: Natural Biodiversity and Synthetic Biology for a Broader Application Range. Front. Bioeng. Biotechnol. 2021, 9, 623701. [Google Scholar] [CrossRef] [PubMed]
  101. Trisuwan, K.; Rukachaisirikul, V.; Sukpondma, Y.; Phongpaichit, S.; Preedanon, S.; Sakayaroj, J. Lactone derivatives from the marine-derived fungus Penicillium sp. PSU-F44. Chem. Pharm. Bull. 2009, 57, 1100–1102. [Google Scholar] [CrossRef]
  102. Ishii, K.; Ueno, Y. Isolation and characterization of two new trichothecenes from Fusarium sporotrichioides strain M-1-1. Appl. Environ. Microbiol. 1981, 42, 541–543. [Google Scholar] [CrossRef]
  103. Abbas, H.K.; Johnson, B.B.; Shier, W.T.; Tak, H.; Jarvis, B.B.; Boyette, C.D. Phytotoxicity and mammalian cytotoxicity of macrocyclic trichothecene mycotoxins from Myrothecium verrucaria. Phytochemistry 2002, 59, 309–313. [Google Scholar] [CrossRef]
  104. Mondol, M.A.M.; Farthouse, J.; Islam, M.T.; Schüffler, A.; Laatsch, H. Metabolites from the Endophytic Fungus Curvularia sp. M12 Act as Motility Inhibitors against Phytophthora capsici Zoospores. J. Nat. Prod. 2017, 80, 347–355. [Google Scholar] [CrossRef]
  105. Horio, T.; Kawabata, Y.; Takayama, T.; Tahara, S.; Kawabata, J.; Fukushi, Y.; Nishimura, H.; Mizutani, J. A potent attractant of zoospores of Aphanomyces cochlioides isolated from its host, Spinacia oleracea. Cell. Mol. Life Sci. 1992, 48, 410–414. [Google Scholar] [CrossRef]
  106. Judelson, H.S.; Blanco, F.A. The spores of Phytophthora: Weapons of the plant destroyer. Nat. Rev. Microbiol. 2005, 3, 47–58. [Google Scholar] [CrossRef]
  107. Ibe, C.; Oladele, R.O.; Alamir, O. Our pursuit for effective antifungal agents targeting fungal cell wall components: Where are we? Int. J. Antimicrob. Agents 2022, 59, 106477. [Google Scholar] [CrossRef] [PubMed]
  108. Freeman, S.; Minz, D.; Kolesnik, I.; Barbul, O.; Zveibil, A.; Maymon, M.; Nitzani, Y.; Kirshner, B.; Rav-david, D.; Bilu, A.; et al. Trichoderma biocontrol of Colletotrichum acutatum and Botrytis cinerea and survival in strawberry. Eur. J. Plant Pathol. 2004, 110, 361–370. [Google Scholar] [CrossRef]
  109. Hoster, F.; Schmitz, J.E.; Daniel, R. Enrichment of chitinolytic microorganisms: Isolation and characterization of a chitinase exhibiting antifungal activity against phytopathogenic fungi from a novel Streptomyces strain. Appl. Microbiol. Biotechnol. 2005, 66, 434–442. [Google Scholar] [CrossRef]
  110. Gomes, R.C.; Sêmedo, L.T.A.S.; Soares, R.M.A. Purification of a thermostable endochitinase from Streptomyces RC1071 isolated from a cerrado soil and its antagonism against phytopathogenic fungi. J. Appl. Microbiol. 2001, 90, 653–661. [Google Scholar] [CrossRef] [PubMed]
  111. Moriguchi, M.; Sakai, K.; Tateyama, R.; Furuta, Y.; Wakayama, M. Isolation and characterization of salt-tolerant glutaminases from marine Micrococcus luteus K-3. J. Ferment. Bioeng. 1994, 77, 621–625. [Google Scholar] [CrossRef]
  112. Höller, U. Isolation, Biological Activity and Secondary Metabolite Investigations of Marine-Derived Fungi and Selected Host Sponges. Ph.D. Thesis, Technical University of Braunschweig, Braunschweig, Germany, 1999. [Google Scholar]
  113. Pandey, A.K.; Kumar, P.; Singh, P.; Tripathi, N.N.; Bajpai, V.K. Essential oils: Sources of antimicrobials and food preservatives. Front. Microbiol. 2017, 7, 02161. [Google Scholar] [CrossRef] [PubMed]
  114. Nett, J.E.; Andes, D.R. Antifungal Agents: Spectrum of Activity, Pharmacology, and Clinical Indications. Infect. Dis. Clin. N. Am. 2016, 30, 51–83. [Google Scholar] [CrossRef]
  115. Beesoo, R.; Neergheen-Bhujun, V.; Bhagooli, R.; Bahorun, T. Apoptosis inducing lead compounds isolated from marine organisms of potential relevance in cancer treatment. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2014, 768, 84–97. [Google Scholar] [CrossRef]
  116. Ngo Mback, M.N.L.; Agnaniet, H.; Nguimatsia, F.; Jazet Dongmo, P.-M.; Hzounda Fokou, J.-B.; Bakarnga-via, I.; Fekam Boyom, F.; Menut, C. Optimization of antifungal activity of Aeollanthus heliotropioides oliv essential oil and Time Kill Kinetic Assay. J. Mycol. Médicale J. Med. Mycol. 2016, 26, 233–243. [Google Scholar] [CrossRef]
  117. Hzounda Fokou, J.B.; Jazet Dongmo, P.M.; Bakarnga-Via, I.; Ngo Mback, M.N.L.; Zeuko’o, M.E.; Fall, A.D.; Bassene, E.; Fekam, B.F. Optimized combinaition of Ocimum essential oils Inhibit growth of four Candida albicans. Int. J. Drug Discov. 2014, 6, 198–206. [Google Scholar]
  118. Van Vuuren, S.F.; Suliman, S.; Viljoen, A.M. The antimicrobial activity of four commercial essential oils in combination with conventional antimicrobials. Lett. Appl. Microbiol. 2009, 48, 440–446. [Google Scholar] [CrossRef] [PubMed]
  119. Foucquier, J.; Guedj, M. Analysis of drug combinations: Current methodological landscape. Pharmacol. Res. Perspect. 2015, 3, e00149. [Google Scholar] [CrossRef] [PubMed]
  120. Van Vuuren, S.; Viljoen, A. Plant-Based Antimicrobial Studies—Methods and Approaches to Study the Interaction between Natural Products. Planta Med. 2011, 77, 1168–1182. [Google Scholar] [CrossRef] [PubMed]
Table 1. Antifungal compounds from M.O symbiont on marine animals.
Table 1. Antifungal compounds from M.O symbiont on marine animals.
TargetsNbCompoundsProducing SourcesInhibited Fungi* Chemical FormulaReferences
M.O. Symbiont Aquatic Animals
Cellular enzymes1Brefeldin APenicillium sp.Annelle sea fanMicrosporum gypseum (M. gypseum) Candida sp.Pathogens 12 00617 i001[26,27,28]
2Roridin AMyrothecium sp.Sponge Axinelle sp.Saccharomyces cerevisiae (S. cerevisiae)
Magnaporthe grisea (M. grisea)
Sclerotinia sclerotiorum (S. sclerotiorum)
Pathogens 12 00617 i002[29,30,31]
Resistance factors3Modiolide AParaphaeosphaeria sp.Modiolus auriculatus (M. auriculatus)
P. capsici
Neurospora
crassa (N. crassa)
P. capsici
Pathogens 12 00617 i003[32,33]
4Fumiquinazoline AAspergillus fumigatus (A. fumigatus)Pseudolabrus japonicusBotrytis cinerea (B. cinerea), Alternaria solani (A. solani);
(A. alternata), Colletotrichum gloeosporioides (C. gloeosporioides), Fusarium solani (F. solani),
Fusarium oxysporum (F. oxysporum), Gibberella saubinettii
Pathogens 12 00617 i004
Pathogens 12 00617 i005
[34,35,36]
Cell wall5Chitinase 34 kDaStreptomyces sp. DA11Sponge Craniella australiensisA. niger
C. albicans
Pathogens 12 00617 i006[37]
Cell differentiation and multiplication6GriseofulvinPenicillium sp.
(Ascomycota (fungus))
A. verrucosaDermatophytesPathogens 12 00617 i007[11,13]
7Majusculamide CLyngbya majuscule
(a cyanobacteria)
Ptilocaulis trachys (P. trachys)Rhizoctonia solani (R. solani)
Pythium aphanidermatu (P. aphanidermatum)
Aphanomyces euteiches (A.euteiches)
Phytophthora infestans (P.infestans)
Pathogens 12 00617 i008[13,38,39,40,41]
8SurfactinBacillus subtilis A190 (B. subtilis)
B. subtilis A184
Aplysina aerophoba (A. aerophoba)Antifungal
Fusarium moniliforme (F. moniliforme)
Pathogens 12 00617 i009[13,42]
Plasma membrane9IturinB. subtilis A202
B. subtilis A184
A. aerophobaFusarium sp.
Penicellium sp.
Monilinia sp.
R. solani
Pathogens 12 00617 i010[13,43,44,45,46]
103,5-dibromo-2-(3,5-dibromo-2-methoxyphenoxy)phenolVibro sp.Dysidea sp.
Dysidea herbacea
A. fumigatus
Aspergillus flavus (A. flavus)
A. niger
C. tropicalis
C. albicans
Candida glabrata (C. glabrata)
Pathogens 12 00617 i011[35,47,48]
11YM-202204Phoma sp. Q60596sponge Halichondria japonicaC. albicans
Cryptococcus neoformans (C. neoformans)
A. fumigatus
Pathogens 12 00617 i012[49]
12Theonellamide F-Theonella sp.Candida sp.
Trichophyton sp.
Aspergillus sp.
Pathogens 12 00617 i013[50,51,52]
13TheopalauamideCandidatus Entotheonella palauensis(C. Entotheonella)Theonella swinhoei (T. swinhoei)FungiPathogens 12 00617 i014[53,54,55,56,57]
Immunomodulatio n and apoptosis14Indole 3-carboxaldehydeJanthinobacterium lividum (J. lividum)Plethodon cinereus (P. cinereus)B. dendrobatidis
C. albicans
Pathogens 12 00617 i015[58,59]
15IsatinAlteromonas sp. (bacterium)Palaemon macrodactylus (P. macrodactylus) embryos Lagenidium callinectes (L. callinectes)
C. albicans
Candida monosa (C. monosa)
C. glabrata
Tricophyton longifusus(T. longifusus)
Microsporum canis (M. canis)
A. flavus
Pathogens 12 00617 i016[60,61,62,63]
Multicellular targets16DiacetylphloroglucinolLysobacter gummosusP. cinereusB. dendrobatidis
Pythium ultimum var. sporangiiferum
S. cereviaseae
Pathogens 12 00617 i017[64,65,66]
17TyrosolBacterium SGT-76Homarus americanus embryosL. callinectes
C. glabrata
Coccidioides posadasii
Histoplasma capsulatum
Pathogens 12 00617 i018[67,68,69,70]
Unknown targets18Tambjamine CPseudoalteromonas tunicata (P. tunicata)Bryozoans and some other marine animalsMallassezia furfur (M. furfur) C. albicansPathogens 12 00617 i019[24,71]
19Tambjamine EP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i020[24,71]
20Roridin DMyrothecium sp.Sponge Axinelle sp.S. cerevisiae
M. grisea
S. sclerotiorum
Pathogens 12 00617 i021[29,30]
21ViolaceinJ. lividumP. cinereusB. dendrobatidisPathogens 12 00617 i022[58,72,73]
22Tambjamine FP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i023[24,71]
23Tambjamine GP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i024[24,71]
24TheonegramideC. Entotheonella palauenis (δ- Proteobacteria)T. swinhoeiC. albicans (ATCC 32354)Pathogens 12 00617 i025[13,74,75]
25Tambjamine HP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i026[24,71]
26Tambjamine IP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i027[24,71]
27Tambjamine JP. tunicataBryozoans and some other marine animalsM. furfur
C. albicans
Pathogens 12 00617 i028[24,71]
28LL-Z1640-2Cochliobolus lunatus (C. lunatus)Sea AnemoneP. calabae, Plasmopara viticola (P. viticola) P. infestansPathogens 12 00617 i029[76,77]
Détail activités y est
291-hydroxy-6-methyl-8-(hydroxymethyl)xanthoneUlocladium botrytisCallyspongia vaginalis (C. vaginalis)FungiPathogens 12 00617 i030[78,79]
30Xestodecalactone BPenicillium cf. montanense (P. montanense)Xestospongia exiguaC. albicansPathogens 12 00617 i031[13,80]
31Peniciadametizine APenicillium adametzioides (P. adametzioides) AS-53An unidentified marine spongeAlternaria brassicae (A. brassicae) Pathogens 12 00617 i032[81,82]
32Caerulomycin AActinoalloteichus sp.A marine invertebrateCandida sp.Pathogens 12 00617 i033[81,83]
33Unidentified compoundF. oxysporum
DLFP2008005
Hymeniacidon
perlevis
Fungi/[13,84]
3487.12 KDa active ProteinNocardiopsis
dassonvillei MAD08
Sponge Dendrilla nigraCandida sp./[85]
351-hydroxy-6-methyl-8-(hydroxylmethyl)xanthoneUlocladium botrylisSponge
C. vaginalis
C. albicans/[79]
36Modiolide BParaphaeosphaeria sp.M. auriculatusN.
crassa
Pathogens 12 00617 i034[32,33]
37(3S)-(3,5-dihydroxyphenyl)butan-2-one Coniothyrium sp.Ectyplasia perox (E. perox)Ustilago violacea (U. violacea)
Mycotypha microspora
Pathogens 12 00617 i035[78]
38(3R,4S)-
Hydroxymellein
Microsphaeropsis sp.Myxilla incrustans (M. incrustans)U. violaceaPathogens 12 00617 i036[78]
393R)-6-methoxy-7-chloromellein Coniothyrium sp.E. peroxEurotium repens
(E. repens)
Pathogens 12 00617 i037[78,86]
40(3R)-6-methoxymellein Coniothyrium sp.E.peroxE. repensPathogens 12 00617 i038[78]
414,8-dihydroxy-3,4-dihydro-2H-naphthalen-1-one Microsphaeropsis sp.M. incrustansE. repens
U. violacea
Pathogens 12 00617 i039[78]
42(3R,4R)-hydroxymellein Microsphaeropsis sp.M. incrustansE. repens
U. violacea
Pathogens 12 00617 i040[78]
43(R)-mellein Microsphaeropsis sp.M. incrustansE. repensPathogens 12 00617 i041[78]
44Seragikinone AUnidentified fungusCeratodictyon spongiosumC. albicansPathogens 12 00617 i042[26,33,87,88]
45MicrosphaeropsinMicrosphaeropsis sp.M. incrustansE. repens
U. violacea
Pathogens 12 00617 i043
microsphaeropsin A
Pathogens 12 00617 i044
microsphaeropsin B
[33,78]
46(3S)-(3,5-dihydroxyphenyl)butan-2-one Coniothyrium sp.E. peroxU. violacea
Mycotypha microspora
Pathogens 12 00617 i045[78]
47Xestolactone BP. cf. montanenseXestospongia exiguaC. albicansPathogens 12 00617 i046[26,33,80]
48Resorcylic acid lactones “zeaenol”C. lunatusPalythoa haddoniP. viticola,
P.infestans
Pathogens 12 00617 i047[76,89]
* Chemical structures of compounds were obtained from spectrabase, pubchem and chemspider https://spectrabase.com; https://pubchem.ncbi.nlm.nih.gov; http://www.chemspider.com/ (accessed on 5 January 2023).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ngo-Mback, M.N.L.; Zeuko’o Menkem, E.; Marco, H.G. Antifungal Compounds from Microbial Symbionts Associated with Aquatic Animals and Cellular Targets: A Review. Pathogens 2023, 12, 617. https://doi.org/10.3390/pathogens12040617

AMA Style

Ngo-Mback MNL, Zeuko’o Menkem E, Marco HG. Antifungal Compounds from Microbial Symbionts Associated with Aquatic Animals and Cellular Targets: A Review. Pathogens. 2023; 12(4):617. https://doi.org/10.3390/pathogens12040617

Chicago/Turabian Style

Ngo-Mback, Madeleine Nina Love, Elisabeth Zeuko’o Menkem, and Heather G. Marco. 2023. "Antifungal Compounds from Microbial Symbionts Associated with Aquatic Animals and Cellular Targets: A Review" Pathogens 12, no. 4: 617. https://doi.org/10.3390/pathogens12040617

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

Article Metrics

Back to TopTop