Next Article in Journal
Profiling Colorectal Cancer in the Landscape Personalized Testing—Advantages of Liquid Biopsy
Next Article in Special Issue
Candida auris: A Quick Review on Identification, Current Treatments, and Challenges
Previous Article in Journal
Impaired 26S Proteasome Assembly Precedes Neuronal Loss in Mutant UBQLN2 Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Highlight on the Inhibition of Fungal Carbonic Anhydrases as Drug Targets for the Antifungal Armamentarium

by
Claudiu T. Supuran
1,* and
Clemente Capasso
2,*
1
Section of Pharmaceutical and Nutraceutical Sciences, Department of Neurofarba, University of Florence, Via U. Schiff 6, Sesto Fiorentino, 50019 Florence, Italy
2
Institute of Biosciences and Bioresources, CNR, Via Pietro Castellino 111, 80131 Napoli, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(9), 4324; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094324
Submission received: 9 April 2021 / Revised: 19 April 2021 / Accepted: 19 April 2021 / Published: 21 April 2021

Abstract

:
Carbon dioxide (CO2), a vital molecule of the carbon cycle, is a critical component in living organisms’ metabolism, performing functions that lead to the building of compounds fundamental for the life cycle. In all living organisms, the CO2/bicarbonate (HCO3) balancing is governed by a superfamily of enzymes, known as carbonic anhydrases (CAs, EC 4.2.1.1). CAs catalyze the pivotal physiological reaction, consisting of the reversible hydration of the CO2 to HCO3 and protons. Opportunistic and pathogenic fungi can sense the environmental CO2 levels, which influence their virulence or environmental subsistence traits. The fungal CO2-sensing is directly stimulated by HCO3 produced in a CA-dependent manner, which directly activates adenylyl cyclase (AC) involved in the fungal spore formation. The interference with CA activity may impair fungal growth and virulence, making this approach interesting for designing antifungal drugs with a novel mechanism of action: the inhibition of CAs linked to the CO2/HCO3/pH chemosensing and signaling. This review reports that sulfonamides and their bioisosteres as well as inorganic anions can inhibit in vitro the β- and α-CAs from the fungi, suggesting how CAs may be considered as a novel “pathogen protein” target of many opportunistic, pathogenic fungi.

1. Introduction

The fossil fuel use, land-use changes as well as the natural carbon sources on land and in oceans has drastically influenced the growth rate of atmospheric CO2 [1]. In the last twenty years, human CO2 emissions have been enormously accelerated, considering the overall rise in energy consummation, the greater use of coal to produce energy, increased consumption per capita, and population development [1]. Changes in atmospheric CO2 mirrors the balance between carbon emissions due to human activity and the dynamics of many terrestrial and ocean processes that remove or emit CO2 [2]. The increased CO2 favors the photosynthetic activity of plants and increases carbon storage in the plants themselves and soil [2]. Cheng et al. believe that carbon storage seems to be mainly due to fungi (the so-called Arbuscular Mycorrhizal Fungi), which settle near the roots of about 80% of plant species, providing essential nutrients to the plants in exchange for carbohydrates [3]. CO2, a key molecule of the carbon cycle, is a critical component in the metabolism of living organisms, performing functions which lead to the building of compounds fundamental for the life cycle [4]. At the same time, CO2 is a waste product since it is the end-product of respiration, reaching a concentration of about 5% in the human bloodstream and tissues. This concentration is higher than the level of CO2 in the atmosphere (about 0.036%) [4]. Intriguingly, opportunistic and pathogenic fungi sense the CO2 difference, which influences fungal differentiation, determining the expression of those fungal features essential for virulent or non-virulent traits [5]. Pathogenic fungi are responsible for superficial diseases such as dermatophytes (infections of skin, hairs, and nails), or may lead to systemic illness (candidiasis, aspergillosis, cryptococcosis, mucormycosis, and others) [6,7,8,9]. Two molecules are crucial for the fungal CO2-sensing: (1) bicarbonate (HCO3), which is a meiosis- and sporulation-promoting ion [10], and (2) adenylyl cyclase (AC) that is involved in the spore formation [11,12,13]. In Cryptococcus, bicarbonate directly activates a soluble form of AC necessary for the polysaccharide capsule formation [14,15,16,17]. AC catalyzes cyclic AMP (cAMP) synthesis, an essential intracellular regulatory molecule, which permits a link between CO2/HCO3/pH chemosensing and signaling [18]. cAMP signaling is involved in many metabolic reactions as well as in fungal development and virulence [19]. The fungal virulence of Cryptococcus neoformans, the etiological agent responsible for cryptococcosis [9], is induced by high CO2 levels in mammalian hosts, causing the production of a massive polysaccharide capsule, which inhibits phagocytosis and impairs cell-mediated immune response [14,15,20]. However, it also true that other factors than high CO2 levels contribute to inhibit mating in the host as demonstrated by the use of a murine model of cryptococcosis. The carbonic anhydrase mutants for Can1 and Can2 (the two CAs encoded by the genome of C. neoformans) were both as virulent as wild type (wt), and quantitative measurements of fungal burden demonstrated that the Can2 mutant proliferates equivalently to the wt strain in the lungs and brain of infected animals [20]. In Candida albicans, the CO2 levels, through the relationship of bicarbonate, adenyl cyclase and cAMP, influence the growth of filamentous structures (hyphae), which are associated with the fungal virulence, adherence, secretion of hydrolases, and cell death in the hosts [19,21,22,23]. Twenty-six thiazolidines against several Candida spp. and Gram-positive and Gram-negative bacteria were tested. Although lacking significant antibacterial activity, the tested compounds exhibited selective antifungal activity with an equal potency to fluconazole and clotrimazole. Interestingly, CA was considered the putative target that could mediate the antifungal effects of these compounds [24].

Fungal Enzymes Involved in the Bicarbonate Production (Physiological Role and Structural Features)

Cloning of the genomes of several pathogenic and non-pathogenic fungi provided the opportunity to identify a superfamily of ubiquitous metalloenzymes, known as carbonic anhydrases (CAs, EC 4.2.1.1), which catalyze a pivotal physiological reaction, consisting of the reversible hydration of the carbon dioxide to bicarbonate and protons [25,26,27,28,29,30,31]. The spontaneous reversible CO2 hydration reaction in the absence of the catalyst occurs very slowly with a rate constant of 0.15 s−1, which arrives at 50 s−1 for the reverse reaction of bicarbonate dehydration at the physiological pH [31]. CA increases the velocity of the CO2 hydration reaction up to 104-106-fold [31].
The CA superfamily is ubiquitously distributed in all living organisms and classified into eight CA classes (α, β, γ, δ, ζ, η, θ, and ι). Their distribution is quite varied from plants, animals, bacteria, and archaea. [25,26,27,28,29]. The genome of mammals, for example, encodes only for the α-CA class, of which 15 isoforms have been identified, which accomplish specialized functions in various tissues and organs [32,33,34,35,36]. In plants, α and β-CAs actively participate in photosynthesis and biosynthetic reactions associated with it, as well as in some aforementioned processes [37]. In Bacteria, Archaea, and cyanobacteria, α, β, γ, and ι -CA classes are present. Their role is to balance the CO2/HCO3 concentration ratio and a role in the carbon dioxide fixation [29,30,31,37,38,39]. Marine diatoms encode for α- δ-,ζ-, θ- and ι-CAs, which are involved in carbon dioxide fixation and metabolism [40,41,42]. In protozoa have been detected α- and η-CAs. Probably, the η-CA-class, recently discovered, has a pivotal role in de novo purine/pyrimidine biosynthetic pathways [43].
The fungal CO2-sensing, related to the CO2/HCO3/pH-sensing, is directly stimulated by HCO3 produced in a CA-dependent manner. In the fungal kingdom, the typical CA class identified is represented by β-class, and the majority of fungi encode at least one β-CA [13,44,45]. The genomes of basidiomycetous and hemiascomycetous yeasts encode only for β-CAs. In contrast, most filamentous ascomycetes contain multiple β-CA genes and, in some of them, it is possible to find genes encoding for α-CAs [13,44,45]. Here, some examples demonstrating that CAs are abundant in fungi and yeasts (the last are microscopic fungi consisting of solitary cells that reproduce by budding) as reported in the following examples. Saccharomyces cerevisiae, Candida albicans, and Candida glabrata have only one β-CA, whereas multiple copies of β-CA and α-CA-encoding genes were reported in other fungi [44,45]. Recently, it has been evidenced that CAs play an important role in fungal pathogen sensing and the control of sexual growth [44,45]. The β-CAs identified in Candida albicans and Candida glabrata indicated with the acronyms CaNce103 and CgNce103, respectively, are necessary for the development of these fungi in environments characterized by low-oxygen conditions, such as the skin [44,45]. The CA (Can2) encoded by the genome of Cryptococcus neoformans allows the growth of the yeast in its natural habitat. It is relevant to note how the link between AC, cAMP signaling, and CO2/HCO3 sensing is conserved in most fungi since it is an essential mediator of fungal metabolism and pathogenesis [13,44,45]. Again, the gene Nce103 identified in the genome of Saccharomyces cerevisiae encodes for a β-CA (ScCA), which is involved in the production of the bicarbonate essential for the enzyme catalyzing carboxylation reactions, such as the pyruvate carboxylase (PC), acetyl-CoA carboxylase (ACC), carbamoyl phosphate synthase (CPSase), and phosphoribosylaminoimidazole (AIR) carboxylase [46,47].
In 2009, the first crystal structure of the β-CA encoded in the genome of a fungus, i.e., Cryptococcus neoformans, was reported by Schlicker and coworkers [45]. It showed a dimeric organization similar to that found in the CA belonging to the plant-type β-class (the two cysteines and a histidine responsible for zinc coordination are conserved in the active site of such enzymes). Intriguingly, a Can2 (acronym used for the CA from C. neoformans) three-dimensional structure showed a peculiar N-terminal extension, which interacts with the entrance of the catalytic pocket of the dimer. The N-terminus is an internal regulator or an interaction site for a regulatory protein, affecting the Can2 activity [45]. It can be considered a switch for the activation/inactivation of the protein, which is regulated by physiological factors, like pH, small molecule, or proteins. [45].
In 2011, the structure of the first fungal α-CA was obtained, which was identified in the fungus Aspergillus oryzae [48]. Like for other α-CAs, the enzyme showed a central core formed by a twisted β sheet consisting of eight mostly anti-parallel strands. The ion cofactor resulted in an atom of Zn(II) coordinated to the three histidines of the catalytic pocket, which is at the bottom of a deep cavity in the protein center [48].
In 2014, the structures of two β-CAs belonging to the fungus Sordaria macrospora were resolved by X-ray crystallography [49]. Like Can2, the two β-CAs from S. macrospora showed a high structural similarity with plant-like β-CAs, but it was assembled in a tetrameric and not a dimeric form. The two CAs (CAS1 and CAS2) were distinguished for the type of conformations they assumed: CAS1 resulted in the open “type- I” conformation, while the CAS2 adopted a close “type-II” conformation [49]. Finally, between 2020 and 2021, CafA and CafB, two of the four β-CAs encoded by the genome of the fungus Aspergillus fumigatus, were crystallized and the structure resolved at 1.8 and 2.0 Å, respectively [50,51]. The catalytic sites of CafA and CafB look similar to those of other β-CAs. CafA showed the typical open conformation. Surprisingly, CafB revealed a unique active site at a low pH or in an oxidative environment, resulting in an inactive enzyme, with a disulfide bond formed by the two zinc-ligating cysteines [50]. Of course, CafB also adopts the typical active/inactive configurations in which a conserved aspartic acid is implicated in switching the enzyme in its open/closed state [52].
This review reports the recent kinetic investigations and inhibition profiles obtained for the fungal CAs encoded in the genomes of Sordaria macrospora, Saccharomyces cerevisiae, Candida glabrata, Malassezia restricta, and Malassezia globosa. As described above, it is readily apparent how fungal CAs could play an essential role in the life cycle of opportunistic and pathogenic fungi. It is reasonable to think that the interference with their activity may impair the fungal growth and virulence, making this approach interesting for designing antifungal drugs with a novel mechanism of action that consists in the inhibition of the CA system linked to CO2/HCO3/pH chemosensing and signaling. The inhibition of the CAs from pathogens represents an essential aspect in fighting the drug-resistance problems developed by many pathogenic microorganisms, whose growth could be impaired through the CA inhibition. We suggested that the activity of CAs is connected to the microbial survival because their activity supports many physiological microorganism functions, which require inorganic carbon [31]. In fungi, the CA inhibition affects the CO2/HCO3 balancing pathway impairing their growth as it happens in bacteria. It has also been demonstrated that carbonic anhydrase inhibitors (CAIs) could inhibit the growth of M. globosa, C. albicans, C. neoformans in vivo in conditions of limited CO2 availability (i.e., the skin surface infected by the fungus) [53]. Species of the genus Malassezia are the most abundant fungi of the skin characterized by a CO2 level very similar to ambient air. A causative link between Malassezia and disease pathogenesis remains unknown since there is a lack of information on the complex interaction of Malassezia with the immune system of the skin [54]. For this reason, an experimental model of Malassezia skin infection in mice was recently established to investigate the interaction of the fungus with the skin immune system in the context of homeostasis and disease [54].
The inhibitors towards the CAs identified in pathogens could improve the chemical arsenal used to contrast the drug-resistance phenomenon. Fortunately, many CAIs exist, as reported in the next paragraph. Therefore, these enzymes could be validated as a “pathogen protein” target, as demonstrated by the recent developments achieved in the field. Recently, FDA-approved carbonic anhydrase inhibitors, such as acetazolamide, methazolamide, and ethoxzolamide, were opportunely modified to target the bacterial CAs of the vancomycin-resistant enterococci (VRE) [55], and a similar approach may be used to develop antifungal agents. In fact, it has been reported that 3 mM ethoxyzolamide (a classical CA inhibitor) significantly reduced the growth of C. neoformans in 0.033% CO2, while, as expected, 5% CO2 reestablishes its growth [44]. These results confirm that ethoxyzolamide inhibits intracellular carbonic anhydrase activity in C. neoformans, and that the pathogen requires CA activity for growing in ambient air concentrations of CO2. Interestingly, the addition of fatty acids can restore the growth of Can2 mutants. This suggests that the lack of bicarbonate production by Can2 mutants affects fatty acid synthesis, causing a growth defect. Interestingly, C. neoformans virulence, which develops during systemic infection characterized by high CO2 concentration, is dependent on the Can2 activity, suggesting the existence of a bicarbonate-dependent signaling cascade [44]. It has been proposed that CO2 diffuses into the cell. Here, it is hydrated to bicarbonate by Can2 when present in limiting concentrations. HCO3 stimulates adenylyl cyclase activity, resulting in the activation of the cAMP-signaling pathway, which controls significant virulence determinants such as capsule biosynthesis [44].

2. Main Class of CA Inhibitors (Sulfonamides and Anions)

2.1. Substituted Benzene-Sulfonamides

The first antimicrobial drug widely used in clinical settings was Prontosil [56], a sulfanilamide prodrug, which is isosteric/isostructural with p-aminobenzoic acid (PABA), the substrate of dihydropteroate synthase (DHPS) [57,58]. After sulfanilamide, a range of analogs, the sulfa drug class, are still used as antibacterials, even if many of them show substantial drug resistance issues. Sulfa drugs are derived from sulfonamides, and the presence of primary sulfonamide moieties in sulfanilamide characterized most of the investigated CAIs until recently [32,59,60,61]. Primary sulfonamides/sulfamates/sulfamides possess the general formula R-X-SO2NH2, where R can be an aromatic, heterocyclic, aliphatic, or sugar scaffold, X = nothing, O or NH (Figure 1).
Most of the sulfonamides acting as CAIs bind Zn (II) in a tetrahedral geometry, showing an extended network of hydrogen bonds with the enzyme amino acid residues, as seen by the enzyme-inhibitor X-ray crystallographic data [32,61,62]. The aromatic/heterocyclic part of the inhibitor interacts with the hydrophilic and hydrophobic residues of the catalytic cavity [32,61]. Compounds containing -SO2NH2 group, including clinically licensed drugs, are generally considered CAIs [27,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78]. Some examples include: AAZ, MZA, EZA, and DCP, which are systemically acting antiglaucoma CAIs; DZA and BRZ are antiglaucoma agents; BZA belongs to the same pharmacological class; ZNS, SLT, and TPM are antiepileptic drugs; and SLP and IND, with COX2 selective inhibitors CLX and VLX. The diuretic hydrochlorothiazide (HCT) is also known to act as a CAI [35,79,80]. FAM is a competitive histamine H2-receptor antagonist [79], and EPA is an inhibitor of the heme-containing enzyme, indoleamine 2,3-dioxygenase-1 (IDO1), but they also act as CAIs [80] (see Figure 1). Table 1 shows selected inhibition data with some of these compounds against selected fungal CAs.

2.2. Inorganic Metal-Complexing Anions or More Complicated Species

These CA inhibitors include inorganic anions as well as several more complex species such as carboxylates, which are in fact organic anions [61,62]. Anions may bind either in the tetrahedral geometry of the metal ion or as trigonal–bipyramidal adducts [86]. Anion inhibitors show KIs in a millimolar range, diversely from the sulfonamides mentioned above, which is generally showed as KIs in the micro to nanomolar range. But their investigation as CA inhibitors offers the possibility to better understand the inhibition/catalytic mechanisms of the CAs, for improving the design of novel types of inhibitors that may have clinical applications [61,62]. A list of anions and their CA inhibitory action against selected fungal CAs is shown in Table 2.
Anions inhibit fungal CAs by coordinating to the metal ion within the enzyme active site, as exemplified in Figure 2 for Can2 complexed with acetate. As for all β-CAs, Can2 is a dimer and the active site contains amino acid residues from both monomers. The zinc ion is coordinated as shown in Figure 2, by Cys68, His124, and Cys127, whereas acetate is the fourth zin ligand, being coordinated monodentately by one of the oxygen atoms. The same type of inhibition mechanism is valid for all anions shown in Table 2, although few X-ray crystal structures of such adducts are available to date [45].

3. Other Classes of Less Investigated CAIs

3.1. Dithiocarbamates

Dithiocarbamates (DTCs) represent another class of CAIs [89,90,91,92,93,94]. They were discovered by considering the inorganic anion trithiocarbonate (TTC, CS32−) as a lead compound [95]. DTCs, as TTC, coordinate through one sulfur atom to the Zn(II) ion from the enzyme active site and also interact with the conserved Thr199 amino acid residue. Besides, DTC organic scaffolds participate in supplementary interactions with the enzyme active site [33,91,96,97]. However, there are no X-ray crystal structures of fungal CAs with DTC inhibitors reported to date.

3.2. Phenols

Other than sulfonamides and their isosteres, as well as anions and DTCs, CAs are also inhibited by phenols, which anchor to the water molecule/hydroxide ion coordinated to the metal ion [62,86,98]. Again, no structural work on phenolic fungal CAIs were reported for the moment, and these compounds are not discussed in detail here.

4. Kinetic Parameters and Inhibition Profiles of Various Fungal CAs

4.1. Saccharomyces Cerevisiae CA

Saccharomyces cerevisiae has been used as a model organism since its high degree of similarity of the biological processes with the human cells [99]. Since it can be easily manipulated, S. cerevisiae was used to develop novel antifungals, such as those altering the mitochondrial functions or inducing oxidative damage [100]. In the genome of S. cerevisiae, the gene Nce103, encoding for a β-CA, designated as scCA, has been identified. It provides the bicarbonate essential to the metabolic carboxylation reactions of the yeast metabolism [101]. scCA is an efficient catalyst for CO2 hydration to bicarbonate and protons, with kinetic parameters as follows, kcat of 9.4 × 105 s−1 and kcat/KM of 9.8 × 107 M−1s−1.

4.1.1. Sulfonamide Inhibition

scCA inhibition with sulfonamides has been investigated. The clinically used sulfonamides/sulfamates, such as acetazolamide, ethoxzolamide, methazolamide, dorzolamide, topiramate, celecoxib, and others, generally showed effective scCA inhibitory activity, with KIs in the range of 82.6–133 nM [102]. Moreover, Benzenesulfonamides substituted in 2-, 4- and 3,4-positions with amino, alkyl, halogen, and hydroxyalkyl moieties had KIs in the range of 0.976–18.45 μM [102]. Lower KIs (154—654 nM) were observed for benzenesulfonamides incorporating aminoalkyl/carboxyalkyl moieties or halogenosulfanilamides; benzene- 1,3-disulfonamides; simple heterocyclic sulfonamides and sulfanilyl-sulfonamides. KI of 15.1 nM was obtained for 4-(2-amino-pyrimidin-4-yl)-benzenesulfonamide [102].

4.1.2. Anion Inhibition

This class of inorganic metal-complexing anions can bind to the metal ion within their enzyme active site, interfering with the enzymatic catalytic process. The anion inhibitors with lower KIs (8.7–10.8 μM) were bromide, iodide, and sulfamide [102].

4.1.3. Dithiocarbamate Inhibition

DTCs are a relatively new class of CAIs. They are usually obtained by the reaction of primary or secondary amines with carbon disulfide, and most of these derivatives incorporated alkyl, mono-/bicyclic aliphatic, and heterocyclic rings but also heterocycles such as piperidine, morpholine, and piperazine. They were investigated for their inhibitory activity against scCA [103]. Some of these DTCs resulted in low nanomolar activity against the yeast enzyme (KIS = 6.4 and 259 nM) [103]. Intriguingly, several of the investigated DTCs showed excellent selectivity ratios for inhibiting scCA over the human cytosolic CA isoforms (hCA I inhibitors with KIs of 66.5–910 nM, hCA II inhibitors with KIs of 8.9–107 nM) [103].

4.1.4. Phenols as Inhibitors

A series of phenols incorporating tertiary amine and trans-pyridylethenyl-carbonyl moieties were assayed as inhibitors of scCA [104]. One of these compounds was a low nanomolar scCA inhibitor, whereas the remainder inhibited the enzyme with KIs in the range of 23.5–95.4 nM [104]. The human isoforms hCA I and hCA II were less sensitive to inhibiton by phenols, since the KIs were of 0.78–23.5 µM against hCA I and of 10.8–52.4 µM against hCA II [104].

4.2. Candida Glabrata CA

Candida glabrata is a haploid yeast belonging to the genus Candida and is considered the most common cause of candidiasis [105]. One of the significant obstacles in infections caused by C. glabrata is its innate resistance to azole antimycotic therapy, which is very effective in treating infections caused by other Candida species [105]. This pathogenic fungus encodes for a β- CA, indicated with the acronym CgNce103 or CgCA. The enzyme showed significant CO2 hydrase activity, with a kcat of 3.8 × 105 s−1 and kcat/KM of 4.8 × 107 M−1 s−1 [106].

4.2.1. Sulfonamide Inhibition

Most simple sulfonamides showed weak or moderate CgNce103 inhibitory properties. In contrast, acetazolamide and a series of 4-substituted ureido-benzene-sulfonamides, sulfamates and sulfamides effectively inhibited CgNce103 with KIs in the range of 4.1–115 nM, whereas many such compounds were ineffective, such as hCA II inhibitors. As C. glabrata offers significant resistance to many classical antifungal agents, sulfonamide inhibition of CgNce103 may allow an exciting means for limiting pathogen growth, leading to the development of antifungals with a novel mechanism of action. Besides, four generations (G0-G3) of poly(amidoamine) (PAMAM) dendrimers incorporating benzenesulfonamide moieties were investigated as inhibitors of CgNce103 [81]. The enzyme was efficiently inhibited by the four generations PAMAM–sulfonamide dendrimer with KIS = 66–509 nM [81]. CaNce103 from C. albicans and CgNce103 from C. glabrata were investigated for their inhibition with structurally novel isatin-containing sulfonamides, too [82]. The compounds show KI values in the low nanomolar range and significantly higher KI values for the human CAs. Unfortunately, no crystal structures are available for both enzymes, and homology models were constructed to rationalize their enzyme inhibition values. Using the obtained homology models, it has been seen that the backbone fold of the two enzymes showed marked differences near the active site, especially in the region where the nitro group is most likely located [82]. CaNce103 was investigated for its inhibition with A series of novel sulfamides incorporating the dopamine scaffold. CaNce103 was inhibited in the low micromolar to nanomolar range by the dopamine sulfamide analogues [107]. Finally, the CA from C. glabrata was inhibited by a series of 6-substituted benzoxaboroles in the nanomolar range, demonstrating that benzoxaborole chemotype may offer exciting development opportunities of antifungal agents [87].

4.2.2. Anion Inhibition

Investigation into its inhibition with a series of simple inorganic anions such as halogenides, pseudohalogenides, bicarbonate, carbonate, nitrate, nitrite, hydrogen sulfide, bisulfite, perchlorate, sulfate, and some isosteric species showed that CgNce103 was moderately inhibited by metal poisons, such as cyanide, azide, cyanate, thiocyanate (KIs of 0.60–1.12 mM), and was strongly inhibited by bicarbonate, nitrate, nitrite, and phenylarsonic acid (KIs of 86–98 μM) [106]. The other anions showed inhibition constants in the low millimolar range, except for bromide and iodide (KIs of 27–42 mM) [106].

4.3. Sordaria Macrospora CAs

Four different CA-genes indicated with the acronyms cas1, cas2, cas3, and cas4 are encoded in the genome of the filamentous ascomycete Sordaria macrospora, which is considered to be a model organism in biology, like S. cerevisiae [83]. The proteins encoded by these genes are indicated as CAS1, CAS2, CAS3, and CAS4 [49,84,108]. CAS1 and CAS2 are strictly related proteins that belong to the plant-like subgroup of β-CAs, CAS3 encodes a cab-type β-CA (cab is the β-CA purified from Methanobacterium thermoautotrophicum [109]), while CAS4 is an α-CA. CAS1 and CAS3 are localized in the cytoplasm, whereas the amino acid sequence of CAS2 is characterized by a signal peptide responsible for its translocation into the mitochondria, diversely from the α-CA CAS4, which was assumed to be a secreted protein [49,84,108]. It has been demonstrated that CAS1 and CAS2 are involved in the bicarbonate-dependent regulation of fruiting body development. CAS2 regulates hyphal growth and germination, too. Intriguingly, the deletion of one of the two—CAS1 or CAS2—is not lethal for the microorganism [13]. CAS1 and CAS2 showed a low activity for the CO2 hydration reaction with a kcat = 1.2 × 104 s−1 and a kcat = 1.3 × 104 s−1, respectively [49]. CAS3 had a higher activity level, showing an order of magnitude higher than catalytic activity (kcat = 7.9 × 105 s−1) for the CAS1 and CAS2. CAS4 has not yet been investigated up to now. Interestingly, CAS3 activity resulted in the same order as those calculated for the β-CAs from Cryptococcus neoformans, Candida albicans, and Saccharomyces cerevisiae [110].

4.3.1. Sulfonamide Inhibition

A panel of aromatic compounds, including heterocyclic, aliphatic sulfonamides, and one sulfamate, were tested to determine their inhibition constants for the related enzymes, CAS1, CAS2, and CAS3 [111,112]. CAS1 was efficiently inhibited by simple aromatic sulfonamides, such as tosylamide, 3-fluoro-/chloro-sulfanilamide, which has inhibition constants in the range of 43.2–79.6 nM. Heterocyclic derivatives such as acetazolamide, methazolamide, topiramate, ethoxzolamide, dorzolamide, and brinzolamide showed medium potency inhibitory action with inhibition constants of 360–445 nM [112]. CAS2 was, on the other hand, less sensitive to inhibition with sulfonamides. However, some effective CAS2 inhibitors comprised 5-amino-1,3,4-thiadiazole-2-sulfonamide, which is in fact the deacetylated precursor of the classical sulfonamide acetazolamide. 4-Hydroxymethyl-benzenesulfonamide was also an effective inhibitor. These two compounds showed inhibition constants of 48.1–92.5nM against CAS2 [112]. Acetazolamide, dorzolamide, ethoxzolamide, topiramate, sulpiride, indisulam, celecoxib, and sulthiame were medium potency CAS2 inhibitors (KIs of 143–857 nM) [112]. The most effective CAS3 inhibitors were benzolamide, brinzolamide, dichlorophenamide, methazolamide, acetazolamide, ethoxzolamide, sulfanilamide, methanilamide, and benzene-1,3-disulfonamide, with KIs in the range of 54–95 nM [111]. CAS3 generally showed a higher affinity for sulfonamide inhibitors compared to CAS1 and CAS2 [111]. Other sulfonamides showed affinities in the high micromolar range or were ineffective as CAS1/2/3 inhibitors [111,112]. Intriguing, small changes in the inhibitor structure led to important differences in the three enzymes’ activity, demonstrating that it is possible to tune the enzyme inhibition, altering the inhibitor scaffold of the inhibitors [49,111,112]. This makes possible the synthesis of selective inhibitors, which may interfere with the activity of the fungal CAs, leaving unaltered the activity of the human α-CAs.

4.3.2. Anion Inhibition

Up to now, only CAS3 has been investigated for its inhibition profiles with this class of inorganic metal-complexing anions, which can bind to the metal ion within their enzyme active site, interfering with the enzymatic catalytic process. The most effective CAS3 anions/small molecule inhibitors were diethyl-dithiocarbamate, sulfamide, sulfamate, phenyl boronic, and phenyl arsonic acids, with KIs in the range of 89–97 μM [110]. Anions such as iodide, the pseudohalides, bicarbonate, carbonate, nitrate, nitrite, hydrogensulfide, stannate, selenate, tellurate, tetraborate, perrhenate, perruthenate, selenocyanide, and trithiocarbonate resulted in low millimolar CAS3 inhibitors [110]. CAS3 was not inhibited by halides, sulfate, hydrogensulfite, peroxydisulfate, diphosphate, divanadate, perchlorate, tetrafluoroborate, fluorosulfonate, and iminodisulfonate [110].

4.4. Malassezia Globosa and M. Restricta

Malassezia is a fungi genus, including seven species: M. furfur, M. pachydermatis, and M. sympodialis, M. globosa, M. obtusa, M. restricta, and M. slooffiae.
M. globosa have been identified as members of the human cutaneous flora, coexisting with the skin’s microbial flora [113]. Malassezia fungi represent the etiologic agents of specific skin diseases, such as pityriasis versicolor, seborrheic dermatitis scalp, and dandruff [113]. The last condition is a frequent skin disorder restricted to the scalp. It is caused by the oleic acid of the scalp sebum, produced through t hydrolysis of triglycerides, which are accomplished by the lipases produced mainly by M. globosa [114]. Other than M. globose, another fungus, M. restricta, is also involved in starting the disequilibrium between the commensals Cutibacterium acnes (formerly named Propionibacterium acnes) and Staphylococcus sp., both of which contribute to dandruff and seborrheic dermatitis symptoms [85,115,116]. Most strategies used to treat the dandruff are based on impairing the growth of the fungi mentioned above, using active ingredients in anti-dandruff shampoos, such as the ketoconazole B, an azole antifungal agent interfering with the biosynthesis of fungal sterols, or the pyridinethione A that interferes with the synthesis of ergosterol, a key component of fungal cell walls [117,118]. However, the efficacy of these compounds in preventing/treating dandruff is not very high. Thus, it is necessary to target new molecules of the Malassezia metabolism, such as the carbonic anhydrases, recently studied as protein-fungal targets by our groups.

4.4.1. Malassezia globosa CA

The genome of the fungal parasite Malassezia globosa contains a single gene encoding a CA (acronym MgCA) belonging to the β-class. The enzyme showed an appreciable CO2 hydrase activity, with a kcat of 9.2 × 105 s−1 and kcat/KM of 8.3 × 107 M−1 s−1 [119].

Sulfonamide Inhibition

Many primary sulfonamides showed KIs in the nanomolar range of 63–174 nM. The clinically used drugs belonging to the series AZZ-HTC, such as MZA, EZA, BRZ, CLX, and SAC, acted as mild inhibitors of MgCA (KIs = 31.5–79 μM) [120]. Interesting to note, in general, MgCA potent inhibitors resulted in mild inhibitors of the β-CA from M. restricta and vice-versa, highlighting how different the inhibition pattern of the two homologous fungal enzymes was [120]. Again, the two fungal enzymes had an inhibition profile highly distinct from those of the two human isoenzymes [120].

Anion Inhibition

Anions, such as halides, pseudohalides, nitrite/nitrate, sulfite/sulfate and anions isoelectronic with them, but also complex anions incorporating heavy metals, as well as the simple small molecules known to have an affinity for Zn (II) in the CAs, such as sulfamide, sulfamic acid, phenylboronic and phenylarsonic acid, bicarbonate and diethyldithiocarbamate, have been known for their interaction with MgCA. The best MgCA inhibitors were sulfamide, sulfamate, phenylboronic acid, phenylarsonic acid, bicarbonate, and diethyldithiocarbamate, with KIs ranging between 83 and 94 µM [119]. Interestingly, bicarbonate is also a substrate/reaction product of the CAs, and this behavior is entirely unexpected, especially considering that carbonate did not show inhibitory properties [119]. This inhibitory behavior of the enzyme, which is very difficult to explain, could be physiologically crucial.

Dithiocarbamate Inhibition

A series of DTCs, incorporating various scaffolds, among which are those of N,N-dimethylaminoethylenediamine, the amino alcohols with 3–5 carbon atoms in their molecule, 3-amino-quinuclidine, piperidine, morpholine and piperazine derivatives, as well as phenethylamine and its 4-sulfamoylated derivative, were investigated for the inhibition of MgCA [121]. Most of the DTCs were shown to be effective, although not low, nanomolar potencies against MgCA, but several of them showed a with KIs ranging between 383 and 6235 nM, resulting in very effective inhibitors of MgCA when compared with the standard sulfonamide drug acetazolamide (KI of 74 µM) [121]. As a result, DTCs could be critical molecules in searching for more potent and efficient fungal CAIs.

Monothiocarbamate Inhibition

Monothiocarbamates (MTCs) were used to inhibit MgCA. They incorporate different scaffolds, among which were aliphatic amine with 1–4 carbons atom in their molecule, morpholine, piperazine, as well as phenethylamine and benzylamine derivatives. The considered MTCs showed KIs spanning between 1.85 and 18.9 µM, producing results better than those observed for the clinically used sulfonamide drug acetazolamide (KI of 74 µM) [88]. The docking studies applied to the homology model of MgCA have highlighted the main differences in the binding mode of MTCs and DTCs within the fungal CA active site [88].

Phenols as Inhibitors

A panel of 22 phenols was investigated as inhibitors of MgCA. All tested phenols possessed a better efficacy in inhibiting MgCA than the clinically used sulfonamide acetazolamide, with KIs in the range of 2.5 and 65.0 µM [122]. A homology-built model of MgCA revealed a vast network of hydrogen bonds and hydrophobic interactions between the phenol and active site residues. The OH moiety of the inhibitor was anchored to the zinc-coordinated water, making hydrogen bonds with Ser48 and Asp49 in the catalytic pocket [122]. MgCA was also investigated for its inhibition profile with various natural phenols, such as xanthomicrol and rosmarinic acid, which inhibited MgCA with a KIS = 0.6 and 2.2 µM, respectively [123].

Phosphonamidates

A phosphorus-based zinc-binding motif, such as phosphonamidate, inhibited the MgCA with KIs in the range 28–256 µM (Can2, CA from C. neoformans, with KIs = 50–3488 µM and CgNce, CA from Candida glabrata, with KIs = 23–656 µM) [124]. Interestingly, the human isoforms (CAs I and II) are inhibited in a high micromolar range (32.8–961.2 mM), suggesting that this group of inhibitors could be considered attractive leads for developing new anti-infective agents.

4.4.2. Malassezia restricta CA

The genome of the opportunistic pathogen Malassezia restricta encodes for a β-CA (acronym MreCA), which had a high catalytic activity for the hydration of CO2 into bicarbonate and protons, with the following kinetic parameters: kcat = 1.06 × 106 s−1 and kcat/KM = 1.07 × 108 M−1s−1 [125].

Sulfonamide Inhibition

The investigation of the sulfonamide inhibition profile of MreCA provides evidence that the MreCA sulfonamide inhibitors, such as dorzolamide, brinzolamide, indisulam, valdecoxib, sulthiam, and acetazolamide, inhibited the enzyme with a KI < 1.0 μM [120]. These inhibitors resulted in the best for MreCA. Noticeably, the MreCA sulfonamide inhibition profile was very different from those of the homologous enzyme MgCA and the human isoenzymes, hCA I and hCA II [120]. The crystallographic structures of MreCA and MgCA not being available, it has been speculated that the fungal and the human CAs (hCA I and hCA II) have a similar catalytic site, but unusual architectural features, which may be responsible for the differences in KIs obtained for the two fungal and human enzymes.

Anion Inhibition

The investigation of the classical anions with their inhibition for MreCA showed that the most effective were diethyldithiocarbamate, sulfamide, phenyl arsenic acid, stannate, tellurate, tetraborate, selenocyanate, trithiocarbonate, and bicarbonate [126]. Moreover, in this case, MreCA and MgCA resulted in different KIs for the inhibitors used [126]. It is possible that the differences in the catalytic site could be the cause of their peculiar inhibitory behavior. All this justifies why the anion inhibition profile studies are crucial for the comprehension of the CA inhibition/catalytic mechanisms, allowing the design of novel types of inhibitors, which may have clinical applications for the management of dandruff and seborrheic dermatitis.

5. Conclusions

Drug-resistant microorganisms are an unavoidable and everyday phenomenon that requires the search for new anti-infectives with a novel mechanism of action. CAs have only recently begun to be studied in depth in microorganisms, since it has been shown that CAs are essential for the life cycle of many pathogens, and their inhibition may lead to their growth deficiency. Many opportunistic or pathogenic fungi, such as C. albicans, C. glabrata, C. neoformans, M. globosa, M. restricta, S. cereviasiae, and many others, encode for β-CAs, which, with their activity, are involved in the production of bicarbonate, an adenyl cyclase inducer. The CA/adenyl cyclase system constitutes the mechanism of CO2-sensing which can be deregulated by inhibiting one of these enzymes. Interesting, some fungi also present α-CAs, which are less investigated concerning the β-class since the α-CAs are rarer to find. The current review reports that sulfonamides and their bioisosteres can inhibit in vitro the β- and α-CAs from the fungi aforementioned above. Moreover, it has also been demonstrated that these compounds could inhibit the growth of M. globosa, C. albicans, and C. neoformans in vivo in conditions of limited CO2 availability (i.e., the skin surface infected by the fungus) [53]. This evidence corroborates how the fungal CAs could be considered druggable targets, mainly because there are no β-class enzymes in humans, and thus, the side effects due to inhibition of the host enzymes are not significant. In conclusion, the tendency to selectively inhibit CAs from pathogens such as fungi might constitute an alternative approach for designing anti-infectives with novel mechanisms of action. This is a rather new research field, which requires many other X-ray crystal structures of CAs from pathogens (as very few of them are presently available) together with massive drug design campaigns for finding selective inhibitors of the pathogenic versus the host enzymes.

Author Contributions

Conceptualization, C.C. and C.T.S.; writing—original draft preparation, C.C.; writing—review and editing, C.C. and C.T.S. Both authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Italian Ministry of University and Research, project FISR2019_04819 BacCAD (to C.T.S. and C.C.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to Valentina Brasiello and Giovanni Del Monaco for technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Canadell, J.G.; Le Quere, C.; Raupach, M.R.; Field, C.B.; Buitenhuis, E.T.; Ciais, P.; Conway, T.J.; Gillett, N.P.; Houghton, R.A.; Marland, G. Contributions to accelerating atmospheric CO2 growth from economic activity, carbon intensity, and efficiency of natural sinks. Proc. Natl. Acad. Sci. USA 2007, 104, 18866–18870. [Google Scholar] [CrossRef] [Green Version]
  2. Cramer, W.; Bondeau, A.; Schaphoff, S.; Lucht, W.; Smith, B.; Sitch, S. Tropical forests and the global carbon cycle: Impacts of atmospheric carbon dioxide, climate change and rate of deforestation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2004, 359, 331–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Cheng, L.; Booker, F.L.; Tu, C.; Burkey, K.O.; Zhou, L.; Shew, H.D.; Rufty, T.W.; Hu, S. Arbuscular mycorrhizal fungi increase organic carbon decomposition under elevated CO2. Science 2012, 337, 1084–1087. [Google Scholar] [CrossRef] [PubMed]
  4. Mitchell, A.P. Fungal CO2 sensing: A breath of fresh air. Curr. Biol. 2005, 15, R934–R936. [Google Scholar] [CrossRef] [Green Version]
  5. Siscar-Lewin, S.; Hube, B.; Brunke, S. Antivirulence and avirulence genes in human pathogenic fungi. Virulence 2019, 10, 935–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gonzalez-Lara, M.F.; Ostrosky-Zeichner, L. Invasive Candidiasis. Semin. Respir. Crit. Care Med. 2020, 41, 3–12. [Google Scholar] [CrossRef] [PubMed]
  7. Ledoux, M.P.; Guffroy, B.; Nivoix, Y.; Simand, C.; Herbrecht, R. Invasive Pulmonary Aspergillosis. Semin. Respir. Crit. Care Med. 2020, 41, 80–98. [Google Scholar] [CrossRef] [PubMed]
  8. Reid, G.; Lynch, J.P., 3rd; Fishbein, M.C.; Clark, N.M. Mucormycosis. Semin. Respir. Crit. Care Med. 2020, 41, 99–114. [Google Scholar] [CrossRef]
  9. Zavala, S.; Baddley, J.W. Cryptococcosis. Semin. Respir. Crit. Care Med. 2020, 41, 69–79. [Google Scholar] [CrossRef]
  10. Ohkuni, K.; Hayashi, M.; Yamashita, I. Bicarbonate-mediated social communication stimulates meiosis and sporulation of Saccharomyces cerevisiae. Yeast 1998, 14, 623–631. [Google Scholar] [CrossRef]
  11. Uno, I.; Matsumoto, K.; Hirata, A.; Ishikawa, T. Outer plaque assembly and spore encapsulation are defective during sporulation of adenylate cyclase-deficient mutants of Saccharomyces cerevisiae. J. Cell Biol. 1985, 100, 1854–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chang, J.C.; Oude-Elferink, R.P. Role of the bicarbonate-responsive soluble adenylyl cyclase in pH sensing and metabolic regulation. Front. Physiol. 2014, 5, 42. [Google Scholar] [CrossRef] [Green Version]
  13. Klengel, T.; Liang, W.J.; Chaloupka, J.; Ruoff, C.; Schroppel, K.; Naglik, J.R.; Eckert, S.E.; Mogensen, E.G.; Haynes, K.; Tuite, M.F.; et al. Fungal adenylyl cyclase integrates CO2 sensing with cAMP signaling and virulence. Curr. Biol. 2005, 15, 2021–2026. [Google Scholar] [CrossRef] [Green Version]
  14. Chadwick, B.J.; Lin, X. On the History and Applications of Congenic Strains in Cryptococcus Research. Pathogens 2020, 9, 750. [Google Scholar] [CrossRef] [PubMed]
  15. Alspaugh, J.A.; Perfect, J.R.; Heitman, J. Cryptococcus neoformans mating and virulence are regulated by the G-protein alpha subunit GPA1 and cAMP. Genes Dev. 1997, 11, 3206–3217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chen, Y.; Cann, M.J.; Litvin, T.N.; Iourgenko, V.; Sinclair, M.L.; Levin, L.R.; Buck, J. Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor. Science 2000, 289, 625–628. [Google Scholar] [CrossRef] [Green Version]
  17. Granger, D.L.; Perfect, J.R.; Durack, D.T. Virulence of Cryptococcus neoformans. Regulation of capsule synthesis by carbon dioxide. J. Clin. Investig. 1985, 76, 508–516. [Google Scholar] [CrossRef] [Green Version]
  18. Buck, J.; Levin, L.R. Physiological sensing of carbon dioxide/bicarbonate/pH via cyclic nucleotide signaling. Sensors 2011, 11, 2112–2128. [Google Scholar] [CrossRef] [Green Version]
  19. D’Souza, C.A.; Heitman, J. Conserved cAMP signaling cascades regulate fungal development and virulence. FEMS Microbiol. Rev. 2001, 25, 349–364. [Google Scholar] [CrossRef]
  20. Bahn, Y.S.; Cox, G.M.; Perfect, J.R.; Heitman, J. Carbonic anhydrase and CO2 sensing during Cryptococcus neoformans growth, differentiation, and virulence. Curr. Biol. 2005, 15, 2013–2020. [Google Scholar] [CrossRef] [Green Version]
  21. Staniszewska, M. Virulence Factors in Candida species. Curr. Protein Pept. Sci. 2020, 21, 313–323. [Google Scholar] [CrossRef]
  22. Rutherford, J.C.; Bahn, Y.S.; van den Berg, B.; Heitman, J.; Xue, C. Nutrient and Stress Sensing in Pathogenic Yeasts. Front. Microbiol. 2019, 10, 442. [Google Scholar] [CrossRef] [PubMed]
  23. Kohler, J.R.; Hube, B.; Puccia, R.; Casadevall, A.; Perfect, J.R. Fungi that Infect Humans. Microbiol. Spectr. 2017, 5, 811–843. [Google Scholar]
  24. Guzel-Akdemir, O.; Carradori, S.; Grande, R.; Demir-Yazici, K.; Angeli, A.; Supuran, C.T.; Akdemir, A. Development of Thiazolidinones as Fungal Carbonic Anhydrase Inhibitors. Int. J. Mol. Sci. 2020, 21, 2960. [Google Scholar] [CrossRef] [Green Version]
  25. Annunziato, G.; Angeli, A.; D’Alba, F.; Bruno, A.; Pieroni, M.; Vullo, D.; De Luca, V.; Capasso, C.; Supuran, C.T.; Costantino, G. Discovery of New Potential Anti-Infective Compounds Based on Carbonic Anhydrase Inhibitors by Rational Target-Focused Repurposing Approaches. ChemMedChem 2016, 11, 1904–1914. [Google Scholar] [CrossRef] [PubMed]
  26. Ozensoy Guler, O.; Capasso, C.; Supuran, C.T. A magnificent enzyme superfamily: Carbonic anhydrases, their purification and characterization. J. Enzym. Inhib. Med. Chem. 2016, 31, 689–694. [Google Scholar] [CrossRef] [PubMed]
  27. Del Prete, S.; Vullo, D.; De Luca, V.; Carginale, V.; Ferraroni, M.; Osman, S.M.; AlOthman, Z.; Supuran, C.T.; Capasso, C. Sulfonamide inhibition studies of the beta-carbonic anhydrase from the pathogenic bacterium Vibrio cholerae. Bioorg. Med. Chem. 2016, 24, 1115–1120. [Google Scholar] [CrossRef]
  28. Del Prete, S.; De Luca, V.; De Simone, G.; Supuran, C.T.; Capasso, C. Cloning, expression and purification of the complete domain of the eta-carbonic anhydrase from Plasmodium falciparum. J. Enzym. Inhib. Med. Chem. 2016, 31 (Suppl. S4), 1–6. [Google Scholar] [CrossRef] [Green Version]
  29. Capasso, C.; Supuran, C.T. An Overview of the Carbonic Anhydrases from Two Pathogens of the Oral Cavity: Streptococcus mutans and Porphyromonas gingivalis. Curr. Top. Med. Chem. 2016, 16, 2359–2368. [Google Scholar] [CrossRef]
  30. Capasso, C.; Supuran, C.T. An overview of the alpha-, beta- and gamma-carbonic anhydrases from Bacteria: Can bacterial carbonic anhydrases shed new light on evolution of bacteria? J. Enzym. Inhib. Med. Chem. 2015, 30, 325–332. [Google Scholar] [CrossRef] [Green Version]
  31. Supuran, C.T.; Capasso, C. An Overview of the Bacterial Carbonic Anhydrases. Metabolites 2017, 7, 56. [Google Scholar] [CrossRef] [Green Version]
  32. Supuran, C.T. Structure and function of carbonic anhydrases. Biochem. J. 2016, 473, 2023–2032. [Google Scholar] [CrossRef] [PubMed]
  33. McKenna, R.; Supuran, C.T. Carbonic anhydrase inhibitors drug design. Subcell Biochem. 2014, 75, 291–323. [Google Scholar] [PubMed]
  34. Neri, D.; Supuran, C.T. Interfering with pH regulation in tumours as a therapeutic strategy. Nat. Rev. Drug Discov. 2011, 10, 767–777. [Google Scholar] [CrossRef] [Green Version]
  35. Supuran, C.T. Carbonic anhydrases: Novel therapeutic applications for inhibitors and activators. Nat. Rev. Drug. Discov. 2008, 7, 168–181. [Google Scholar] [CrossRef] [PubMed]
  36. Supuran, C.T. Carbonic anhydrases—An overview. Curr. Pharm. Des. 2008, 14, 603–614. [Google Scholar] [CrossRef] [PubMed]
  37. Supuran, C.T.; Capasso, C. Biomedical applications of prokaryotic carbonic anhydrases. Expert Opin. Ther. Pat. 2018, 28, 745–754. [Google Scholar] [CrossRef]
  38. Supuran, C.T.; Capasso, C. Carbonic Anhydrase from Porphyromonas Gingivalis as a Drug Target. Pathogens 2017, 6, 30. [Google Scholar] [CrossRef] [Green Version]
  39. Capasso, C.; Supuran, C.T. An Overview of the Selectivity and Efficiency of the Bacterial Carbonic Anhydrase Inhibitors. Curr. Med. Chem. 2015, 22, 2130–2139. [Google Scholar] [CrossRef]
  40. Rogato, A.; Del Prete, S.; Nocentini, A.; Carginale, V.; Supuran, C.T.; Capasso, C. Phaeodactylum tricornutum as a model organism for testing the membrane penetrability of sulphonamide carbonic anhydrase inhibitors. J. Enzym. Inhib. Med. Chem. 2019, 34, 510–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Angeli, A.; Pinteala, M.; Maier, S.S.; Del Prete, S.; Capasso, C.; Simionescu, B.C.; Supuran, C.T. Inhibition of alpha-, beta-, gamma-, delta-, zeta- and eta-class carbonic anhydrases from bacteria, fungi, algae, diatoms and protozoans with famotidine. J Enzym. Inhib. Med. Chem. 2019, 34, 644–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Berrino, E.; Bozdag, M.; Del Prete, S.; Alasmary, F.A.S.; Alqahtani, L.S.; AlOthman, Z.; Capasso, C.; Supuran, C.T. Inhibition of alpha-, beta-, gamma-, and delta-carbonic anhydrases from bacteria and diatoms with N’-aryl-N-hydroxy-ureas. J. Enzym. Inhib. Med. Chem. 2018, 33, 1194–1198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Angeli, A.; Del Prete, S.; Alasmary, F.A.S.; Alqahtani, L.S.; AlOthman, Z.; Donald, W.A.; Capasso, C.; Supuran, C.T. The first activation studies of the eta-carbonic anhydrase from the malaria parasite Plasmodium falciparum with amines and amino acids. Bioorg. Chem. 2018, 80, 94–98. [Google Scholar] [CrossRef] [PubMed]
  44. Mogensen, E.G.; Janbon, G.; Chaloupka, J.; Steegborn, C.; Fu, M.S.; Moyrand, F.; Klengel, T.; Pearson, D.S.; Geeves, M.A.; Buck, J.; et al. Cryptococcus neoformans senses CO2 through the carbonic anhydrase Can2 and the adenylyl cyclase Cac1. Eukaryot Cell 2006, 5, 103–111. [Google Scholar] [CrossRef] [Green Version]
  45. Schlicker, C.; Hall, R.A.; Vullo, D.; Middelhaufe, S.; Gertz, M.; Supuran, C.T.; Muhlschlegel, F.A.; Steegborn, C. Structure and inhibition of the CO2-sensing carbonic anhydrase Can2 from the pathogenic fungus Cryptococcus neoformans. J. Mol. Biol. 2009, 385, 1207–1220. [Google Scholar] [CrossRef]
  46. Isik, S.; Kockar, F.; Aydin, M.; Arslan, O.; Guler, O.O.; Innocenti, A.; Scozzafava, A.; Supuran, C.T. Carbonic anhydrase inhibitors: Inhibition of the beta-class enzyme from the yeast Saccharomyces cerevisiae with sulfonamides and sulfamates. Bioorg. Med. Chem. 2009, 17, 1158–1163. [Google Scholar] [CrossRef]
  47. Isik, S.; Kockar, F.; Arslan, O.; Guler, O.O.; Innocenti, A.; Supuran, C.T. Carbonic anhydrase inhibitors. Inhibition of the beta-class enzyme from the yeast Saccharomyces cerevisiae with anions. Bioorg. Med. Chem. Lett. 2008, 18, 6327–6331. [Google Scholar] [CrossRef]
  48. Cuesta-Seijo, J.A.; Borchert, M.S.; Navarro-Poulsen, J.C.; Schnorr, K.M.; Mortensen, S.B.; Lo Leggio, L. Structure of a dimeric fungal alpha-type carbonic anhydrase. FEBS Lett. 2011, 585, 1042–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Lehneck, R.; Neumann, P.; Vullo, D.; Elleuche, S.; Supuran, C.T.; Ficner, R.; Poggeler, S. Crystal structures of two tetrameric beta-carbonic anhydrases from the filamentous ascomycete Sordaria macrospora. FEBS J. 2014, 281, 1759–1772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Kim, S.; Yeon, J.; Sung, J.; Kim, N.J.; Hong, S.; Jin, M.S. Structural insights into novel mechanisms of inhibition of the major beta-carbonic anhydrase CafB from the pathogenic fungus Aspergillus fumigatus. J. Struct. Biol. 2021, 213, 107700. [Google Scholar] [CrossRef]
  51. Kim, S.; Yeon, J.; Sung, J.; Jin, M.S. Crystal Structure of beta-Carbonic Anhydrase CafA from the Fungal Pathogen Aspergillus fumigatus. Mol. Cells 2020, 43, 831–840. [Google Scholar] [PubMed]
  52. Covarrubias, A.S.; Bergfors, T.; Jones, T.A.; Hogbom, M. Structural mechanics of the pH-dependent activity of beta-carbonic anhydrase from Mycobacterium tuberculosis. J. Biol. Chem. 2006, 281, 4993–4999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hewitson, K.S.; Vullo, D.; Scozzafava, A.; Mastrolorenzo, A.; Supuran, C.T. Molecular cloning, characterization, and inhibition studies of a beta-carbonic anhydrase from Malassezia globosa, a potential antidandruff target. J. Med. Chem. 2012, 55, 3513–3520. [Google Scholar] [CrossRef] [PubMed]
  54. Sparber, F.; LeibundGut-Landmann, S. Infecting Mice with Malassezia spp. to Study the Fungus-Host Interaction. J. Vis. Exp. 2019, 153, e60175. [Google Scholar] [CrossRef]
  55. Kaur, J.; Cao, X.; Abutaleb, N.S.; Elkashif, A.; Graboski, A.L.; Krabill, A.D.; AbdelKhalek, A.H.; An, W.; Bhardwaj, A.; Seleem, M.N.; et al. Optimization of Acetazolamide-Based Scaffold as Potent Inhibitors of Vancomycin-Resistant Enterococcus. J. Med. Chem. 2020, 63, 9540–9562. [Google Scholar] [CrossRef] [PubMed]
  56. Otten, H. Domagk and the development of the sulphonamides. J. Antimicrob. Chem. 1986, 17, 689–696. [Google Scholar] [CrossRef]
  57. Capasso, C.; Supuran, C.T. Sulfa and trimethoprim-like drugs—Antimetabolites acting as carbonic anhydrase, dihydropteroate synthase and dihydrofolate reductase inhibitors. J. Enzym. Inhib. Med. Chem. 2014, 29, 379–387. [Google Scholar] [CrossRef]
  58. Achari, A.; Somers, D.O.; Champness, J.N.; Bryant, P.K.; Rosemond, J.; Stammers, D.K. Crystal structure of the anti-bacterial sulfonamide drug target dihydropteroate synthase. Nat. Struct. Biol. 1997, 4, 490–497. [Google Scholar] [CrossRef]
  59. Supuran, C.T. Carbonic anhydrase inhibition and the management of neuropathic pain. Expert Rev. Neurother. 2016, 16, 961–968. [Google Scholar] [CrossRef]
  60. Supuran, C.T. Drug interaction considerations in the therapeutic use of carbonic anhydrase inhibitors. Expert Opin. Drug Metab. Toxicol. 2016, 12, 423–431. [Google Scholar] [CrossRef] [PubMed]
  61. Supuran, C.T. Advances in structure-based drug discovery of carbonic anhydrase inhibitors. Expert Opin. Drug Discov. 2017, 12, 61–88. [Google Scholar] [CrossRef]
  62. Supuran, C.T. How many carbonic anhydrase inhibition mechanisms exist? J. Enzym. Inhib. Med. Chem. 2016, 31, 345–360. [Google Scholar] [CrossRef] [PubMed]
  63. Vullo, D.; Del Prete, S.; Fisher, G.M.; Andrews, K.T.; Poulsen, S.A.; Capasso, C.; Supuran, C.T. Sulfonamide inhibition studies of the eta-class carbonic anhydrase from the malaria pathogen Plasmodium falciparum. Bioorg. Med. Chem. 2015, 23, 526–531. [Google Scholar] [CrossRef] [PubMed]
  64. Vullo, D.; De Luca, V.; Del Prete, S.; Carginale, V.; Scozzafava, A.; Capasso, C.; Supuran, C.T. Sulfonamide inhibition studies of the gamma-carbonic anhydrase from the Antarctic bacterium Pseudoalteromonas haloplanktis. Bioorg. Med. Chem. Lett. 2015, 25, 3550–3555. [Google Scholar] [CrossRef] [Green Version]
  65. Vullo, D.; De Luca, V.; Del Prete, S.; Carginale, V.; Scozzafava, A.; Capasso, C.; Supuran, C.T. Sulfonamide inhibition studies of the gamma-carbonic anhydrase from the Antarctic cyanobacterium Nostoc commune. Bioorg. Med. Chem. 2015, 23, 1728–1734. [Google Scholar] [CrossRef] [PubMed]
  66. Dedeoglu, N.; DeLuca, V.; Isik, S.; Yildirim, H.; Kockar, F.; Capasso, C.; Supuran, C.T. Sulfonamide inhibition study of the beta-class carbonic anhydrase from the caries producing pathogen Streptococcus mutans. Bioorg. Med. Chem. Lett. 2015, 25, 2291–2297. [Google Scholar] [CrossRef] [PubMed]
  67. Alafeefy, A.M.; Ceruso, M.; Al-Tamimi, A.M.; Del Prete, S.; Supuran, C.T.; Capasso, C. Inhibition studies of quinazoline-sulfonamide derivatives against the gamma-CA (PgiCA) from the pathogenic bacterium, Porphyromonas gingivalis. J. Enzym. Inhib. Med. Chem. 2015, 30, 592–596. [Google Scholar] [CrossRef]
  68. Alafeefy, A.M.; Abdel-Aziz, H.A.; Vullo, D.; Al-Tamimi, A.M.; Awaad, A.S.; Mohamed, M.A.; Capasso, C.; Supuran, C.T. Inhibition of human carbonic anhydrase isozymes I, II, IX and XII with a new series of sulfonamides incorporating aroylhydrazone-, [1,2,4]triazolo [3,4-b][1,3,4]thiadiazinyl- or 2-(cyanophenylmethylene)-1,3,4-thiadiazol-3(2H)-yl moieties. J. Enzym. Inhib. Med. Chem. 2015, 30, 52–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Diaz, J.R.; Fernandez Baldo, M.; Echeverria, G.; Baldoni, H.; Vullo, D.; Soria, D.B.; Supuran, C.T.; Cami, G.E. A substituted sulfonamide and its Co (II), Cu (II), and Zn (II) complexes as potential antifungal agents. J. Enzym. Inhib. Med. Chem. 2016, 31 (Suppl. S2), 51–62. [Google Scholar] [CrossRef] [Green Version]
  70. Del Prete, S.; Vullo, D.; De Luca, V.; Carginale, V.; Osman, S.M.; AlOthman, Z.; Supuran, C.T.; Capasso, C. Comparison of the sulfonamide inhibition profiles of the alpha-, beta- and gamma-carbonic anhydrases from the pathogenic bacterium Vibrio cholerae. Bioorg. Med. Chem. Lett. 2016, 26, 1941–1946. [Google Scholar] [CrossRef] [PubMed]
  71. Del Prete, S.; Vullo, D.; De Luca, V.; Carginale, V.; Osman, S.M.; AlOthman, Z.; Supuran, C.T.; Capasso, C. Cloning, expression, purification and sulfonamide inhibition profile of the complete domain of the eta-carbonic anhydrase from Plasmodium falciparum. Bioorg. Med. Chem. Lett. 2016, 26, 4184–4190. [Google Scholar] [CrossRef]
  72. Abdel Gawad, N.M.; Amin, N.H.; Elsaadi, M.T.; Mohamed, F.M.; Angeli, A.; De Luca, V.; Capasso, C.; Supuran, C.T. Synthesis of 4-(thiazol-2-ylamino)-benzenesulfonamides with carbonic anhydrase I, II and IX inhibitory activity and cytotoxic effects against breast cancer cell lines. Bioorg. Med. Chem. 2016, 24, 3043–3051. [Google Scholar] [CrossRef] [PubMed]
  73. Supuran, C.T. Legionella pneumophila Carbonic Anhydrases: Underexplored Antibacterial Drug Targets. Pathogens 2016, 5, 44. [Google Scholar] [CrossRef] [Green Version]
  74. Nishimori, I.; Vullo, D.; Minakuchi, T.; Scozzafava, A.; Capasso, C.; Supuran, C.T. Sulfonamide inhibition studies of two beta-carbonic anhydrases from the bacterial pathogen Legionella pneumophila. Bioorg. Med. Chem. 2014, 22, 2939–2946. [Google Scholar] [CrossRef]
  75. Vullo, D.; Sai Kumar, R.S.; Scozzafava, A.; Capasso, C.; Ferry, J.G.; Supuran, C.T. Anion inhibition studies of a beta-carbonic anhydrase from Clostridium perfringens. Bioorg. Med. Chem. Lett. 2013, 23, 6706–6710. [Google Scholar] [CrossRef]
  76. Nishimori, I.; Minakuchi, T.; Maresca, A.; Carta, F.; Scozzafava, A.; Supuran, C.T. The beta-carbonic anhydrases from Mycobacterium tuberculosis as drug targets. Curr. Pharm. Des. 2010, 16, 3300–3309. [Google Scholar] [CrossRef]
  77. Carta, F.; Maresca, A.; Covarrubias, A.S.; Mowbray, S.L.; Jones, T.A.; Supuran, C.T. Carbonic anhydrase inhibitors. Characterization and inhibition studies of the most active beta-carbonic anhydrase from Mycobacterium tuberculosis, Rv3588c. Bioorg. Med. Chem. Lett. 2009, 19, 6649–6654. [Google Scholar] [CrossRef]
  78. Supuran, C.T. Special Issue: Sulfonamides. Molecules 2017, 22, 1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Nguyen, K.; Ahlawat, R. Famotidine. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  80. Komiya, T.; Huang, C.H. Updates in the Clinical Development of Epacadostat and Other Indoleamine 2,3-Dioxygenase 1 Inhibitors (IDO1) for Human Cancers. Front. Oncol. 2018, 8, 423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Carta, F.; Osman, S.M.; Vullo, D.; AlOthman, Z.; Del Prete, S.; Capasso, C.; Supuran, C.T. Poly(amidoamine) dendrimers show carbonic anhydrase inhibitory activity against alpha-, beta-, gamma- and eta-class enzymes. Bioorg. Med. Chem. 2015, 23, 6794–6798. [Google Scholar] [CrossRef] [PubMed]
  82. Akdemir, A.; Guzel-Akdemir, O.; Karali, N.; Supuran, C.T. Isatin analogs as novel inhibitors of Candida spp. beta-carbonic anhydrase enzymes. Bioorg. Med. Chem. 2016, 24, 1648–1652. [Google Scholar] [CrossRef] [PubMed]
  83. Teichert, I.; Poggeler, S.; Nowrousian, M. Sordaria macrospora: 25 years as a model organism for studying the molecular mechanisms of fruiting body development. Appl. Microbiol. Biotechnol. 2020, 104, 3691–3704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Elleuche, S.; Poggeler, S. Carbonic anhydrases in fungi. Microbiology 2010, 156, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wang, L.; Clavaud, C.; Bar-Hen, A.; Cui, M.; Gao, J.; Liu, Y.; Liu, C.; Shibagaki, N.; Gueniche, A.; Jourdain, R.; et al. Characterization of the major bacterial-fungal populations colonizing dandruff scalps in Shanghai, China, shows microbial disequilibrium. Exp. Dermatol. 2015, 24, 398–400. [Google Scholar] [CrossRef] [PubMed]
  86. De Simone, G.; Supuran, C.T. (In)organic anions as carbonic anhydrase inhibitors. J. Inorg. Biochem. 2012, 111, 117–129. [Google Scholar] [CrossRef] [PubMed]
  87. Nocentini, A.; Cadoni, R.; Del Prete, S.; Capasso, C.; Dumy, P.; Gratteri, P.; Supuran, C.T.; Winum, J.Y. Benzoxaboroles as Efficient Inhibitors of the beta-Carbonic Anhydrases from Pathogenic Fungi: Activity and Modeling Study. ACS Med. Chem. Lett. 2017, 8, 1194–1198. [Google Scholar] [CrossRef] [PubMed]
  88. Nocentini, A.; Vullo, D.; Del Prete, S.; Osman, S.M.; Alasmary, F.A.S.; AlOthman, Z.; Capasso, C.; Carta, F.; Gratteri, P.; Supuran, C.T. Inhibition of the beta-carbonic anhydrase from the dandruff-producing fungus Malassezia globosa with monothiocarbamates. J. Enzym. Inhib. Med. Chem. 2017, 32, 1064–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Carta, F.; Aggarwal, M.; Maresca, A.; Scozzafava, A.; McKenna, R.; Masini, E.; Supuran, C.T. Dithiocarbamates strongly inhibit carbonic anhydrases and show antiglaucoma action in vivo. J. Med. Chem. 2012, 55, 1721–1730. [Google Scholar] [CrossRef] [Green Version]
  90. Carta, F.; Aggarwal, M.; Maresca, A.; Scozzafava, A.; McKenna, R.; Supuran, C.T. Dithiocarbamates: A new class of carbonic anhydrase inhibitors. Crystallographic and kinetic investigations. Chem. Commun. 2012, 48, 1868–1870. [Google Scholar] [CrossRef] [Green Version]
  91. Monti, S.M.; Maresca, A.; Viparelli, F.; Carta, F.; De Simone, G.; Muhlschlegel, F.A.; Scozzafava, A.; Supuran, C.T. Dithiocarbamates are strong inhibitors of the beta-class fungal carbonic anhydrases from Cryptococcus neoformans, Candida albicans and Candida glabrata. Bioorg. Med. Chem. Lett. 2012, 22, 859–862. [Google Scholar] [CrossRef] [Green Version]
  92. Maresca, A.; Carta, F.; Vullo, D.; Supuran, C.T. Dithiocarbamates strongly inhibit the beta-class carbonic anhydrases from Mycobacterium tuberculosis. J. Enzym. Inhib. Med. Chem. 2013, 28, 407–411. [Google Scholar] [CrossRef]
  93. Scozzafava, A.; Mastrolorenzo, A.; Supuran, C.T. Arylsulfonyl-N,N-dialkyl-dithiocarbamates as tumor cell growth inhibitors: Novel agents targeting beta-tubulin? J. Enzym. Inhib. Med. Chem. 2001, 16, 55–63. [Google Scholar] [CrossRef] [PubMed]
  94. Scozzafava, A.; Mastrolorenzo, A.; Supuran, C.T. Arylsulfonyl-N,N-diethyl-dithiocarbamates: A novel class of antitumor agents. Bioorg. Med. Chem. Lett. 2000, 10, 1887–1891. [Google Scholar] [CrossRef]
  95. Innocenti, A.; Scozzafava, A.; Supuran, C.T. Carbonic anhydrase inhibitors. Inhibition of transmembrane isoforms IX, XII, and XIV with less investigated anions including trithiocarbonate and dithiocarbamate. Bioorg. Med. Chem. Lett. 2010, 20, 1548–1550. [Google Scholar] [CrossRef] [PubMed]
  96. Supuran, C.T. Structure-based drug discovery of carbonic anhydrase inhibitors. J. Enzym. Inhib. Med. Chem. 2012, 27, 759–772. [Google Scholar] [CrossRef] [PubMed]
  97. Adak, A.K.; Leonov, A.P.; Ding, N.; Thundimadathil, J.; Kularatne, S.; Low, P.S.; Wei, A. Bishydrazide glycoconjugates for lectin recognition and capture of bacterial pathogens. Bioconjug. Chem. 2010, 21, 2065–2075. [Google Scholar] [CrossRef] [Green Version]
  98. Lomelino, C.L.; Supuran, C.T.; McKenna, R. Non-Classical Inhibition of Carbonic Anhydrase. Int. J. Mol. Sci. 2016, 17, 1150. [Google Scholar] [CrossRef] [Green Version]
  99. Resnick, M.A.; Cox, B.S. Yeast as an honorary mammal. Mutat. Res. 2000, 451, 1–11. [Google Scholar] [CrossRef]
  100. Stefanini, I.; Trabocchi, A.; Marchi, E.; Guarna, A.; Cavalieri, D. A systems biology approach to dissection of the effects of small bicyclic peptidomimetics on a panel of Saccharomyces cerevisiae mutants. J. Biol. Chem. 2010, 285, 23477–23485. [Google Scholar] [CrossRef] [Green Version]
  101. Alafeefy, A.M.; Isik, S.; Al-Jaber, N.A.; Vullo, D.; Abdel-Aziz, H.A.; Ashour, A.E.; Awaad, A.S.; Capasso, C.; Supuran, C.T. Carbonic anhydrase inhibitors. Benzenesulfonamides incorporating cyanoacrylamide moieties strongly inhibit Saccharomyces cerevisiae beta-carbonic anhydrase. Bioorg. Med. Chem. Lett. 2013, 23, 3570–3575. [Google Scholar] [CrossRef]
  102. Isik, S.; Guler, O.O.; Kockar, F.; Aydin, M.; Arslan, O.; Supuran, C.T. Saccharomyces cerevisiae beta-carbonic anhydrase: Inhibition and activation studies. Curr. Pharm. Des. 2010, 16, 3327–3336. [Google Scholar] [CrossRef]
  103. Bozdag, M.; Carta, F.; Vullo, D.; Isik, S.; AlOthman, Z.; Osman, S.M.; Scozzafava, A.; Supuran, C.T. Dithiocarbamates with potent inhibitory activity against the Saccharomyces cerevisiae beta-carbonic anhydrase. J. Enzym. Inhib. Med. Chem. 2016, 31, 132–136. [Google Scholar] [CrossRef] [PubMed]
  104. Bilginer, S.; Unluer, E.; Gul, H.I.; Mete, E.; Isik, S.; Vullo, D.; Ozensoy-Guler, O.; Beyaztas, S.; Capasso, C.; Supuran, C.T. Carbonic anhydrase inhibitors. Phenols incorporating 2- or 3-pyridyl-ethenylcarbonyl and tertiary amine moieties strongly inhibit Saccharomyces cerevisiae beta-carbonic anhydrase. J. Enzym. Inhib. Med. Chem. 2014, 29, 495–499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Fidel, P.L., Jr.; Vazquez, J.A.; Sobel, J.D. Candida glabrata: Review of epidemiology, pathogenesis, and clinical disease with comparison to C. albicans. Clin. Microbiol. Rev. 1999, 12, 80–96. [Google Scholar] [CrossRef] [Green Version]
  106. Innocenti, A.; Leewattanapasuk, W.; Muhlschlegel, F.A.; Mastrolorenzo, A.; Supuran, C.T. Carbonic anhydrase inhibitors. Inhibition of the beta-class enzyme from the pathogenic yeast Candida glabrata with anions. Bioorg. Med. Chem. Lett. 2009, 19, 4802–4805. [Google Scholar] [CrossRef] [PubMed]
  107. Aksu, K.; Nar, M.; Tanc, M.; Vullo, D.; Gulcin, I.; Goksu, S.; Tumer, F.; Supuran, C.T. Synthesis and carbonic anhydrase inhibitory properties of sulfamides structurally related to dopamine. Bioorg. Med. Chem. 2013, 21, 2925–2931. [Google Scholar] [CrossRef] [PubMed]
  108. Elleuche, S.; Poggeler, S. Evolution of carbonic anhydrases in fungi. Curr. Genet. 2009, 55, 211–222. [Google Scholar] [CrossRef] [Green Version]
  109. Strop, P.; Smith, K.S.; Iverson, T.M.; Ferry, J.G.; Rees, D.C. Crystal structure of the “cab”-type beta class carbonic anhydrase from the archaeon Methanobacterium thermoautotrophicum. J. Biol. Chem. 2001, 276, 10299–10305. [Google Scholar] [CrossRef] [Green Version]
  110. Vullo, D.; Lehneck, R.; Donald, W.A.; Poggeler, S.; Supuran, C.T. Anion Inhibition Studies of the beta-Class Carbonic Anhydrase CAS3 from the Filamentous Ascomycete Sordaria macrospora. Metabolites 2020, 10, 93. [Google Scholar] [CrossRef] [Green Version]
  111. Vullo, D.; Lehneck, R.; Donald, W.A.; Poggeler, S.; Supuran, C.T. Sulfonamide Inhibition Studies of the beta-Class Carbonic Anhydrase CAS3 from the Filamentous Ascomycete Sordaria macrospora. Molecules 2020, 25, 1036. [Google Scholar] [CrossRef] [Green Version]
  112. Vullo, D.; Lehneck, R.; Poggeler, S.; Supuran, C.T. Sulfonamide inhibition studies of two beta-carbonic anhydrases from the ascomycete fungus Sordaria macrospora, CAS1 and CAS2. J. Enzym. Inhib. Med. Chem. 2018, 33, 390–396. [Google Scholar] [CrossRef] [Green Version]
  113. Barac, A.; Pekmezovic, M.; Milobratovic, D.; Otasevic-Tasic, S.; Radunovic, M.; Arsic Arsenijevic, V. Presence, species distribution, and density of Malassezia yeast in patients with seborrhoeic dermatitis—A community-based case-control study and review of literature. Mycoses 2015, 58, 69–75. [Google Scholar] [CrossRef] [PubMed]
  114. Gaitanis, G.; Magiatis, P.; Hantschke, M.; Bassukas, I.D.; Velegraki, A. The Malassezia genus in skin and systemic diseases. Clin. Microbiol. Rev. 2012, 25, 106–141. [Google Scholar] [CrossRef] [Green Version]
  115. Stalhberger, T.; Simenel, C.; Clavaud, C.; Eijsink, V.G.; Jourdain, R.; Delepierre, M.; Latge, J.P.; Breton, L.; Fontaine, T. Chemical organization of the cell wall polysaccharide core of Malassezia restricta. J. Biol. Chem. 2014, 289, 12647–12656. [Google Scholar] [CrossRef] [Green Version]
  116. Grice, E.A.; Dawson, T.L., Jr. Host-microbe interactions: Malassezia and human skin. Curr. Opin. Microbiol. 2017, 40, 81–87. [Google Scholar] [CrossRef] [PubMed]
  117. Loden, M.; Wessman, C. The antidandruff efficacy of a shampoo containing piroctone olamine and salicylic acid in comparison to that of a zinc pyrithione shampoo. Int. J. Cosmet. Sci. 2000, 22, 285–289. [Google Scholar] [CrossRef] [PubMed]
  118. Hay, R.J. Malassezia, dandruff and seborrhoeic dermatitis: An overview. Br. J. Dermatol. 2011, 165 (Suppl. S2), 2–8. [Google Scholar] [CrossRef]
  119. Del Prete, S.; Vullo, D.; Osman, S.M.; AlOthman, Z.; Capasso, C.; Supuran, C.T. Anion inhibition studies of the dandruff-producing fungus Malassezia globosa beta-carbonic anhydrase MgCA. Bioorg. Med. Chem. Lett. 2015, 25, 5194–5198. [Google Scholar] [CrossRef]
  120. Del Prete, S.; Angeli, A.; Ghobril, C.; Hitce, J.; Clavaud, C.; Marat, X.; Supuran, C.T.; Capasso, C. Sulfonamide Inhibition Profile of the beta-Carbonic Anhydrase from Malassezia restricta, An Opportunistic Pathogen Triggering Scalp Conditions. Metabolites 2020, 10, 39. [Google Scholar] [CrossRef] [Green Version]
  121. Vullo, D.; Del Prete, S.; Nocentini, A.; Osman, S.M.; AlOthman, Z.; Capasso, C.; Bozdag, M.; Carta, F.; Gratteri, P.; Supuran, C.T. Dithiocarbamates effectively inhibit the beta-carbonic anhydrase from the dandruff-producing fungus Malassezia globosa. Bioorg. Med. Chem. 2017, 25, 1260–1265. [Google Scholar] [CrossRef]
  122. Entezari Heravi, Y.; Bua, S.; Nocentini, A.; Del Prete, S.; Saboury, A.A.; Sereshti, H.; Capasso, C.; Gratteri, P.; Supuran, C.T. Inhibition of Malassezia globosa carbonic anhydrase with phenols. Bioorg. Med. Chem. 2017, 25, 2577–2582. [Google Scholar] [CrossRef]
  123. Paloukopoulou, C.; Govari, S.; Soulioti, A.; Stefanis, I.; Angeli, A.; Matheeussen, A.; Capasso, C.; Cos, P.; Supuran, C.T.; Karioti, A. Phenols from Origanum dictamnus L. and Thymus vulgaris L. and their activity against Malassezia globosa carbonic anhydrase. Nat. Prod. Res. 2021, 1–7. [Google Scholar] [CrossRef] [PubMed]
  124. Alissa, S.A.; Alghulikah, H.A.; Alothman, Z.A.; Osman, S.M.; Del Prete, S.; Capasso, C.; Nocentini, A.; Supuran, C.T. Phosphonamidates are the first phosphorus-based zinc binding motif to show inhibition of beta-class carbonic anhydrases from bacteria, fungi, and protozoa. J. Enzym. Inhib. Med. Chem. 2020, 35, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Del Prete, S.; Vullo, D.; Ghobril, C.; Hitce, J.; Clavaud, C.; Marat, X.; Capasso, C.; Supuran, C.T. Cloning, Purification, and Characterization of a beta-Carbonic Anhydrase from Malassezia restricta, an Opportunistic Pathogen Involved in Dandruff and Seborrheic Dermatitis. Int. J. Mol. Sci. 2019, 20, 2447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Prete, S.D.; Angeli, A.; Ghobril, C.; Hitce, J.; Clavaud, C.; Marat, X.; Supuran, C.T.; Capasso, C. Anion Inhibition Profile of the beta-Carbonic Anhydrase from the Opportunist Pathogenic Fungus Malassezia Restricta Involved in Dandruff and Seborrheic Dermatitis. Metabolites 2019, 9, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Sulfonamide/sulfamate/sulfamide of types 124 (pink background) and AAZ-EPA (gray background) investigated as fungal CA inhibitors. Legend: AAZ, acetazolamide; MZA, methazolamide; EZA, ethoxzolamide; DCP, dichlorophenamide; DZA, dorzolamide; BRZ, brinznolamide; BZA, benzolamide; TPM, topiramate; ZNS, zonisamide; SLP, sulpiride; IND, indisulam; VLX, valdecoxib; CLX, celecoxib; SLT, sulthiame; HCT, hydrochlorothiazide; FAM, famotidine; EPA, epacadostat.
Figure 1. Sulfonamide/sulfamate/sulfamide of types 124 (pink background) and AAZ-EPA (gray background) investigated as fungal CA inhibitors. Legend: AAZ, acetazolamide; MZA, methazolamide; EZA, ethoxzolamide; DCP, dichlorophenamide; DZA, dorzolamide; BRZ, brinznolamide; BZA, benzolamide; TPM, topiramate; ZNS, zonisamide; SLP, sulpiride; IND, indisulam; VLX, valdecoxib; CLX, celecoxib; SLT, sulthiame; HCT, hydrochlorothiazide; FAM, famotidine; EPA, epacadostat.
Ijms 22 04324 g001
Figure 2. Active site view of Can2 (pdb 2W3N) complexed to the anion inhibitor acetate [45]. Protomers A and B are colored green and cyan respectively. Residues from protomers A and B are labeled black and light blu, respectively. The Zn2+ ion, represented as a grey sphere, is coordinated by two cysteines and one histidine residue from monomer A and by one acetate ion as a ligand. The salt bridge in the Asp-Arg dyad is represented as a yellow dashed line.
Figure 2. Active site view of Can2 (pdb 2W3N) complexed to the anion inhibitor acetate [45]. Protomers A and B are colored green and cyan respectively. Residues from protomers A and B are labeled black and light blu, respectively. The Zn2+ ion, represented as a grey sphere, is coordinated by two cysteines and one histidine residue from monomer A and by one acetate ion as a ligand. The salt bridge in the Asp-Arg dyad is represented as a yellow dashed line.
Ijms 22 04324 g002
Table 1. Inhibition data of human isoenzymes (CA I and CA II) and fungal CAs (MreCA, MgCA, CAS1, CAS2, CAS3; Figure 1) by a stopped-flow CO2 hydrase assay. The other fungal CAs have been previously reviewed by Elleuche and Poggeler (see references [81,82]).
Table 1. Inhibition data of human isoenzymes (CA I and CA II) and fungal CAs (MreCA, MgCA, CAS1, CAS2, CAS3; Figure 1) by a stopped-flow CO2 hydrase assay. The other fungal CAs have been previously reviewed by Elleuche and Poggeler (see references [81,82]).
InhibitorKI (nM) *
hCA I 1
(α-CA)
hCA II 1
(α-CA)
MgCA 2
(β-CA)
MreCA 2
(β-CA)
CAS1 1
(β-CA)
CAS2 1
(β-CA)
CAS3 1
(β-CA)
128,00030098041236138690
225,00024024.5462144348084
379815.2>10,000225363083
478,50032067440447.16900560
525,00017017.4>10,0003238720726
621,0001607.9>10,0002417650441
783006011.645943.27360585
8980011012.1>10,00079.691202078
965004034.9>10,00058012,000712
1073005454.3>10,000>50,00023,500350
11580063967689018,700235
128400759.2>10,0003350>50,00090
138600607900>10,000865048.188
149300198500>10,000721528094
1555008023.6>10,0003160143605
1695009410.4651452092.582
1721,0001256.3>10,000>50,000390507
18164466.8>10,00044433250226
19109333500779475676091
206223.491363988085
21691111.87404550406095
22164469.4374198525,20085
23109334530>10,000282>50,00089
249530256>10,000294>50,00084
AAZ2501276001044581694
MZA50147455390421814091
EZA2583800379440317095
DCP12003834.63061220579073
DZA50,0009790081360742274
BRZ45,000384007045173961
BZA15948.2715211541054
TPM25010146383414673363
ZNS5635765>10,00018201885710
SLP120040324851715670493
IND3115n.d.87424021694
VLX54,0004331507744253730831
CLX50,0002134801402513857669
SLT3749n.d.6732104964838
SAC18,5405959n.d.62052807075191
HCT328290n.d.85033506680545
FAMn.d.n.d.n.d.>10,000n.d.n.d.n.d.
EPAn.d.n.d.n.d.n.d.n.d.n.d.n.d.
* Errors were in the range of ±5–10% on three different assays. 1 From reference [83] and [84]; 2 From reference [85]; n.d.: not detected.
Table 2. Inhibition constants obtained using anionic inhibitors versus the α-CA isozymes of human origin (hCA I and hCA II), and Table 1. CAS2, CAS3; for the acronyms, see the text) by a stopped flow CO2 hydrase assay.
Table 2. Inhibition constants obtained using anionic inhibitors versus the α-CA isozymes of human origin (hCA I and hCA II), and Table 1. CAS2, CAS3; for the acronyms, see the text) by a stopped flow CO2 hydrase assay.
AnionKI (mM) *
hCA I 1
(α-CA)
hCA II 1
(α-CA)
MgCA 2
(β-CA)
MreCA 2
(β-CA)
CAS1 1
(β-CA)
CAS2 1
(β-CA)
CAS3 1
(β-CA)
F>300>3007.13>50>100>100>100
Cl62007.98>509.2>100>100
Br46318.6>509.3>100>100
I0.326 8.738.68.67.79.9
CNO0.00070.036.81>500.90.823.2
SCN0.21.608.39>505.45.67.3
CN0.00050.027.19>500.940.758.7
N30.00121.5145.2>50>1006.17.2
NO28.4637.56>50>100>1008.3
NO37358.139>100>1008.5
HCO312850.590.863.37.3>100
CO32−1573>100>50>1008.88
HSO31889>100>503.37.3>100
SO42−63>20019.5>50>1004.8>100
HS0.00060.0411.9>500.898.58.3
SnO32−0.570.835.070.564.30.927.9
SeO42−1181127.411.72.49.23.4
TeO42−0.660.925.750.562.56.38.1
OsO52−0.920.956.168.5n.d.n.d.n.d.
P2O74−25.7748.506.03>503.10.96>100
V2O74−0.540.576.89>50>1001.4>100
B4O72−0.640.958.450.46.76.95.9
ReO40.110.7516.7>508.2>1008.8
RuO40.1010.698.827.43.9>1009.2
S2O82−0.1070.084>100>505>100>100
SeCN0.0850.0861.730.652.99.37.1
CS32−0.00870.00881.770.920.79>1008.6
Et2NCS20.000790.00310.300.0750.380.930.89
CF3SO3n.d.n.d.2.284.5n.d.n.d.n.d.
PF6n.d.n.d.6.473.9n.d.n.d.n.d.
ClO4>200>200>1009.2>100>100>100
BF4>200>200>100383>100>100>100
FSO30.790.464.06>500.938.4>100
NH(SO3)22−0.310.7621.4>500.889.2>100
H2NSO2NH0.311.130.0940.720.0840.0480.094
H2NSO3H0.0210.390.0837.70.0690.0720.095
Ph-B(OH)258.623.10.0898.70.0090.0560.097
Ph-AsO3H231.749.20.0900.830.0350.0540.091
* Errors were in the range of ±5–10% on three different assays. 1 From reference [87]; 2 From reference [88]; n.d.: not detected.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Supuran, C.T.; Capasso, C. A Highlight on the Inhibition of Fungal Carbonic Anhydrases as Drug Targets for the Antifungal Armamentarium. Int. J. Mol. Sci. 2021, 22, 4324. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094324

AMA Style

Supuran CT, Capasso C. A Highlight on the Inhibition of Fungal Carbonic Anhydrases as Drug Targets for the Antifungal Armamentarium. International Journal of Molecular Sciences. 2021; 22(9):4324. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094324

Chicago/Turabian Style

Supuran, Claudiu T., and Clemente Capasso. 2021. "A Highlight on the Inhibition of Fungal Carbonic Anhydrases as Drug Targets for the Antifungal Armamentarium" International Journal of Molecular Sciences 22, no. 9: 4324. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094324

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