Next Article in Journal
Characterization of Transcriptomic and Proteomic Changes in the Skin after Chronic Fluocinolone Acetonide Treatment
Next Article in Special Issue
Analysis of Antioxidant and Antiviral Effects of Olive (Olea europaea L.) Leaf Extracts and Pure Compound Using Cancer Cell Model
Previous Article in Journal
Comprehensive Genomic Profiling of Cell-Free Circulating Tumor DNA Detects Response to Ribociclib Plus Letrozole in a Patient with Metastatic Breast Cancer
Previous Article in Special Issue
Biodefensive Based on Piper nigrum Essential Oil for Controlling of Anopheles aquasalis Larvae: Influence of Temperature (35 °C) and Preservatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Secondary Metabolites Diversity of Aspergillus unguis and Their Bioactivities: A Potential Target to Be Explored

by
Levy Tenório Sousa Domingos
1,
Raquel dos Santos Martins
2,
Leonardo Melo de Lima
2,
Angela Michelato Ghizelini
1,
Antonio Ferreira-Pereira
1,* and
Fernando Cotinguiba
2,*
1
Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro CEP 21941-902, Brazil
2
Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco H, Cidade Universitária, Rio de Janeiro CEP 21941-902, Brazil
*
Authors to whom correspondence should be addressed.
Biomolecules 2022, 12(12), 1820; https://doi.org/10.3390/biom12121820
Submission received: 12 October 2022 / Revised: 10 November 2022 / Accepted: 15 November 2022 / Published: 6 December 2022
(This article belongs to the Special Issue Bioactive Natural Compounds against Animal and Human Pathogens)

Abstract

:
Aspergillus unguis belongs to the Aspergillus section Nidulantes. This species is found in soils and organisms from marine environments, such as jellyfishes and sponges. The first chemical study reported in the literature dates from 1970, with depsidones nidulin (1), nornidulin (2), and unguinol (3) being the first isolated compounds. Fifty-two years since this first study, the isolation and characterization of ninety-seven (97) compounds have been reported. These compounds are from different classes, such as depsides, depsidones, phthalides, cyclopeptides, indanones, diarylethers, pyrones, benzoic acid derivatives, orcinol/orsenillate derivatives, and sesterpenoids. In terms of biological activities, the first studies on isolated compounds from A. unguis came only in the 1990s. Considering the tendency for antiparasitic and antibiotics to become ineffective against resistant microorganisms and larvae, A. unguis compounds have also been extensively investigated and some compounds are considered very promising. In addition to these larvicidal and antimicrobial activities, these compounds also show activity against cancer cell lines, animal growth promotion, antimalarial and antioxidant activities. Despite the diversity of these compounds and reported biological activities, A. unguis remains an interesting target for studies on metabolic induction to produce new compounds, the determination of new biological activities, medicinal chemistry, structural modification, biotechnological approaches, and molecular modeling, which have yet to be extensively explored.

1. Introduction

Many fungi are capable of producing heterogeneous low-molecular-mass compounds, also called secondary metabolites. These metabolites are not directly necessary for organism growth, unlike primary metabolites [1,2]. The versatile metabolism of fungi allows different types of interactions with other organisms, ranging from bacteria to metazoa, and substrates, which play essential roles in ecosystems. Over hundreds of millions of years, fungi use these metabolites as chemical signals for communication, defending their habitat, or inhibiting the growth of competitors, leading to the evolution of natural products and, consequently, acting in the ecological success of fungi in colonizing approximately all habitats on the planet [3,4,5,6].
Penicillin, considered the “wonder drug” of World War II and the first broad-spectrum antibiotic, was discovered in the fungus Penicillium notatum by Alexander Fleming in 1928. This metabolite significantly changed the search for new natural products from plants to microorganisms, inaugurating “the golden age of antibiotics [7,8,9]. Currently, 45 % of the known microbial metabolites are of fungal origin such as filamentous fungi, including Penicillium, Trichoderma, and Aspergillus, which represent almost 99 % of the total known fungal metabolites [10].
Aspergillus is an ascomycetous fungus with the greatest bioactive potential in nature. Species of this genus have been studied extensively for years, and they produce metabolites of considerable medical, industrial, agricultural, and economic importance. For example, the phytohormone gibberellin produced by A. fumigatus improves plant growth, and the anti-cholesterol drug lovastatin produced by A. terreus, with worldwide sales of USD 10 billion annually. The metabolic roles of these metabolites have been increasingly explored in biotechnological research that seeks new fungal natural products of commercial interest [11,12,13].
Fungal species such as A. niger, A. oryzae, and A. terreus are considered the work horses in biotechnology [14]. However, other species that are not in the spotlight and are less well known have great biotechnological value that has already been described, such as A. unguis. Research into this fungus include its application for the removal of heavy metals from industrial wastewater [15], expression systems for heterologous protein expression [16], production of enzymes of industrial interest [17,18] and, since the last century, the rich production of secondary metabolites. Therefore, this review highlights the secondary metabolites of A. unguis and their biological activities, with a brief description of the biology of A. unguis.

2. Biology of A. unguis

The fungus A. unguis, described in 1935 by Emile-Weill and L. Gaudin, belongs to the Aspergillus section Nidulantes, forming the series Unguium with four other species: A. israelensis [19], A. croceus [20], A. longistipitatus [21], and A. croceiaffinis [21], all of which were described between 2016 and 2020 using polyphasic taxonomy; that is, with the application of phenotypic, genotypic (using multigenic DNA sequences), and chemotaxonomic techniques [19,20,21,22]. Aspergillus taxonomy is complex, and it changes over time as species identification technology reorganizes just as new species are described [23]. In recent years, many efforts have been made by the scientific community to revise the taxonomy and phylogeny of several species that are already known through polyphasic taxonomy, mainly using DNA sequence data, in order to reorganize these thousands of species in light of new knowledge and modern techniques [24].
Emericella unguis is the sexual state (teleomorph) of A. unguis (anamorph), and it became synonymous with A. unguis with the new nomenclatural rules based on a single-name system established by the International Commission of Penicillium and Aspergillus (ICPA) in 2012 [24]. A. unguis has other possible synonyms, such as Sterigmatocystis unguis and A. mellinus, and there may be others; however, the differentiation of A. unguis and non-cleistothecial A. nidulans isolates was problematic in the past [20].
The complete A. unguis genome was sequenced in 2016 as part of a project to sequence Aspergillus species (Joint Genome Institute, https://jgi.doe.gov, accessed on 24 April 2022) [24]. This information may soon contribute to the taxonomic reordering of the species mainly by using the supporting information on metabolite production.
A. unguis can be isolated from soils [25], lichens [26], and organisms in marine environments, such as jellyfishes [27], seaweeds [28], sponges [29], and others. This ubiquitous fungus has great potential for the “one strain many compounds” (OSMAC) strategy, as it is capable of growing in culture broth with different sources of carbon and nitrogen, such as potato dextrose, oatmeal, glycerol casein, yeast extract sucrose, Czapek-Dox, and malt extract [30,31]. Solid media, in addition to solid agar media, are also used as the main media [26]. On some occasions, the medium can be supplemented with specific salts to study the assimilation and modulation of the production of secondary metabolites or with sea water to mimic the environment of marine isolates [29,30]. The use of A. unguis biology and its different growth methods are excellent alternatives for the discovery of new metabolites of this fungus.

3. Secondary Metabolism

A review of the 52-year-long chemistry studies of A. unguis (1970–2022) was performed, from the first articles published by Kamal in 1970 to the last published by Cao in 2022 [32,33]. In this review, ninety-seven chemical structures of compounds isolated and identified from A. unguis are reported. In Table 1, these compounds are organized by code, presenting the following information: (I) the class of metabolites to which they belong, (II) the biological activities of each compound, and (III) the references that report the isolation and identification. The compounds found in A. unguis are classified into depsides, depsidones, phthalides, cyclopeptides, indanones, diarylethers, pyrones, benzoic acid derivatives, orcinol/orsenillate derivatives, and sesterpenoids. Most of the compounds are depsidones, representing approximately 30 % of the total. Among the ninety-seven compounds isolated from A unguis, only twelve were also isolated from other microorganisms: nidulin (1), nornidulin (2), unguinol (3), 2-chlorounguinol (8), (3S)-3-ethyl-5,7-dihydroxy-3,6-dimethylphthalide (9), folipastatin (13), unguisin E (20) aspergillusidone C (23), pilobolusate (37), (+)-montagnetol (38), averantin (75), and corynesidone D (84) (see Table 1). The chemical structure of each compound is also presented in Figure 1.
A549 = human lung carcinoma, AChE = acetylcholinesterase, BS = Bacillus subtilis (ATCC 6633 or KCTC 1021), CA = Candida albicans NCPF3153, CN = Cryptococcus neoformans ATCC90113, HepG2 = human hepatocellular liver carcinoma, HT-29 = human colorectal adenocarcinoma cell line, HuCCA-1 = human cholangiocarcinoma, KB = human oral carcinoma cell line, MRSA = methicillin-resistant Staphylococcus aureus, MCF-7 = breast cancer cells, MG = Microsporum gypseum clinical isolate, MOLT-3 = acute lymphoblastic leukemia, NS-1= mouse myeloma cells (ATCC TIB-18), PA = Pseudomonas aeruginosa, PN = Penicillium marnefeii, SA = Staphylococcus aureus (ATCC25923, or ATCC29213, or KCTC 1927), SC = Saccharomyces cerevisiae (ATCC 9763), SMMC-7721 = human hepatoma cells, SOAT1 = sterol O-acyltransferase ubiquitously expressed in all tissues and cells, SOAT2 = sterol O-acyltransferase expressed predominantly in the liver (hepatocytes) and intestine, U251 = glioblastoma cell line, U87 = human primary glioblastoma cell, PF = Plasmodium falciparum (K1, multidrug-resistant strain), MT = Mycobacterium tuberculosis (ATCC25177); BC = Bacillus cereus (ATCC11778); AB = Alternaria brassicicola (BCC42724); CA = Colletotrichum acutatum (BCC58146); ML = Micrococcus luteus (KCTC 1915); MV = Microbulbifer variabilis; MJ = Marinobacterium jannaschii; VP = Vibrio pelagius.

3.1. Depsidones

Depsidones consist of two aromatic rings (A and C rings) joined by a -CO-O- bridge (ester group) and an ether group, forming a third seven-membered ring (B ring). This class of compounds is biosynthesized through oxidative coupling of the depsides [42,70].
Three depsidones, nidulin (1), nornidulin (2), and yasimin (3), were isolated [33]. In the same year, the authors published a biosynthetic study of yasimin (3) by incorporating labeled acetate (1-14C and 2-14C) and malonate-1-14C using a fungus culture growing in Czapek-Dox medium [54]. The labeled compound 3 was isolated, and the authors were able to make a series of considerations about the biosynthesis pathways of this compound, classifying them as polyketides [41,54]. Furthermore, in 1970, Kamal published another study describing the identification of four new depsidones from A. unguis: haiderin (4), rubinin (5), (-)-shirin (6), and nasrin (7). None of the four compounds described in this study were reisolated from A. unguis or any other species of fungus; therefore, this is the only report of these four compounds in literature.
Stodola et al. reisolated compound 3 in 1972 but did not publish the results; however, it was renamed unguinol (3) [56]. Since then, most articles have referred to this compound as unguinol, not yasimin, despite having exactly the same chemical structure. In 1983, Turner and Aldridge identified nidulin (1), nornidulin (2), and unguinol (3) in E. nidulans (anamorph A. nidulans) and A. mellinus; however, these two organisms have been reidentified as A. unguis [27]. In 1988, Kawahara et al. published two studies describing a series of new depsidones, including emeguisin A (10), emeguisin B (11), and emeguisin C (12) [38,39]. These compounds are the first examples of depsidones bearing two 1-methylprop-1-enyl groups in one molecule. Uchida et al. described the structure of a supposedly unprecedented compound, which they named 7-chlorofolipastatin (10). However, this compound was previously identified as emeguisin A (10) [47]. Sureram et al. (2013) subjected A. unguis to growth in medium containing different halogen salts (potassium bromide [KBr], potassium iodide [KI], and potassium fluoride). The fungus grown in medium containing KBr produced three new brominated depsidones, namely aspergillusidones D–F (26–28). Meanwhile, when the fungus was subjected to medium containing KI, the fungus did not incorporate iodine atoms into the compounds, but a new depsidone, 2,4-dichlorounguinol (32) was isolated and identified. Morshed et al. (2018) manipulated the concentration of halides in the culture medium, inducing A. unguis to produce a series of new compounds, by OSMAC strategy. When the fungus was grown in yeast extract with supplements (YES) medium without saline supplementation, several compounds were produced in addition to an unprecedented substance, 7-carboxyfolipastatin (45). When the YES medium was supplemented with 0.5 % sodium chloride, it was possible to decrease the production of unguinol (3), in addition to producing new compounds such as 45 and 4,7-dichlorounguinol (46). In an experiment with 0.5 % KBr supplementation, the fungus produced new substances, such as 7-bromounguinol (48), 2-chloro-7-bromounguinol (49), and 7-bromofolipastatin (50). Sureram (2013) and Morshed (2018) explored the concept of precursor-directed biosynthesis, which is an attempt to exploit the metabolic potential of fungi in the face of a precursor. Modifying the concentration of the halides is a powerful tool for modulating secondary metabolite production and triggering quiescent pathways in the fungus. The fungus must be able to recognize, metabolize and generate new unnatural structures [71]. Sureram (2013) and Morshed (2018) showed that this approach was efficient in generating new structures of secondary metabolites. The structural diversity of depsidones has increased due to the recent discoveries of Saetang et al. (2021). They described two new depsidones (69 and 70), both harboring an interesting structure different from all other depsidones: the substitution of the 2-butenyl unit (C ring) for the hydroxy-3-butenyl moiety group. These recent discoveries show that the metabolic potential of A. unguis remains to be elucidated and may elicit surprise.

3.2. Depsides

Depsides are substances that are biosynthesized from the union of two orsellinic acid derivative units or the union of an orsellinic acid derivative and an orcinol derivative [42]. Depsides are related to depsidones and are generally accepted as biosynthetic precursors of this depsidones [43]. Nielsen et al. (1999) isolated and identified the first depside in A. unguis: guisinol (15). After almost 20 years, Phainuphong et al. (2018) described the identification of three new depsides: aspergiside A (54), aspergiside B (52), and aspergiside C (58). Coincidentally, Morshed et al. (2018) also described the isolation and structural characterization of compounds 52 and 54, but they called these structures unguidepside A (52) and decarboxyunguidepside A (54). Yang et al. (2018) used a plasma-induced mutant and the combined use of an epigenetic modifier (procaine) with sodium bromide as a strategy to diversify the production of secondary metabolites by a marine strain of A. unguis. Ten compounds were isolated and identified, one of which was a new depside named aspergillusidone G (52). However, this compound is structurally identical to those described by Phainuphong et al. (2018) and Morshed et al. (2018): aspergiside B (52) and unguidepside A (52), respectively. Yang et al. (2018) classified aspergillusidone G (52) as a depsidone, but the other authors classified it as a depside [25,28,30].

3.3. Phthalides

Phthalide or isobenzofuran-1(3H)-one is a chemical group that joins an aromatic ring and a dihydrofuranone group. The 3-ethyl-5,7-dihydroxy-3,6-dimethylphthalide (9) was the first phthalide isolated from A. unguis. This compound is also considered the first example of a 3,3-disubstituted phthalide obtained from a natural source [38]. However, there are still a few descriptions of this class of compounds in A. unguis, especially the following: asperlide (34), aspergiside C (58), asperunguislide A (71), and asperunguislide B (72).

3.4. Cyclopeptides

Cyclic heptapeptides unguisin A (16) and unguisin B (17) isolated from a marine-derived strain of E. unguis are considered to contain gamma-aminobutyric acid (GABA) in the ring. The only difference between the two peptides is that unguisin A (16) contains L-phenylalanine, while unguisin B (17) has L-leucine [55]. The cyclopeptide unguisin C (18) was also isolated as a minor component in 2002 [63]. This peptide is structurally similar to unguisin A (16), except for the substitution of D-alanine to D-serine, yielding a more hydrophilic peptide. Unguisin D (19) was produced when L-leucine was added to the fermentation medium and was detected only by liquid chromatography mass spectrophotometry analysis (LC-MS), with the structure cyclo-(tryptophyl-GABA-alanyl-valyl-leucyl-leucyl-alanyl). This compound is similar to unguisin B (17) with the replacement of valine-2 to leucine [63]. In 2011, Liu and Chen (2011) reported the isolation of a new cyclopeptide, unguisin E (20), from Aspergillus sp. AF119 [64]. The only difference was that the amino acid phenylalanine in unguisin A (16) was replaced by β-methylphenylalanine in 20.

3.5. Pyrone

Pyrones are a class of heterocyclic chemical compounds that contain an unsaturated six-membered ring containing one cyclic ester functional group. Despite its presence in fungi, the metabolism of pyrones is still not well known in A. unguis; only one compound of this class has been isolated and identified. The 3-methyl-4-hydroxy-6-(1-trans-methyl-1-propenyl)-2-pyrone (24) was isolated as a natural source for the first time. This pyrone was a des-O-methyl derivative of nectriapyrone, a metabolite previously isolated from the fungus Gyrostroma missouriense [29].

3.6. Diarylethers

Several diarylethers were isolated from A. unguis from 2010. The first was an unprecedented compound described by Sureram et al. (2012) as aspergillusether A (25). The authors considered the possibility that compound 25 could be an artifact obtained from the cleavage of an ester bond of nidulin (1), followed by methylation at the carboxylic group, which possibly occurred during extraction and purification. Subsequently, several new diaryethylethers were isolated as aspergillusethers B-D (41–43) [31], unguolic acid (55), decarboxyunguolic acid (56), 5-chlorounguinolic acid (57) [30] and aspergillusethers E–F (67–68) [41].

3.7. Orcinol/Orsenillate Derivatives and Benzoic Acid Derivatives

Structurally, orcinol consists of an aromatic ring interspersed by two hydroxyls and one methyl group. Orcinol (44), despite being a known substance, has not yet been reported in A. unguis [31]. Two new orcinol derivatives, aspergillusphenol A (29) and aspergillusphenol B (30), were identified by Sureram et al. (2013). Orselinates had not been reported in A. unguis until the study by Klaiklay et al. (2017) who identified methyl orsellinate (36), pilobolusate (37), and (+)-montagnetol (38). The (+)-montagnetol is commonly isolated from lichen, especially from the genus Roccella [72].

3.8. Indanones

Phainuphong described the isolation and structural characterization of a class of compounds that had not yet been identified in A. unguis: indanones. Two new indanones, asperunguisone A (39) and asperunguisone B (40), were described. Indanones structurally exhibit a junction between an aromatic ring and a cyclopropane ring with a ketone function [31].

3.9. Anthraquinones

Anthraquinones (polyketides compounds) are an important chemical group. The skeleton is basically two aromatic rings separated by two carbonyl groups. Although these compounds have already been isolated from other species of the genus Aspergillus, it was only in 2021 that the anthraquinones averantin (75), 7-chloroaverantin (76) and 1’-O-methylaverantion (77) were isolated from strains 158SC-067 of A. unguis [68].

3.10. Terpenoids and Sterols

Terpenoids belong to a class of compounds formed by the coupling of isoprene units (dimethylallyl pyrophosphate and isopentenyl pyrophosphate) and are classified into subclasses according to the multiplicity of five carbons. In fungi, these isoprene units originate from the mevalonate pathway. Li et al. (2019) isolated and identified six new asperane-type sesterterpenoids (terpenes with 25 carbon atoms), asperunguisins A–F (59–64), together with a known analogue, aspergilloxide (65). These are rare asperane-type of sesterterpenoids, characterized by a unique hydroxylated 7/6/6/5 tetracyclic system. Compound 65 was discovered as the first asperane-type sesterterpenoid from the marine-derived fungus Aspergillus sp. in 2002 [73]. Rare ergostane-type sterols with an unusual unsaturated side chain (94–97) were reported by Cao and collaborators [32]. It was the first time that this type of compound had been isolated from A. unguis.

4. Biological Potential of Natural Products Isolated from A. unguis

Several biological activities were attributed to the various compounds isolated from A. unguis: enzymatic inhibition (acetylcholinesterase, aromatase, phospholipase A, sterol O-acyltransferase, xanthine oxidase), activity against cancer cell lines (A549, HB, HuCCA-1, HepG2, HT-29, KB, MCC-7, MOLT-3, NS-1, SMMC-7721, T47D, U251, U87, Vero), antibacterial properties (against Bacillus subtilis, Staphylococcus aureus, methicillin-resistant S. aureus, Pseudomonas aeruginosa), antifungal properties (against Candida albicans, Cryptococcus neoformans, Microsporum gypseum), herbicidal effects, larvicidal effects (against brine shrimp larva lethality test), DNA repair activity, animal growth promotion, antimalarial properties, and antioxidant activities. Table 1 presents data on the biological activities of all ninety-seven compounds already identified in A. unguis. All this information shows the immense potential, indicating the ways that can be better explored and others that have not been investigated.

4.1. Antifungal, Antibacterial, Antimalarial and Larvicidal Activities

Considering the tendency for antibiotics to become ineffective against resistant strains, A. unguis compounds have also been extensively investigated and some of them are very promising against larvae and pathogenic microorganisms. A good example is the study by Yang et al. (2022) which show the potent activity of depsidones nornidulin (2), emeguisin A (10) and folipastatin (13) against vancomycin-resistant bacteria Enterococcus faecium [44]. Synthetic derivatives of nidulin (1) were also investigated for their significant activities against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus bacterial properties. These derivatives, in particular 8-O-aryl ether derivatives, proved to be very promising [74].
Saetang et al. pointed out the activity of the depsides 10 and 13 that were strongly active against the pathogenic yeast Candida neoformans with respective MIC values of 1 and 0.5 μg/mL, an identical value to the positive control, the antifungal amphotericin B. Another despside, aspergillusidone C (23) showed the strongest antifungal activity against Microsporum gypseum with the MIC value of 2 μg/mL, similar to positive control clotrimazole.
Yang et al. (2018) considers that the compounds nidulin (1), nornidulin (2) and aspergillusidone F (28) could be used for the development of pesticides for their larvicidal activity. These depsidones exhibited potent larvicidal activity against brine shrimp, with close or even lower LC50 values compared with the positive control mercury (II) nitrate, Hg(NO3)2 [31]. Klaiklay and colleagues also demonstrated the potent antimicrobial activities of the depsidone emeguisin A (13). This compound 13 exhibited the most potent antibacterial (against S. aureus and methicillin-resistant S. aureus), and antifungal (against C. neoformans) activities with MIC values of 0.5 µg/mL. Emeguisin A (13) also showed antimalarial activity, with MIC values of 2.2 µM against P. falciparum, identical to the positive control dihydroartemisinin [40].

4.2. Anti-Osteoclastogenic Activity

With the aging of the population, some diseases such as osteoporosis have become a concern in recent times. Therefore, the search for new bioactive molecules that can be applied to treatments and therapies becomes extremely important and some research groups have moved towards this. Zhang et al. (2022) evaluated several pure substances and guisinol (15) proved to be a further dose-dependent suppressed RANKL-induced osteoclast differentiation without any evidence of cytotoxicity in bone marrow macrophage cells. This is the first article that reports the identification of an A. unguis compound that has this type of activity [46].

4.3. Cytotoxic Activity against Cancer Cell Lines

Cancer is one of the most feared diseases and its treatment can still leave the patient debilitated and sometimes discouraged. Compounds isolated from A. unguis have been extensively evaluated against several tumor cell lines. As an example, Cao et al. (2022) evaluated ergosterane-type sterol against ACHN (renal), HCT-15 (colon), NUGC-3 (stomach), PC-3 (prostate), NCI-H23 (lung), and MDA-MB-231 (breast) cancer cell lines. The new substance aspersterol A (94) showed cytotoxicity against all the tested cell lines [32]. The orsellinate pilobolusate (37) exhibited potent activity against the KB cell line with an IC50 value of 4.5 µM which was much stronger than the standard drug, ellipticin [40]. The asperane sesterterpenoid asperunguisin C (61) showed cytotoxicity against the human cancer cell line A549 with an IC50 value of 6.2 μM, a value very close to the positive control adriamycin (2.9 μM) [26].

4.4. Anti-Inflammatory Activity

Cao and colleagues monitored the anti-inflammatory potential of ergostanes and concluded that the compound aspersterol C (96) showed the most potent anti-inflammatory activity. This compound inhibited the production of inflammatory mediators, including IL-6 and iNOS in LPS-induced macrophages (RAW 264.7 cells). The authors consider that this molecule could be used as a lead for additional studies for anti-inflammatory models [32]. In a similar study, this research group isolated A. unguis and the anti-inflammatory properties of compound variotin B (90), the first linear nitrogenous secondary metabolite isolated from A. unguis [62].

4.5. Enzyme Inhibitors

Biological tests involving natural products were greatly advanced in the 1990s. Among all the studies analyzed, the first one that shows the attribution of biological activity to a compound isolated directly from A. unguis was reported by Hamano and collaborators in 1992, wherein depsidone folipastatin (13) inhibited the enzyme phospholipase A2 enzyme isolated from rabbit peritoneal exudate. These enzymes are involved in inflammatory processes [52]. Studies suggest that unguinol inhibits C4 plant enzyme pyruvate phosphate dikinase (PPDK) via a novel mechanism of action which also translates to a herbicidal effect on whole plants. This compound had deleterious effects on a model C4 plant but no effect on a model C3 plant [49].
The depsidone aspergillusidone C (23) showed the inhibitory activity of the aromatase enzyme with an IC50 value of 0.74 µM, being more potent than the positive standard ketoconazole (IC50 value 2.4 µM). It was concluded, from the structure activity relationship view, that the depsidone structure is important for this type of activity. The aromatase enzyme participates in aromatization reactions from androgens to the aromatic ring of estrogens and the inhibition of this enzyme reduces the incidence of breast cancer [29]. The same research group showed in another publication that unguinol (3) and aspergillusidone A (21) are also inhibitors of this enzyme [50]. The depsidones unguinol (3) and aspergillusidone D (26) were selected for further studies. Unguinol (3) induced apoptosis and cell cycle arrest in the breast cancer cell line-MB 231. Unguinol (3) and aspergillusidone D (26) also inhibit lifetime cell viability at low concentrations (μM) [51].
Depsidones were also evaluated for their potential to inhibit the enzyme acetylcholinesterase (AChE). Aspergillusidone A (21) showed AChE inhibition with IC50 value of 56.8 μM, with donepezil used as a positive control (IC50 = 0.3 μM). Docking studies were also performed, showing that this depsidone interacts with the enzyme in different ways [28].

5. Summary and Future Perspectives

In the past fifty-two years (1970–2022), since the first chemical study of A. unguis, ninety-seven compounds from different classes of microbial secondary metabolites have been isolated and identified by spectrometric and spectroscopic techniques. Many of the reported compounds are produced exclusively by A. unguis. The substances isolated from A. unguis have promising biological activity against pathogens such as larvae, bacteria and yeasts, with several examples where these substances are even more active in in vitro assays than the positive controls. Considering the tendencies of antiparasitic and antibiotics to become less effective, it is very important to continue these studies in order to investigate their action in vivo. In addition to antimicrobial and larvicidal activities, these substances also demonstrate promising activities against the cancer cell lineage, anti-inflammatory, and antimalarial activities. Several compounds of A. unguis have also been shown to be enzyme inhibitors, such as aromatase and AChE. The metabolic potential of A. unguis is still largely untapped; therefore, it has great potential for innovation. Variations in the concentration or depletion of salts in the culture media proved to be an extremely interesting approach to produce new structures. The OSMAC strategy, despite having been successfully applied by several authors in A. unguis, can still be widely explored, changing the parameters of microorganism cultures, in order to allow the fungus to produce new chemical structures [28,30,68]. Approaches using epigenetic regulation also have not been widely used in A. unguis. Epigenetic regulation is critical for fungal secondary metabolism biosynthesis and the activation of gene clusters.The modulation of epigenetic regulation is an interesting alternative for discovering new secondary metabolites and improving their production [75]. Another tool that could also accelerate the discovery of new minority metabolites as well as understand the metabolic capacity of A. unguis are metabolomics-based dereplication methods. These approaches have not yet been applied to this fungus and deserve attention [76]. Medicinal chemists and molecular modeling specialists could also contribute significantly to the understanding of the interaction of secondary metabolite X enzyme inhibition. Despite the diversity of chemical structures, few studies have focused on the structural modification of these compounds which deserve attention in order to obtain semisynthetic derivatives even more potent than the natural substances.

Author Contributions

Individual contributions of the authors are the following: conceptualization, F.C. and L.T.S.D.; methodology, F.C. and L.T.S.D.; formal analysis, L.T.S.D., R.d.S.M., L.M.d.L., A.M.G., A.F.-P. and F.C.; investigation, L.T.S.D., R.d.S.M., L.M.d.L., A.M.G., A.F.-P. and F.C.; resources, A.F.-P. and F.C.; writing original draft preparation, L.T.S.D., R.d.S.M., L.M.d.L., A.M.G., A.F.-P. and F.C.; writing review and editing, L.T.S.D., R.d.S.M., L.M.d.L., A.M.G., A.F.-P. and F.C.; supervision, A.F.-P. and F.C.; project administration, FC; funding acquisition, A.F.-P. and F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financed by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brazil (CAPES)—Finance Code 001 and Conselho Nacional de Desenvolvimento Científico e Tecnológico—Brazil (CNPq). L.T.S.D. thanks CNPq for the PhD scholarship; R.d.S.M. and L.M.d.L. thanks CAPES for the PhD scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

L.T.S.D. thanks CNPq for the PhD scholarship; R.d.S.M., L.M.d.L. and, A.M.G. thanks CAPES for the PhD scholarship. A.F.-P. and F.C. thank FAPERJ, CNPq and CAPES for their financial support.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Newman, D.J.; Cragg, G.M.; Snader, K.M. The influence of natural products upon drug discovery. Nat. Prod. Rep. 2000, 17, 215–234. [Google Scholar] [CrossRef] [Green Version]
  2. Brakhage, A.A. Regulation of fungal secondary metabolism. Nat. Rev. Microbiol. 2013, 11, 21–32. [Google Scholar] [CrossRef]
  3. Bills, G.F.; Gloer, J.B. Biologically Active Secondary Metabolites from the Fungi. Microbiol. Spectr. 2016, 4, 1087–1119. [Google Scholar]
  4. Brakhage, A.A.; Schroeckh, V. Fungal secondary metabolites—Strategies to activate silent gene clusters. Fungal Genet. Biol. 2011, 48, 15–22. [Google Scholar] [CrossRef]
  5. Blackwell, M. The fungi: 1, 2, 3 …5.1 million species? Am. J. Bot. 2011, 98, 426–438. [Google Scholar] [CrossRef]
  6. Richards, T.A.; Leonard, G.; Wideman, J.G. What Defines the “Kingdom” Fungi? Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef] [PubMed]
  7. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta 2013, 1830, 3670–3695. [Google Scholar]
  8. Pham, J.V.; Yilma, M.A.; Feliz, A.; Majid, M.T.; Maffetone, N.; Walker, J.R.; Kim, E.; Cho, H.J.; Reynolds, J.M.; Song, M.C.; et al. A Review of the Microbial Production of Bioactive Natural Products and Biologics. Front. Microbiol. 2019, 10, 1404. [Google Scholar]
  9. Keller, N.P. Fungal secondary metabolism: Regulation, function and drug discovery. Nat. Rev. Microbiol. 2019, 17, 167–180. [Google Scholar] [CrossRef] [PubMed]
  10. Berdy, J. Thoughts and facts about antibiotics: Where we are now and where we are heading. J. Antibiot. 2012, 65, 385–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Inglis, D.O.; Binkley, J.; Skrzypek, M.S.; Arnaud, M.B.; Cerqueira, G.C.; Shah, P.; Wymore, F.; Wortman, J.R.; Sherlock, G. Comprehensive annotation of secondary metabolite biosynthetic genes and gene clusters of Aspergillus nidulans, A. fumigatus, A. niger and A. oryzae. BMC Microbiol. 2013, 13, 91. [Google Scholar] [CrossRef] [Green Version]
  12. Sanchez, J.F.; Somoza, A.D.; Keller, N.P.; Wang, C.C. Advances in Aspergillus secondary metabolite research in the post-genomic era. Nat. Prod. Rep. 2012, 29, 351–371. [Google Scholar] [CrossRef] [Green Version]
  13. Khan, A.L.; Hamayun, M.; Kim, Y.-H.; Kang, S.-M.; Lee, J.-H.; Lee, I.-J. Gibberellins producing endophytic Aspergillus fumigatus sp. LH02 influenced endogenous phytohormonal levels, isoflavonoids production and plant growth in salinity stress. Process Biochem. 2011, 46, 440–447. [Google Scholar] [CrossRef]
  14. Frisvad, J.C.; Larsen, T.O. Chemodiversity in the genus Aspergillus. Appl. Microbiol. Biotechnol. 2015, 99, 7859–7877. [Google Scholar] [CrossRef]
  15. Sharaf, E.F.; Alharbi, E. Removal of heavy metals from waste water of tanning leather industry by fungal species isolated from polluted soil. Afr. J. Biotechnol. 2013, 12, 4351–4355. [Google Scholar]
  16. Madhavan, A.; Pandey, A.; Sukumaran, R.K. Expression system for heterologous protein expression in the filamentous fungus Aspergillus unguis. Bioresour. Technol. 2017, 245, 1334–1342. [Google Scholar] [CrossRef]
  17. Shruthi, K.; Yadav, P.S.; Prasad, B.V.S.; Chandra, M.S. Cellulase production by Aspergillus unguis in solid state fermentation. J. For. Res. 2019, 30, 205–212. [Google Scholar] [CrossRef]
  18. Kooloth Valappil, P.; Rajasree, K.P.; Abraham, A.; Christopher, M.; Sukumaran, R.K. Characterization of a glucose tolerant beta-glucosidase from Aspergillus unguis with high potential as a blend-in for biomass hydrolyzing enzyme cocktails. Biotechnol. Lett. 2019, 41, 1201–1211. [Google Scholar] [CrossRef]
  19. Chen, A.J.; Frisvad, J.C.; Sun, B.D.; Varga, J.; Kocsube, S.; Dijksterhuis, J.; Kim, D.H.; Hong, S.B.; Houbraken, J.; Samson, R.A. Aspergillus section Nidulantes (formerly Emericella): Polyphasic taxonomy, chemistry and biology. Stud. Mycol. 2016, 84, 1–118. [Google Scholar]
  20. Hubka, V.; Nováková, A.; Peterson, S.W.; Frisvad, J.C.; Sklenář, F.; Matsuzawa, T.; Kubátová, A.; Kolařík, M. A reappraisal of Aspergillus section Nidulantes with descriptions of two new sterigmatocystin-producing species. Plant. Syst. Evol. 2016, 302, 1267–1299. [Google Scholar] [CrossRef]
  21. Sklenar, F.; Jurjevic, Z.; Peterson, S.W.; Kolarik, M.; Novakova, A.; Flieger, M.; Stodulkova, E.; Kubatova, A.; Hubka, V. Increasing the species diversity in the Aspergillus section Nidulantes: Six novel species mainly from the indoor environment. Mycologia 2020, 112, 342–370. [Google Scholar] [CrossRef]
  22. Houbraken, J.; Kocsube, S.; Visagie, C.M.; Yilmaz, N.; Wang, X.C.; Meijer, M.; Kraak, B.; Hubka, V.; Bensch, K.; Samson, R.A.; et al. Classification of Aspergillus, Penicillium, Talaromyces and related genera (Eurotiales): An overview of families, genera, subgenera, sections, series and species. Stud. Mycol. 2020, 95, 5–169. [Google Scholar]
  23. Batista, L.R.; Souza, S.C.; Evangelista, S.R.; Azevedo, N.L. Identificação de fungos: Gêneros Aspergillus, Penicillium e Talaromyces. In Conhecimento, Conservação e Uso de Fungos; Oliveira, L.A., Jesus, M.A., Matsuura, A.B.J., Oliveira, J.G.S., Gasparotto, L., Lima-Neto, R.G., Rocha, L.C., Eds.; Editora INPA: Manaus, Brazil, 2019; pp. 1–10. [Google Scholar]
  24. Samson, R.A.; Visagie, C.M.; Houbraken, J.; Hong, S.B.; Hubka, V.; Klaassen, C.H.; Perrone, G.; Seifert, K.A.; Susca, A.; Tanney, J.B.; et al. Phylogeny, identification and nomenclature of the genus Aspergillus. Stud. Mycol. 2014, 78, 141–173. [Google Scholar] [CrossRef] [Green Version]
  25. Phainuphong, P.; Rukachaisirikul, V.; Phongpaichit, S.; Sakayaroj, J.; Kanjanasirirat, P.; Borwornpinyo, S.; Akrimajirachoote, N.; Yimnual, C.; Muanprasat, C. Depsides and depsidones from the soil-derived fungus Aspergillus unguis PSU-RSPG204. Tetrahedron 2018, 74, 5691–5699. [Google Scholar] [CrossRef]
  26. Li, Y.L.; Gao, Y.; Liu, C.Y.; Sun, C.J.; Zhao, Z.T.; Lou, H.X. Asperunguisins A-F, Cytotoxic Asperane Sesterterpenoids from the Endolichenic Fungus Aspergillus unguis. J. Nat. Prod. 2019, 82, 1527–1534. [Google Scholar] [CrossRef]
  27. Nielsen, J.; Nielsen, P.H.; Frisvad, J.C. Fungal depside, guisinol, from a marine derived strain of Emericella unguis. Phytochemistry 1999, 50, 263–265. [Google Scholar] [CrossRef]
  28. Yang, W.C.; Bao, H.Y.; Liu, Y.Y.; Nie, Y.Y.; Yang, J.M.; Hong, P.Z.; Zhang, Y. Depsidone Derivatives and a Cyclopeptide Produced by Marine Fungus Aspergillus unguis under Chemical Induction and by Its Plasma Induced Mutant. Molecules 2018, 23, 2245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Sureram, S.; Wiyakrutta, S.; Ngamrojanavanich, N.; Mahidol, C.; Ruchirawat, S.; Kittakoop, P. Depsidones, aromatase inhibitors and radical scavenging agents from the marine-derived fungus Aspergillus unguis CRI282-03. Planta Med. 2012, 78, 582–588. [Google Scholar] [CrossRef]
  30. Morshed, M.T.; Vuong, D.; Crombie, A.; Lacey, A.E.; Karuso, P.; Lacey, E.; Piggott, A.M. Expanding antibiotic chemical space around the nidulin pharmacophore. Org. Biomol. Chem. 2018, 16, 3038–3051. [Google Scholar] [CrossRef]
  31. Phainuphong, P.; Rukachaisirikul, V.; Phongpaichit, S.; Preedanon, S.; Sakayaroj, J. Diphenyl ethers and indanones from the soil-derived fungus Aspergillus unguis PSU-RSPG204. Tetrahedron 2017, 73, 5920–5925. [Google Scholar] [CrossRef]
  32. Cao, V.A.; Kwon, J.H.; Kang, J.S.; Lee, H.S.; Heo, C.S.; Shin, H.J.; Aspersterols, A.-D. Ergostane-Type Sterols with an Unusual Unsaturated Side Chain from the Deep-Sea-Derived Fungus Aspergillus unguis. J. Nat. Prod. 2022, 85, 2177–2183. [Google Scholar] [CrossRef]
  33. Kamal, A.; Haider, Y.; Khan, Y.A.; Qureshi, I.H.; Qureshi, A.A. Studies in the biochemistry of microorganisms. Part X. Isolation, structure and stereochemistry of yasminin and other metabolic products of Aspergillus unguis Emile-Weil and Gaudin. Pakistan J. Sci. Ind. Res. 1970, 13, 244–250. [Google Scholar]
  34. Zhang, Y.; Mu, J.; Feng, Y.; Wen, L.; Han, J. Four chlorinated depsidones from a seaweed-derived strain of Aspergillus unguis and their new biological activities. Nat. Prod. Res. 2014, 28, 503–506. [Google Scholar] [CrossRef]
  35. Sadorn, K.; Saepua, S.; Bunbamrung, N.; Boonyuen, N.; Komwijit, S.; Rachtawee, P.; Pittayakhajonwut, P. Diphenyl ethers and depsidones from the endophytic fungus Aspergillus unguis BCC54176. Tetrahedron 2022, 105, 132612. [Google Scholar] [CrossRef]
  36. Anh, C.V.; Kwon, J.H.; Kang, J.S.; Lee, H.S.; Heo, C.S.; Shin, H.J. Antibacterial and Cytotoxic Phenolic Polyketides from Two Marine-Derived Fungal Strains of Aspergillus unguis. Pharmaceuticals 2022, 15, 74. [Google Scholar] [CrossRef] [PubMed]
  37. Handayani, D.; Rendowati, A.; Aminah, I.; Ariantari, N.P.; Proksch, P. Bioactive compounds from marine sponge derived fungus Aspergillus unguis WR8. Rasayan. J. Chem. 2020, 13, 2633–2638. [Google Scholar] [CrossRef]
  38. Kawahara, N.; Nakajima, S.; Satoh, Y.; Yamazaki, M.; Kawai, K.-c. Studies on fungal products. XVIII.: Isolation and structures of a new fungal depsidone related to nidulin and a new phthalide from Emericella unguis. Chem. Pharm. Bull. 1988, 36, 1970–1975. [Google Scholar] [CrossRef]
  39. Kawahara, N.; Nozawa, K.; Nakajima, S.; Kawai, K. Isolation and structures of novel fungal depsidones, emeguisins, A, B, and C, from Emericella unguis. J. Chem. Soc. Perkin Trans. 1988, 1, 2611–2614. [Google Scholar] [CrossRef]
  40. Klaiklay, S.; Rukachaisirikul, V.; Aungphao, W.; Phongpaichit, S.; Sakayaroj, J. Depsidone and phthalide derivatives from the soil-derived fungus Aspergillus unguis PSU-RSPG199. Tetrahedron. Lett. 2016, 57, 4348–4351. [Google Scholar] [CrossRef]
  41. Saetang, P.; Rukachaisirikul, V.; Phongpaichit, S.; Preedanon, S.; Sakayaroj, J.; Hadsadee, S.; Jungsuttiwong, S. Antibacterial and Antifungal Polyketides from the Fungus Aspergillus unguis PSU-MF16. J. Nat. Prod. 2021, 84, 1498–1506. [Google Scholar] [CrossRef]
  42. Sureram, S.; Kesornpun, C.; Mahidol, C.; Ruchirawat, S.; Kittakoop, P. Directed biosynthesis through biohalogenation of secondary metabolites of the marine-derived fungus Aspergillus unguis. RSC Adv. 2013, 3, 1781–1788. [Google Scholar] [CrossRef]
  43. Turner, W.B.; Aldridge, D.C. Fungal Metabolites II; Academic Press: London, UK, 1983. [Google Scholar]
  44. Yang, J.; Zhou, L.; Zhou, Z.; Song, Y.; Ju, J. Anti-pathogenic depsidones and its derivatives from a coral-derived fungus Aspergillus sp. SCSIO SX7S7. Biochem. Syst. Ecol. 2022, 102, 104415. [Google Scholar] [CrossRef]
  45. Dean, F.M.; Roberts, J.C.; Robertson, A. The chemistry of fungi. Part XXII. Nidulin and nornidulin (“ustin”): Chlorine-containing metabolic products of Aspergillus nidulans. J. Chem. Soc. 1954, 1432–1439. [Google Scholar] [CrossRef]
  46. Zhang, Y.; Li, Z.; Huang, B.; Liu, K.; Peng, S.; Liu, X.; Gao, C.; Liu, Y.; Tan, Y.; Luo, X. Anti-Osteoclastogenic and Antibacterial Effects of Chlorinated Polyketides from the Beibu Gulf Coral-Derived Fungus Aspergillus unguis GXIMD 02505. Mar. Drugs 2022, 20, 178. [Google Scholar] [CrossRef] [PubMed]
  47. Uchida, R.; Nakajyo, K.; Kobayashi, K.; Ohshiro, T.; Terahara, T.; Imada, C.; Tomoda, H. 7-Chlorofolipastatin, an inhibitor of sterol O-acyltransferase, produced by marine-derived Aspergillus ungui NKH-007. J. Antibiot. 2016, 69, 647–651. [Google Scholar] [CrossRef] [PubMed]
  48. Feighner, S.D.; Salituro, G.M.; Smith, J.L.; Tsou, N.N. Unguinol and Analogs as Animal Growth Promoters. U.S. Patent 5350763A, 1994. [Google Scholar]
  49. Motti, C.A.; Bourne, D.G.; Burnell, J.N.; Doyle, J.R.; Haines, D.S.; Liptrot, C.H.; Llewellyn, L.E.; Ludke, S.; Muirhead, A.; Tapiolas, D.M. Screening marine fungi for inhibitors of the C4 plant enzyme pyruvate phosphate dikinase: Unguinol as a potential novel herbicide candidate. Appl. Environ. Microbiol. 2007, 73, 1921–1927. [Google Scholar] [CrossRef] [Green Version]
  50. Chottanapund, S.; Van Duursen, M.B.M.; Zwartsen, A.; Timtavorn, S.; Navasumrit, P.; Kittakoop, P.; Sureram, S.; Ruchirawat, M.; Van den Berg, M. Depsidones inhibit aromatase activity and tumor cell proliferation in a co-culture of human primary breast adipose fibroblasts and T47D breast tumor cells. Toxicol. Rep. 2017, 4, 165–171. [Google Scholar] [CrossRef]
  51. Zwartsen, A.; Chottanapund, S.; Kittakoop, P.; Navasumrit, P.; Ruchirawat, M.; Van Duursen, M.B.M.; Van den Berg, M. Evaluation of anti-tumour properties of two depsidones—Unguinol and Aspergillusidone D—In triple-negative MDA-MB-231 breast tumour cells. Toxicol. Rep. 2019, 6, 1216–1222. [Google Scholar] [CrossRef]
  52. Hamano, K.; Kinoshita-Okami, M.; Hemmi, A.; Sato, A.; Hisamoto, M.; Matsuda, K.; Yoda, K.; Haruyama, H.; Hosoya, T.; Tanzawa, K. Folipastatin, a new depsidone compound from Aspergillus unguis as an inhibitor of phospholipase A2. Taxonomy, fermentation, isolation, structure determination and biological properties. J. Antibiot. 1992, 45, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
  53. Kamal, A.; Haider, Y.; Qureshi, A.A.; Khan, Y.A. Studies in the biochemistry of microorganisms XII. Isolation and structures of haiderin, rubinin, shirin and nasrin metabolic products of Aspergillus unguis Emile-Weil and Gaudin. Pak. J. Sci. Ind. Res. 1970, 13, 364–372. [Google Scholar]
  54. Kamal, A.; Haider, Y.; Qureshi, A.A.; Wahid, M.A.; Khan, Y.A. Studies in the biochemistry of microorganisms. Part XIII. Biosynthesis of yasimin, a metabolic product of Aspergillus unguis. Pak. J. Sci. Ind. Res. 1970, 13, 373–377. [Google Scholar]
  55. Malmstrom, J. Unguisins A and B: New cyclic peptides from the marine-derived fungus Emericella unguis. J. Nat. Prod. 1999, 62, 787–789. [Google Scholar] [CrossRef] [PubMed]
  56. Stodola, F.H.; Vesonder, R.F.; Fennell, D.I.; Weisleder, D. A new depsidone from Aspergillus unguis. Phytochemistry 1972, 11, 2107–2108. [Google Scholar] [CrossRef]
  57. Sierankiewicz, J.; Gatenbeck, S. A new depsidone from Aspergillus nidulans. Acta Chem. Scand. 1972, 26, 455–458. [Google Scholar] [CrossRef] [Green Version]
  58. Maha, A.; Phainuphong, P.; Rukachaisirikul, V.; Saithong, S.; Phongpaichit, S.; Hadsadee, S.; Jungsuttiwong, S.; Preedanon, S.; Sakayaroj, J. Blennolide derivatives from the soil-derived fungus Trichoderma asperellum PSU-PSF14. Tetrahedron 2018, 74, 5659–5664. [Google Scholar] [CrossRef]
  59. Hosoe, T.; Gloer, J.B.; Wicklow, D.T.; Raja, H.A.; Shearer, C.A. New nonadride analogues from a freshwater isolate of an undescribed fungus belonging to the order Pleosporales. Heterocycles 2010, 81, 2123–2130. [Google Scholar] [CrossRef]
  60. Okoye, F.B.C.; Nworu, C.S.; Akah, P.A.; Esimone, C.O.; Debbab, A.; Proksch, P. Inhibition of inflammatory mediators and reactive oxygen and nitrogen species by some depsidones and diaryl ether derivatives isolated from Corynespora cassiicola, an endophytic fungus of Gongronema latifolium leaves. Immunopharmacol Immunotoxicol. 2013, 35, 662–668. [Google Scholar] [CrossRef]
  61. Zhang, S.; Chen, D.; Kuang, M.; Peng, W.; Chen, Y.; Tan, J.; Kang, F.; Xu, K.; Zou, Z. Rhytidhylides A and B, Two New Phthalide Derivatives from the Endophytic Fungus Rhytidhysteron sp. BZM-9. Molecules 2021, 26, 6092. [Google Scholar] [CrossRef]
  62. Anh, C.V.; Kang, J.S.; Choi, B.K.; Lee, H.S.; Heo, C.S.; Shin, H.J. Polyketides and Meroterpenes from the Marine-Derived Fungi Aspergillus unguis 158SC-067 and A. flocculosus 01NT-1.1.5 and Their Cytotoxic and Antioxidant Activities. Mar. Drugs 2021, 19, 415. [Google Scholar] [CrossRef]
  63. Malmstrom, J.; Ryager, A.; Anthoni, U.; Nielsen, P.H. Unguisin C, a GABA-containing cyclic peptide from the fungus Emericella unguis. Phytochemistry 2002, 60, 869–872. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, S.; Shen, Y. A new cyclic peptide from the marine fungal strain Aspergillus sp. AF119. Chem. Nat. Compd. 2011, 47, 786–788. [Google Scholar] [CrossRef]
  65. Akone, S.H.; Daletos, G.; Lin, W.; Proksch, P. Unguisin F, a new cyclic peptide from the endophytic fungus Mucor irregularis. Z. Naturforsch. C J. Biosci. 2016, 71, 15–19. [Google Scholar] [CrossRef] [PubMed]
  66. Rajachan, O.A.; Kanokmedhakul, S.; Kanokmedhakul, K.; Soytong, K. Bioactive depsidones from the fungus Pilobolus heterosporus. Planta Med. 2014, 80, 1635–1640. [Google Scholar] [CrossRef] [PubMed]
  67. Rao, V.S.; Seshadri, T.R. Chemical examination of Indian lichens. V. Occurrence of active montagnetol in Roccella montagnei. Proceedings 1942, 15, 429–431. [Google Scholar]
  68. Anh, C.V.; Yoon, Y.D.; Kang, J.S.; Lee, H.S.; Heo, C.S.; Shin, H.J. Nitrogen-Containing Secondary Metabolites from a Deep-Sea Fungus Aspergillus unguis and Their Anti-Inflammatory Activity. Mar. Drugs 2022, 20, 217. [Google Scholar] [CrossRef]
  69. Lee, Y.M.; Li, H.; Hong, J.; Cho, H.Y.; Bae, K.S.; Kim, M.A.; Kim, D.K.; Jung, J.H. Bioactive metabolites from the sponge-derived fungus Aspergillus versicolor. Arch. Pharm. Res. 2010, 33, 231–235. [Google Scholar] [CrossRef] [PubMed]
  70. Abdou, R.; Scherlach, K.; Dahse, H.M.; Sattler, I.; Hertweck, C. Botryorhodines A-D, antifungal and cytotoxic depsidones from Botryosphaeria rhodina, an endophyte of the medicinal plant Bidens pilosa. Phytochemistry 2010, 71, 110–116. [Google Scholar] [CrossRef]
  71. Cane, D.E.; Kudo, F.; Kinoshita, K.; Khosla, C. Precursor-directed biosynthesis: Biochemical basis of the remarkable selectivity of the erythromycin polyketide synthase toward unsaturated triketides. Chem. Biol. 2002, 9, 131–142. [Google Scholar] [CrossRef] [Green Version]
  72. Duong, T.H.; Huynh, B.L.C.; Chavasiri, W.; Chollet-Krugler, M.; Nguyen, V.K.; Nguyen, T.H.T.; Hansen, P.E.; Le Pogam, P.; Thus, H.; Boustie, J.; et al. New erythritol derivatives from the fertile form of Roccella montagnei. Phytochemistry 2017, 137, 156–164. [Google Scholar] [CrossRef]
  73. Cueto, M.; Jensen, P.R.; Fenical, W. Aspergilloxide, a novel sesterterpene epoxide from a marine-derived fungus of the genus Aspergillus. Org. Lett. 2002, 4, 1583–1585. [Google Scholar] [CrossRef] [PubMed]
  74. Isaka, M.; Yangchum, A.; Supothina, S.; Veeranondha, S.; Komwijit, S.; Phongpaichit, S. Semisynthesis and antibacterial activities of nidulin derivatives. J. Antibiot. 2019, 72, 181–184. [Google Scholar] [CrossRef]
  75. Wei, Q.; Bai, J.; Yan, D.; Bao, X.; Li, W.; Liu, B.; Zhang, D.; Qi, X.; Yu, D.; Hu, Y. Genome mining combined metabolic shunting and OSMAC strategy of an endophytic fungus leads to the production of diverse natural products. Acta Pharm. Sin. B 2021, 11, 572–587. [Google Scholar] [CrossRef] [PubMed]
  76. Lu, Q.P.; Huang, Y.M.; Liu, S.W.; Wu, G.; Yang, Q.; Liu, L.F.; Zhang, H.T.; Qi, Y.; Wang, T.; Jiang, Z.K.; et al. Metabolomics Tools Assisting Classic Screening Methods in Discovering New Antibiotics from Mangrove Actinomycetia in Leizhou Peninsula. Mar. Drugs 2021, 19, 688. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Depsidones (1–8, 10–14, 21–23, 26–28, 32, 35, 39, 45–46, 48–50, 69–70, 83–84, 86, 88), depsides (15, 33, 47, 51–54, 85, 87, 93), phthalides (9, 34, 59, 71–72), cyclopeptides (16–20), nitrogen-containing compounds (90–91), indanones (40–41), diarylethers (25, 41–43, 55–57, 67–68, 79–82), pyrones (24), benzoic acid derivative (31), orcinol/orsenillate derivatives (29–30, 36–38, 44, 66, 73–74, 89, 92), anthraquinones (75–77), chromone (78), sesterpenoids (59–65) and sterols (94–97) from A. unguis.
Figure 1. Depsidones (1–8, 10–14, 21–23, 26–28, 32, 35, 39, 45–46, 48–50, 69–70, 83–84, 86, 88), depsides (15, 33, 47, 51–54, 85, 87, 93), phthalides (9, 34, 59, 71–72), cyclopeptides (16–20), nitrogen-containing compounds (90–91), indanones (40–41), diarylethers (25, 41–43, 55–57, 67–68, 79–82), pyrones (24), benzoic acid derivative (31), orcinol/orsenillate derivatives (29–30, 36–38, 44, 66, 73–74, 89, 92), anthraquinones (75–77), chromone (78), sesterpenoids (59–65) and sterols (94–97) from A. unguis.
Biomolecules 12 01820 g001aBiomolecules 12 01820 g001bBiomolecules 12 01820 g001cBiomolecules 12 01820 g001dBiomolecules 12 01820 g001eBiomolecules 12 01820 g001fBiomolecules 12 01820 g001gBiomolecules 12 01820 g001h
Table 1. All the compounds identified in A. unguis were organized by code, showing for each one (I) the references that report the isolation and identification; (II) the chemical class to which they belong and (III) the biological activities of each compound.
Table 1. All the compounds identified in A. unguis were organized by code, showing for each one (I) the references that report the isolation and identification; (II) the chemical class to which they belong and (III) the biological activities of each compound.
CodeCompoundBiological ActivitiesIsolation and Identification
Depsidones
1nidulinInhibition of xanthine oxidase [29]; aromatase inhibitory activity [29]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (weak cytotoxicity) [29]; anti-MRSA and brine shrimp larva lethality test strong bioactivity [34]; DNA-damage repair test (anti-AB3027) [34]; antimicrobial activity against BS and SA [30]; antimicrobial activity against SA and MRSA [25]; larvicidal activity using brine shrimp model [28]; antimicrobial activity against BC [35]; anti-phytopathogenic activity against CA [35]; growth inhibition against human cancer cell lines [36][25,28,29,30,33,34,35,36,37,38,39,40,41,42,43,44] also isolated from A. nidulans [45]
2nornidulinInhibition of xanthine oxidase [29]; aromatase inhibitory activity [29]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (weak cytotoxicity) [29]; anti-MRSA and brine shrimp larva lethality test [34]; DNA-damage repair test (anti-AB3027) [34]; antimicrobial in SA, MRSA, and CN [42]; antimicrobial activity against BS and SA [30]; antimicrobial activity against SA, MRSA and CN [25]; larvicidal activity/brine shrimp model [28]; antimicrobial activity against BC [35]. anti-phytopathogenic activity against AB and CA [35]; growth inhibition against human cancer cell lines [36]; inhibition effect on LPS-induced NF-kβ activation [46]. Antimicrobial activity against MRSA, MV, MJ and VP [46]; antimicrobial activity against Gram-positive and Gram-negative bacteria [41][25,27,28,29,30,33,34,35,36,40,41,42,43,44,46,47] also isolated from A. nidulans [45]
3unguinol (yasimin)Animal growth permittant [48]; pyruvate phosphate dikinase (PPDK) inhibitor (a potential herbicide target) [49]; aromatase inhibitory activity [37]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (weak cytotoxicity) [37]; KB, MCF-7 and Vero cancer cell line (cytotoxicity) [42]; T47D tumor cells most likely via inhibition of aromatase (CYP19) activity [50]; SOAT1 and SOAT2 isozymes inhibition [47]; antimicrobial activity against BS [30]; apoptosis induction and cell cycle arrest in MDA-MB-231 cells induction [51]; antimicrobial activity against MT [35]; anti-phytopathogenic activity against AB [35]; growth inhibition against human cancer cell lines [36]; inhibition effect on LPS-induced NF-kβ activation [46][25,28,30,33,35,36,38,39,40,41,42,43,46,47,48,49,50,52,53,54,55,56] also isolated from Aspergillus nidulans and Trichoderma asperellum [57,58]
4haiderin-[53]
5rubinin-[53]
6(-)-shirin-[53]
7nasrin-[53]
82-chlorounguinolInhibition of xanthine oxidase [29]; aromatase inhibitory activity [29]; antimicrobial in SA, MRSA, CA, CN and MG [42]; SOAT1 and SOAT2 isozymes inhibition [47]; antimicrobial activity against SA, MRSA, CA, and MG [25]; antimicrobial activity against PA and MRSA [28]; larvicidal activity/brine shrimp model [28]; antimicrobial activity against MT, BC [35]. Anti-phytopathogenic activity against AB and CA [35]; growth inhibition against human cancer cell lines [36][25,28,29,30,35,36,38,39,40,41,42,44,47,48] also isolated from Trichoderma asperellum [58]
10emeguisin A/7-chlorofolipastatinAntimicrobial in SA, MRSA and CN [42]; antimalarial activity against PF [35,42]; antimicrobial activity against BS, SA and SC [30]; antimicrobial activity against SA, MRSA, CA and CN [25]; cytotoxic activity against KB and Vero cells [25]; SOAT1 and SOAT2 isozymes inhibition [47]; antimicrobial activity against MT, BC [35]; anti-phytopathogenic activity against CA [35]; growth inhibition against human cancer cell lines [36]; antimicrobial activity against Gram-positive and Gram-negative bacteria [41][25,30,35,36,39,40,41,44]
11emeguisin BAntimalarial activity against PF [35][35,39,41,44]
12emeguisin CAntimalarial activity against PF [35]; Antimicrobial activity against MT, BC [35][35,39]
13folipastatinPhospholipase A inhibitor [55]; antimicrobial in SA, MRSA, and CN [42]; antimalarial activity against PF [35,42]; SOAT1 and SOAT2 isozymes inhibition [47]; antimicrobial activity against BS and SA [30]; cytotoxity against NS-1 [30]; antimicrobial activity against SA, MRSA and CN [25]; cytotoxic activity against Vero cells [25]; growth inhibition against human cancer cell lines [36]; antimicrobial activity against Gram-positive and Gram-negative bacteria [41][25,30,35,36,40,41,44,47,48,52] also isolated from Wicklowia aquatica [59]
144-methylunguinol-[48]
21aspergillusidone AInhibition of superoxide anion radical formation by xanthine/xanthine oxidase [29]; aromatase inhibitory activity [29]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (only weak cytotoxicity) [29]; T47D tumor cells most likely via inhibition of aromatase (CYP19) activity [50]; antimicrobial activity against MRSA and CA [28]. AChE inhibitory activity [28]. Larvicidal activity using brine shrimp model [28]; anti-phytopathogenic activity against CA [35][25,28,29,35,41,44,50]
22aspergillusidone BInhibition of superoxide anion radical formation by xanthine/xanthine oxidase [29]; aromatase inhibitory activity [29]; antimalarial activity against PF [35]; antimicrobial activity against BC [35]; inhibition effect on LPS-induced NF-kβ activation [46][25,29,35,44,46]
23aspergillusidone C(2,7-dichlorounginol)Aromatase inhibitory activity [29]; inhibition of xanthine oxidase [29]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (only weak cytotoxicity) [29]; anti-A549 tumor cell line [34]; anti-MRSA and brine shrimp larva lethality test strong bioactivity [34]; DNA-damage repair test (anti-AB3027) [34]; antimicrobial in SA, MRSA, CA, CN and MG [42]; antimalarial [42]; VERO cell line (strong cytotoxicity) [42]; antimicrobial activity against BS, SA and SC [30]; antimicrobial activity against SA, MRSA, and MG [25]. Cytotoxic activity against Vero cells [25]; antimicrobial activity against PA [28]. Larvicidal activity using brine shrimp model [28]; antimicrobial activity against MT, BC [35]; anti-phytopathogenic activity against AB and CA [35]; growth inhibition against human cancer cell lines [36]; antimicrobial activity against MRSA, MV, MJ and VP [46];[25,28,29,30,34,35,36,40,41,44,46,47] also isolated from Trichoderma asperellum [58]
26aspergillusidone DAromatase inhibitory activity [37]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (only weak cytotoxicity) [37]; antimicrobial activity against BS, SA and SC [30]; apoptosis induction and cell cycle arrest in MDA-MB-231 cells induction [51][30,42]
27aspergillusidone EAromatase inhibitory activity [37]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (only weak cytotoxicity) [37]; antimicrobial activity against BS, SA and SC [30][30,42]
28aspergillusidone FAromatase inhibitory activity [37]; HuCCA-1, HepG2, A549, MOLT-3 cancer cell line (only weak cytotoxicity) [37]; antimicrobial activity against BS, SA and SC [30]; antimicrobial activity against PA and MRSA [28]. Larvicidal activity using brine shrimp model [28].[28,30,42]
322,4-dichlorounguinolAntimicrobial activity against SA and MRSA [25][25,42]
35aspersidoneAntimicrobial activity against SA and MRSA [25]. Cytotoxic activity against Vero cells [25]; antimicrobial activity against BC [35]; anti-phytopathogenic activity against AB and CA [35]; growth inhibition against human cancer cell lines [36][25,35,36,40,41]
457-carboxifolipastatin-[30,44]
464,7-dichlorounguinolAntimicrobial activity against BS, SA and SC [30][30]
487-bromounguinolAntimicrobial activity against BS, SA and SC [30][30]
492-chloro-7-bromounguinolAntimicrobial activity against BS, SA and SC [30][30]
507-bromofolipastatinAntimicrobial activity against BS and SA [30][30]
69asperunguissidone AAntimicrobial activity against SA and MRSA [38][41]
70asperunguissidone B-[41]
83emeguisin DAntimalarial activity against PF [35]; antimicrobial activity against MT, BC and SA [35];[35]
84corynesidone DAntimicrobial activity against BC [35];[35] also isolated from Corynespora cassiicola [60]
86aspersidone BAntimicrobial activity against BS, ML and SA [36]; growth inhibition against human cancer cell lines [36][36]
88aspergillusidone HInhibition effect on LPS-induced NF-kβ activation [46][46]
Depsides
15guisinolGrowth inhibition against human cancer cell lines [36]; antimicrobial activity against MRSA, MV, MJ and VP [46][27,36,46]
33agonodepside AGrowth inhibition against human cancer cell lines [36];[28,36,41,42,44]
47agonodepside B Antimicrobial activity against BS [30]; growth inhibition against human cancer cell lines [36][30,36]
515-bromoagonodepside BAntimicrobial activity against BS [30][30]
52aspergiside B/unguidepside A/aspergillusidone G-[25,30,41,44]
533-bromounguidepside A-[30]
54aspergiside A/decarboxyunguidepside AAntimicrobial activity against BS [30];antimicrobial activity against SA and MRSA [25]; growth inhibition against human cancer cell lines [36][25,30,36,41]
85unguidepside CAntimicrobial activity against BS, ML and SA [36]; growth inhibition against human cancer cell lines [36][36]
87agonodepside CAntimicrobial activity against BS, ML and SA [36][36]
93asperdepside A-[44]
Phthalides
9*(3S)-3-ethyl-5,7-dihydroxy-3,6-dimethylphthalideAntimicrobial in SA, MRSA, CA, CN and MG [42]; antimalarial [42]; HB carcinoma cell line (strong cytotoxicity) [42]; antimicrobial activity against SA, MRSA, and MG [25][25,38,40,41,42] also isolated from Rhytidhysteron sp. [61]
34asperlide-[40,41]
58aspergiside C -[25,41]
71asperunguislide A-[41]
72asperunguislide B-[41]
Cyclopeptides
16unguisin A-[28,55,62]
17unguisin B-[55]
18unguisin C-[63]
19unguisin D-[63]
20unguisin E-[55] also isolated from Mucor irregularis and Aspergillus candidus [64,65]
Nitrogen-containing Compounds
90variotin BAnti-inflamatory activity [62][62]
91coniosulfide E-[62]
Indanones
39asperunguisone A -[31,41]
40asperunguisone B-[31,41]
Diarylethers
25aspergillusether AHuCCA-1, HepG2, A549, MOLT-3 cancer cell line (weak cytotoxicity) [29][29,34]
41aspergillusether B-[31]
42aspergillusether C-[31,41]
43aspergillusether DAntimicrobial activity against CA, CN and PM [31][31,35,41]
55unguinolic acid-[30]
56decarboxyunguinolic acid-[30]
575-chlorounguinolic acid [30]
67aspergillusether EAntimicrobial activity against SA, MRSA, CA, CN and MG [38]; cytotoxic activity against Vero cells [38][41]
68aspergillusether FAntimicrobial activity against BC [35]; anti-phytopathogenic activity against CA [35]; inhibition effect on LPS-induced NF-kβ activation [46]. Antimicrobial activity against MRSA, MV, MJ and VP [46][35,41,46]
79aspergillusether G-[35]
80aspergillusether HAntimicrobial activity against MT, BC and SA [35];[35]
81aspergillusether IAntimicrobial activity against BC and SA [35]; Anti-phytopathogenic activity against CA [35];[35]
82aspergillusether JAntimicrobial activity against MT, BC and SA [35]; Anti-phytopathogenic activity against CA [35]; Inhibition effect on LPS-induced NF-kβ activation [46]. Antimicrobial activity against MRSA, MV, MJ and VP [46]; [35,46]
Pyrone
243-methyl-4-hydroxy-6-(1-trans-methyl-1-propenyl)-2-pyroneAntimicrobial activity against MT [35];[29,35,41]
Orcinol/Orsenillate Derivatives/Phloroglucinol
29aspergillusphenol AAntimicrobial activity against SA and MRSA [31] [29,31,40,41,44]
30aspergillusphenol B-[42]
36methyl orsellinate-[31,40,41]
37pilobolusateAntimicrobial in SA, MRSA, CA, CN and MG [42]; antimalarial [42]; KB cell line (cytotoxicity) [31][31,40,41] also isolated from Pilobolus heterosporus [66]
38(+)-montagnetol-[31,40,41] also isolated from Roccella montagnei [67]
44orcinol-[31,41]
66aspergillusphenol C-[41]
73grifolin A-[68]
74grifolin BDPPH radical scavenging activity [68][68]
891-(2,6-dihydroxy-4-methoxy-3,5-dimethylphenyl)-methylbutan-1-oneInhibition effect on LPS-induced NF-kβ activation [46]; antimicrobial activity against MV, MJ and VP [46] [46]
92aspergillusphenol A carboxylic acid-[44]
Benzoic Acid Derivatives
313,5-dibromo-2,4-dihydroxy-6-methyl-benzoic acid methyl ester-[42]
Anthraquinones
75*averantin-[68] also isolated from Aspergillus versicolor [69]
767-chloroaveratin-[68]
771′-O-methylaveration-[68]
Chromone
787-hydroxy-2-(2-hydroxypropyl-5-pentyl-chromone-[68]
Sesterterpenoids
59asperunguisin ACytotoxicity against A549 and HepG2 human cancer cells [26][26]
60asperunguisin BCytotoxicity against SMMC-7721 human cancer cells [26][26]
61asperunguisin CCytotoxicity against HT-29, A549, U251, U87, SMMC-7721 and HepG2 human cancer cells [26][26]
62asperunguisin D-[26]
63asperunguisin E-[26]
64asperunguisin F-[26]
65aspergilloxide-[26]
Sterols
94aspersterol ACytotoxicity against cancer cell lines [32][32]
95aspersterol BAnti-inflammatory activity [32][32]
96aspersterol CAnti-inflammatory activity [32][32]
97aspersterol DAnti-inflammatory activity [32][32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Domingos, L.T.S.; Martins, R.d.S.; Lima, L.M.d.; Ghizelini, A.M.; Ferreira-Pereira, A.; Cotinguiba, F. Secondary Metabolites Diversity of Aspergillus unguis and Their Bioactivities: A Potential Target to Be Explored. Biomolecules 2022, 12, 1820. https://doi.org/10.3390/biom12121820

AMA Style

Domingos LTS, Martins RdS, Lima LMd, Ghizelini AM, Ferreira-Pereira A, Cotinguiba F. Secondary Metabolites Diversity of Aspergillus unguis and Their Bioactivities: A Potential Target to Be Explored. Biomolecules. 2022; 12(12):1820. https://doi.org/10.3390/biom12121820

Chicago/Turabian Style

Domingos, Levy Tenório Sousa, Raquel dos Santos Martins, Leonardo Melo de Lima, Angela Michelato Ghizelini, Antonio Ferreira-Pereira, and Fernando Cotinguiba. 2022. "Secondary Metabolites Diversity of Aspergillus unguis and Their Bioactivities: A Potential Target to Be Explored" Biomolecules 12, no. 12: 1820. https://doi.org/10.3390/biom12121820

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