Next Article in Journal
Long-Term Effectiveness of BNT162b2 Pfizer-BioNTech mRNA-Based Vaccine on B Cell Compartment: Efficient Recall of SARS-CoV-2-Specific Memory B Cells
Previous Article in Journal
Optogenetic Therapy for Visual Restoration
Previous Article in Special Issue
Comparative Genomic Analysis of Agarolytic Flavobacterium faecale WV33T
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Polyenic Antibiotics and Other Antifungal Compounds Produced by Hemolytic Streptomyces Species

1
Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studničkova 7, 128 00 Prague, Czech Republic
2
Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, České mládeže 8, 400 96 Ústí nad Labem, Czech Republic
3
Contipro a.s., Dolní Dobrouč 401, 561 02 Dolní Dobrouč, Czech Republic
4
Institute of Soil Biology and Biogeochemistry, Biology Centre of the Czech Academy of Sciences, Na Sádkách 7, 370 05 České Budějovice, Czech Republic
5
Doctoral School of Environmental Sciences, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
6
Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 31, 370 05 České Budějovice, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15045; https://doi.org/10.3390/ijms232315045
Submission received: 27 September 2022 / Revised: 14 November 2022 / Accepted: 26 November 2022 / Published: 30 November 2022
(This article belongs to the Special Issue Microbial Genomics and Biosynthesis)

Abstract

:
Streptomyces are of great interest in the pharmaceutical industry as they produce a plethora of secondary metabolites that act as antibacterial and antifungal agents. They may thrive on their own in the soil, or associate with other organisms, such as plants or invertebrates. Some soil-derived strains exhibit hemolytic properties when cultivated on blood agar, raising the question of whether hemolysis could be a virulence factor of the bacteria. In this work we examined hemolytic compound production in 23 β-hemolytic Streptomyces isolates; of these 12 were soil-derived, 10 were arthropod-associated, and 1 was plant-associated. An additional human-associated S. sp. TR1341 served as a control. Mass spectrometry analysis suggested synthesis of polyene molecules responsible for the hemolysis: candicidins, filipins, strevertene A, tetrafungin, and tetrin A, as well as four novel polyene compounds (denoted here as polyene A, B, C, and D) in individual liquid cultures or paired co-cultures. The non-polyene antifungal compounds actiphenol and surugamide A were also identified. The findings indicate that the ability of Streptomyces to produce cytolytic compounds (here manifested by hemolysis on blood agar) is an intrinsic feature of the bacteria in the soil environment and could even serve as a virulence factor when colonizing available host organisms. Additionally, a literature review of polyenes and non-polyene hemolytic metabolites produced by Streptomyces is presented.

1. Introduction

1.1. The Role of Hemolysis in Streptomyces’ Interactions

Streptomyces produce a large variety of secondary metabolites (SM) that correspond to their environmental needs [1], of which antibacterial and antifungal agents are of utmost importance [2]. The chemical diversity of the SMs produced by Streptomyces species has most likely evolved as a direct result of their interactions with other organisms. Streptomyces’ interactions with plants and animals can be parasitic, as is the case of potato scab-causing streptomycetes, which infect the plant tuberosphere [3], or S. somaliensis and other strains that infect humans [4]. However, in most cases, they are beneficial and growth- promoting organisms. Streptomycetes form numerous mutualistic relationships with invertebrates and plants [5], and they protect their hosts against infection using antibiotics and antifungals.
Hemolysis—the rupture of erythrocytes—is a virulence factor of many pathogens with E. coli, Streptococci, Vibrio, and Staphylococcus aureus being some prominent examples [6]. The activity of hemolysins, however, is not restricted to erythrocytes only; hemolysins, by acting on cellular membranes, can also damage other eukaryotic cells [6,7]. Hemolysins are mostly lytic proteins—enzymes or channel-forming porins [8] or, rarely, polyene compounds (also referred to as polyene antibiotics or polyene antimycotics) [9].

1.2. Polyenes and Non-Polyene Hemolytic Metabolites: A Literature Review

Polyenes are poly-unsaturated compounds with linear or cyclic structures. Cyclic polyene antibiotics belong to the macrolide class of SMs, representing a large and variable group of antibiotics produced mostly by Streptomyces. They have a macrolactone ring to which typically two sugars and one amino sugar are attached [10]. Antibiotics in the polyene class possess a macrocyclic ring of carbon atoms closed by lactonization; the polyene group has, in addition, a series of conjugated double bonds of various length.
We searched the literature extensively to review mostly actinomycete-derived polyenes identified so far. They are listed in groups based on their structures together with their chemical formulas, molecular weight (MW), and assessed activities (Table 1); the representative structures are shown in Figure S1. Selected non-polyene, human cells-targeting metabolites included in the study are placed in Table 2. The primary selection criterion was the origin of compounds in streptomycetes or related bacteria and their reported activity towards eukaryotic cells. The majority of the compounds were discovered more than 50 years ago, therefore the data often lack sufficient structure determination and complex activity screenings. Crucial structural characteristics include a combination of a hydrophobic polyene region with a hydrophilic polyol part, often glycosylated, which enables the molecules to enter the cytoplasmic membranes of various organisms, exhibiting either irreversible destruction of the membrane or transient and reversible channel formation (e.g., in pentaenes or heptaenes, respectively). They form complexes, in which the polyene chain faces the lipid environment and the polyol chain is oriented towards the aqueous environment in the interior of the pores [11]. Polyenes often form complexes with sterols and exhibit variable specificities to ergosterols and cholesterols [12]. These traits strongly influence their cytotoxicity and, subsequently, their possible medical application.
Almost all polyene compounds have been identified due to their antifungal activity [11] and some have been shown to possess other bioactivities, such as antibacterial (often targeting cell-wall lacking bacteria), antiparasitic (anti-Trichomonas activity has been reported most frequently, implying the compounds may find use in the treatment of combined vaginal infections), cytolytic, and anti-cancer activities.
Most of polyene antibiotics, including filipin and candicidin compounds with larger rings, exhibit hemolytic properties [89]. Filipin forms large aggregates within the erythrocyte membrane that render it permeable [90]. Other polyenes, however, impair plasma membranes by direct binding to ergosterol, as is the case of natamycin [91] or amphotericin [92]. All these polyene compounds are fungicidal and those with lower toxicity are used in medicine. For example, amphotericin B, natamycin and nystatin A1 are used in antifungal and antiprotozoal medications [90].

1.3. Hemolysis as a Virulence Factor

Whilst SM production in Actinobacteria has been extensively studied [2,69], the impact of hemolytic metabolites has not received much attention so far. As hemolysis can be considered a virulence factor [93,94], it may well be viewed as one of the adaptations that the bacteria employ to compete with other organisms in their environment. This theory is further supported by the example of the streptomycete strain S. sp. TR1341, extracted from the lungs of a senior male patient with relapsing bronchopneumonia, whose taxonomy indicates a distance from plant and human pathogenic strains [89]. It has been demonstrated that S. sp. TR1341 possesses a filipin biosynthetic gene cluster responsible for the bacteria’s hemolytic capabilities [89].
The β-hemolytic activity is not exclusively related to human-associated strains, but also occurs in soil-dwelling strains. About half of soil-derived Streptomyces strains exhibit β-hemolytic activity, whereas three out of four clinical Streptomyces isolates are β-hemolytic, according to Wurzer [95]. The soil-derived strains exhibiting β-hemolysis were collected (BCCO strains) and used here to search for the β-hemolytic compound production. The search was focused on polyene antibiotics as their role as hemolysins has not yet been systematically studied in Streptomyces. In addition, the 16S rRNA phylogeny of the BCCO strains used in this study was compared to that of well-known polyene producers.

2. Results

2.1. Sequencing Data and Phylogenetic Tree

According to the 16S rRNA-encoding gene similarities, 22 BCCO strains with β-hemolytic activities clustered with other Streptomyces and one strain with Nocardioides (BCCO 10_0486). Phylogenetic analysis showed no clear association between phylogeny and the production of particular polyene compounds (Figure 1); the clustering seems to correlate more with the isolation source (arthropod or soil) or the country of origin. However, there are a few clusters that contain strains of various origins (country/source): (i) BCCO 10_1099 (Papua New Guinea/ambrosia beetle) and BCCO 10_2196 (Czechia/millipede) clustered together with S. albidoflavus DSM40455; (ii) BCCO 10_0670 (Czechia/soil) and BCCO 10_1092 (Papua New Guinea/ambrosia beetle) clustered together with S. griseus subsp. griseus KCTC9080; and (iii) BCCO 10_1747 (Czechia/soil) and BCCO 10_2389 (Hungary/soil) clustered as S. drozdowiczii NRRL-B-24297.
Apparently, filipin, candicidin, nystatin A1, pimaricin, and actiphenol can be produced by strains from different phylogenetic groups. Our analysis suggests that strains with high phylogenetic relatedness and originating from the same habitat and country produce either the same polyene B (strains associated with ambrosia beetle from Papua New Guinea: BCCO 10_1093, 10_1095, 10_1104) or different compounds (soil strains from Hungary: BCCO 10_2295, 10_2309, 10_2325), as can be seen in our results below.

2.2. Characteristics of Morphological Differentiation and Hemolytic Activities in the Strains

Investigated strains were streaked on blood agar plates, and after 120 h of cultivation, pictures of growing mycelia and hemolytic zones were taken from the top and bottom sides of the dish. All lab-tested strains expressed β-hemolytic activity when grown on blood agar (Table S2).

2.3. Gamma-Butyrolactone-Induced Polyene Production

Gamma-butyrolactone (GBL) is an activator of SM production in Streptomyces. To test its capacity to induce polyene production, we inoculated a strain of Streptomyces as one line on the blood agar. Five microliters of GBL solution (ReagentPlus, ≥99%, Sigma-Aldrich Co. St. Louis, MO, USA) were dropped on a sterile filter paper strip which was then inserted perpendicularly to the line of Streptomyces. An increase in the hemolytic zone was observed after O/N cultivation on the cross-sections with the GBL-soaked paper strip in BCCO 10_0524, BCCO 10_0670, BCCO 10_1093, BCCO 10_1747, BCCO 10_2179, and BCCO 10_2389 (Figure S2). Remaining strains did not reveal any phenotypic difference in the presence of GBL.

2.4. Hemolytic Activity of Ethyl Acetate Extracts

To determine whether SMs produced by Streptomyces spp. into the medium could cause hemolysis, the culture supernatants were subjected to ethyl acetate extraction. A similar approach has already been used [89] to demonstrate that the polyene compound filipin is the only compound responsible for the hemolytic activity of their tested strain. The crude extracts (5 μL) were dropped on blood agar plates and incubated for 3 days with 5 μL of chloroform in the middle as a negative control (Figure S3). These experiments were performed in duplicate. Hemolytic activity was spotted on the blood agar for BCCO 10_1093, BCCO 10_1094, BCCO 10_1095, BCCO 10_1106, and BCCO 10_1499 strains. No hemolytic zone was observed in samples isolated from the BCCO 10_1099 strain and from the negative control. Nevertheless, the ethyl acetate extraction procedure led to a considerable reduction in the hemolytic activity when compared to hemolytic zones produced by the cell-free supernatants shown in Figure 2 (see below).

2.5. LC-MS Analysis of the Supernatant Extracts

After cultivation in liquid medium with or without blood, SPE was performed using the supernatants, followed by LC-MS. The presence of the polyenes listed in Table 1 was assayed in the metabolic extracts of selected beta-hemolytic streptomycete strains. The length of the polyene part influences the typical three-peak UV-VIS spectrum of the compounds [42]. This was used as a clue to identify putative, so far uncharacterized, polyenes in some extracts. The LC-MS analysis results are summarized in Table S3. The metabolites found are also listed in Figure 1 and Table S2.
Out of the 23 β-hemolytic tested strains, known polyene substances were likely detected in 12 strains (Table S2). Of these, candicidins A1–A3 were detected in two strains (BCCO 10_0524, BCCO 10_1099). Strevertene A was detected in two strains (BCCO 10_1499, BCCO 10_2155) and filipin III in BCCO 10_2155. Tetrafungin and tetrin A were found in BCCO 10_2325, BCCO 10_1092, BCCO 10_1093, BCCO 10_1094, BCCO 10_1095, BCCO 10_1104, and BCCO 10_2179.
A new polyene (retention time (tR) = 9.42 min, [M+H]+ m/z = 745.4166, UV/VIS wavelength of maximum absorbance: (289 nm), 327 nm, 343 nm, 362 nm), designated here as polyene B, was likely detected in 10 strains (BCCO 10_1092, BCCO 10_1093, BCCO 10_1094, BCCO 10_1095, BCCO 10_1097, BCCO 10_1104, BCCO 10_2179, BCCO 10_2282, BCCO 10_2325, and BCCO 10_2389). Its absorption spectrum suggests the presence of a pentaene structure, and the production is often associated with the formation of tetrafungin and tetrin A compounds (Table S2). The second novel polyene compound (tR = 8.23 min, UV/VIS wavelength of maximum absorbance: 311 nm, 326 nm, 343 nm) was designated polyene A and was present only in the extract of BCCO 10_1106 strain. The third, polyene D, tR = 10.6 min, UV/VIS wavelength of maximum absorbance: 287 nm, 345 nm, 362 nm, 384 nm, most probably has hexaene or methylhexaene structure [42]. No polyene-like compound was detected in individual cultures of five β-hemolytic strains (BCCO 10_0670, BCCO 10_1331, BCCO 10_2259, BCCO 10_2295, BCCO 10_2309), in both 7% blood-containing and blood-free media.
Besides the above-described polyenes, the extracts were screened for the presence of non-polyene, human cells-targeting SMs (Table 2). Of these, we identified Surugamide A, a non-ribosomal peptide with cathepsin B-inhibitory activity [70], in BCCO 10_0524 and BCCO 10_1099, the two strains that also produced candicidin A1-A3. The latter strain belongs to the S. albidoflavus clade according to our phylogenetic tree. In a recent study, S. albidoflavus J11074 has also been reported as a surugamide producer during cultivation under stress conditions [68].

2.6. Selected Hemolytic Activity Is Likely Not Due to Lytic Proteins

Four β-hemolytic strains (BCCO 10_1331, BCCO 10_2259, BCCO 10_2295, BCCO 10_2309), in which no polyene metabolite, and no other metabolites with possible hemolytic activity, were detected following standard cultivation, were further tested to show whether their hemolytic activity could be caused by extracellular protein(s). Each cell-free supernatant from these four 72-h-old cultures was split into three thirds: one was treated by proteinase K (1 μg/mL) for 60 min at 37 °C (to degrade extracellular hemolytic proteins), the second was incubated in the same conditions without the enzyme (serving as a negative control in which hemolytic activity was expected), and the third sample was incubated at 100 °C for 5 min (we expected it to lose its hemolytic activity completely). All samples lost their hemolytic activity after boiling, but not after the proteinase K treatment, suggesting that the hemolytic activity is associated with the SMs production rather than hemolytic proteins [97] in the tested streptomycetes (Figure 2).
Figure 2. Hemolytic activity of supernatants after proteinase K treatment or boiling. Cell-free supernatants from 72-h-old cultures of strains BCCO 10_1331, BCCO 10_2259, BCCO 10_2295, BCCO 10_2309 (listed from left) were treated by proteinase K (upper discs on each plate) or were boiled (discs on the left of each plate), or no additional treatment have been performed (control discs on the right side of each plate).
Figure 2. Hemolytic activity of supernatants after proteinase K treatment or boiling. Cell-free supernatants from 72-h-old cultures of strains BCCO 10_1331, BCCO 10_2259, BCCO 10_2295, BCCO 10_2309 (listed from left) were treated by proteinase K (upper discs on each plate) or were boiled (discs on the left of each plate), or no additional treatment have been performed (control discs on the right side of each plate).
Ijms 23 15045 g002

2.7. Hemolytic Compounds Identified in Paired Co-Cultures

Four β-hemolytic strains (BCCO 10_1331, BCCO 10_2259, BCCO 10_2295, BCCO 10_2309), in which no polyene compound was detected, nor was hemolytic activity unique for extracytoplasmic proteins in those strains (see Section 2.7), were subjected to paired co-cultures to promote the production of SMs in these strains. In the sample isolated from the paired co-culture [BCCO 10_1331 + BCCO 10_2309] we detected a fourth novel polyene-like compound (UV/VIS wavelength of maximum absorbance: 346 nm, 364 nm, and 386 nm; tR = 7.77 min, acquired [M+H]+ = 743.4131), here designated as polyene C, with a putative (methyl-)hexaene structure. The second paired co-culture [BCCO 10_2259 + BCCO 10_2295] revealed a known hemolytic compound, namely actiphenol (a non-polyene antibiotic with MW 275; tR = 4.65 min, identified pseudomolecular ion [M+H]+ = 276.1219 and after the loss of hydroxyl group as a water molecule 258.1144), with antifungal effects [82] (Table S3). We were not able to detect any compound in other paired co-cultures (BCCO 10_2259 + BCCO 10_2309; BCCO 10_2259 + BCCO 10_1331; BCCO 10_2295 + BCCO 10_2309; BCCO 10_2295 + BCCO 10_1331).

2.8. Inhibitory Activity of Streptomyces against Candida albicans or Filamentous Fungi

C. albicans and filamentous micromycetes were cultured with streptomycetes to verify that the hemolytic streptomycetes also possess antifungal properties. The strains of Streptomyces were inoculated in lines and cultivated for 72 h. Then, lines of C. albicans were added perpendicularly to those of Streptomyces and the culture was continued for an additional 24 h. Pictures of the results were taken and the inhibitory zone of C. albicans was measured (Table 3).
Subsequently, strains of Streptomyces were cultured with Aspergillus niger, Aspergillus fumigatus, Fusarium spp., and Paecilomyces spp. to test for the antifungal activity of the produced SMs. The results of this experiment are summarized in Table 4. Based on these results, we may briefly conclude that streptomycetes producing polyene B are, to some extent, able to inhibit the growth of Paecilomyces spp. and Aspergillus niger, whereas Aspergillus fumigatus was mostly resistant. Fusarium spp. did not grow in the proximity of some polyene B-producing species (BCCO 10_1092, BCCO 10_1093, BCCO 10_1094), whereas it grew in the proximity of others (BCCO 10_1095, BCCO 10_1097, BCCO 10_1104, BCCO 10_2282, and BCCO 10_2389). The polyene A-producing BCCO 10_1106 inhibited only the growth of Paecilomyces spp., as was also the case of BCCO 10_2259, the strain which probably produces actiphenol. Strain BCCO 10_2309, the probable producer of polyene C, inhibited the growth of Paecilomyces spp. and Aspergillus niger but not that of Fusarium spp. and Aspergillus fumigatus. The candicidin-producing strain BCCO 10_1099 inhibited the growth of Aspergillus niger, Fusarium spp., and Paecilomyces spp., but it had no impact on that of Aspergillus fumigatus.

3. Discussion

Hemolytic capabilities might be beneficial to lyse eukaryotic cells in any streptomycete habitat, either to kill competitors or to obtain nutrients from their lysed cells. They might secondarily protect the streptomycete plant or animal symbionts against mycotic infections.
The SMs produced by streptomycetes traditionally receive considerable attention because of their promising and often successful clinical application [98]. Their hemolytic activities (or, in a broader sense, cytolytic activity, i.e., their ability to lyse eukaryotic cells) have, however, largely been overlooked, as the organisms are generally considered non-pathogenic. Nevertheless, hemolysis remains a natural part of the secondary metabolism of a substantial number of streptomycetes [90]. Recent studies have suggested that hemolysis may serve as a virulence factor [99,100], with filipin produced by Streptomyces sp. TR1341 as an example [89]. Cytolytic SMs may also provide the producer with the ability to acquire larger variability of substrates. Next, we cannot exclude the role of some hemolytic metabolites in the streptomycete programmed cell death, as has also been reported for other cytotoxins acting on DNA [101,102]; though the effect of polyenes in sterol-free bacterial membranes is much less severe [103].
To gain a better understanding of the role hemolysis might play in the soil ecosystem, we used streptomycete strains isolated from various environments for this study—12 isolates directly from the soil, 10 isolates from the bodies of invertebrates, such as ambrosia beetles (Hadrodemius globus, Dinoplatypus pallidus, Xyleborus perforans, Diapus pussilimus, Crossotarsus mniszechi, Hypoborus ficus) and millipedes (Archispirostreptus gigas, Telodeinopus aoutii), and 1 strain from plant roots (Zea mays). Ambrosia beetles live in symbiosis with ambrosia fungi to cover their nutritional needs and to create so-called fungal gardens. As such, they have developed a defensive symbiosis with actinomycetes, especially Streptomyces to protect themselves against fungi [104]. As the diversity of ambrosia beetles is huge (more than 3200 species), and ambrosia fungi are host-specific [105], it can be assumed that many different actinobacteria and their as yet unexplored secondary metabolites may be involved in defensive symbiosis in a particular species.
Our results indicate that a number of streptomycetes that live symbiotically or parasitically with invertebrates and plants produce polyenes as well as those living freely in soil. Interestingly, both beetle- and millipede-derived strains produce similar compounds, although the expected ecological functions differ: the defensive symbiosis of streptomycetes with ambrosia beetles is a well-known phenomenon [104]. On the other hand, there are no data about the symbiotic relationship of intestinal streptomycetes in millipedes. Their interaction may start incidentally, as a transient colonization. However, the fact that the same antifungal compounds produced in the beetles are produced in the millipedes may suggest a much tighter relationship, linked with the prevention of fungal pathogens growth in the host body.
To demonstrate a particular clash between streptomycetes and fungi, we performed a co-culture experiment using a yeast C. albicans (Table 3), and four different ascomycetes: Aspergillus niger, Aspergillus fumigatus, Fusarium spp., and Paecilomyces spp. (Table 4). When both organisms were inoculated at the same time, no inhibitory zones were observed, probably due to the time required for the onset of SM synthesis during streptomycete development. When Streptomyces spp. had a 24-h or 72-h advantage, various zones inhibiting the growth of fungi were observed after an additional 24 h or 72 h, a condition that may reflect a natural event when a streptomycete inhabits a symbiotic organism and protects it from fungal pathogens [106,107,108,109,110]. Thus, cultures of Streptomyces spp. with C. albicans revealed inhibitory zones of the yeast that vary between 3 and 12 mm among various, mostly tetrafungin/tetrin A/polyene B-producing streptomycete species. Aspergillus fumigatus looked highly resistant to the cytolytic metabolites produced by our set of streptomycetes. Aspergillus niger, on the other hand, could not grow in the vicinity of most of the streptomycetes. Fusarium spp. was sensitive only to several tetrafungin/tetrin A/polyene B-producing strains, whereas the sole polyene B-producers did not inhibit its growth. Nevertheless, all hemolytic streptomycetes, regardless of their compound production, were more or less able to inhibit the growth of Paecilomyces spp. which seemed to be most sensitive of the micromycetes tested.
Our LC-MS data combined with the UV-VIS absorption possibly revealed production of six previously characterized cyclic polyenes: candicidins A1, A2, filipin III, pentamycin (fungichromin), strevertene A, tetrafungin, and tetrin A [24,26,33,34,42,51], and four novel polyenes (A-D) in our set of Streptomyces species. Only four strains of the 23 assayed did not show any detectable putative polyenes. This may or may not necessarily mean that another non-polyenic cytotoxic SM is produced. In case of an undetected polyene production, the compounds can be unstable or they are not produced in the amount needed for the isolation method used in this study. Besides this, our results suggest that the production of polyenes is widespread among streptomycetes and often associated with their β-hemolytic and/or antifungal activities.
Tetraenes produced by seven strains belonged to two structural types: small tetrins with MW < 700 [26] and large nystatin and amphotericin B ressembling tetrafungin of MW > 1000 [24]. Unlike tetrins, the structure of tetrafungin in unknown, though the compound was discovered almost 40 years ago. Antifungal activities of the tetraenes have already been reported; however, their hemolytic capabilities were not explored.
Pentaenes were produced by the TR1341 control strain (filipin III and pentamycin). Filipin III was also found in the BCCO 10_2155 strain; however, the associated pentamycin was missing there. The hemolytic properties of the filipin-type compounds, produced by multiple streptomycete species, have long been recognized [111], as well as their antifungal activity. They exhibit equal affinity to ergosterol (the main fungal sterol) and to mammalian cholesterol, which complicates their wider medical use. The sterol-binding feature leads to perforations of the erythrocyte membranes explaining their hemolytic properties [90]. Activity of the filipin gene cluster is essential for β-hemolysis of the human-associated Streptomyces sp. TR1341 [89]. Filipin production does not seem to be linked to any particular phylogenetic clade. On the other hand, strevertene A, a similar pentaene lacking the methylated side chain, was found to be produced by two soil strains belonging to the same phylogenetic clade of S. avidinii NBRC13426, only (Figure 1). Another producer of strevertenes, the tomato-associated S. psammoticus, uses the compound to protect the host plants against Fusarium wilt [112]. Of the novel compounds, polyene B shared by 10 strains, shows the typical methylpentaene-specific absorption spectrum as filipin, but has substantially higher molecular mass. The compound is typically found in strains putatively producing tetraenes (Table S2). It seems unusual for a single strain to produce macrolides differing in the ring lengths as this is determined by the type I polyketide synthase (PKS-I) structure. Of course, we cannot exclude the presence of two different biosynthetic gene clusters in the producer strains, or PKS-I flexibility in the starter unit selection/the chain length control, as possible explanations. Next, a possibility of non-enzymatic degradation (e.g., oxidation) of the original, enzymatically synthesized compounds have to be taken in account as documented in nystatin [113]. However, a more detailed genomic analysis is needed to better understand the biosynthesis of these molecules.
Two novel compounds seem to belong to methylhexaenes—if we assume that a methyl group adjacent to the polyene chain shifts the absorption spectrum peaks to slightly higher wavelengths, as documented for methylpentaenes [42]. Polyene D was produced in a single strain pure culture (BCCO 10_2179), whereas polyene C in the culture of BCCO 10_1331 and BCCO 10_2309 strains. Hexaenes are the smallest, most under characterized polyene group, with just one well-described member: linearmycin [67]. The production of a hexaene derivative of nystatin as a result of a spontaneous mutation of the relevant PKS genes in S. noursei has also been reported [114]. All other hexaenic compounds have been detected based solely on their characteristic UV-VIS absorption and their antifungal activity (Table 1) and the relevant reports lack detailed structure and activity data. Therefore, the two potential novel hexaenes represent perfect targets for future purification as well as structure and activity assessments.
Heptaenes were represented by candicidins detected in two strains coming from different phylogenetic clades (Figure 1). Candicidins have been first identified in S. griseus IMRU3570 [115]. The candicidin complex consists of up to nine compounds (A-I) where only candicidin D has been fully structurally characterized [116]. Candicidins change permeability in the cell membrane of C. albicans [117], followed by the release of K+ ions from the intracellular space, which completes the cell lysis [118].
The mass spectrum of the polyene A peak (BCCO 10_1106) was indeterminate and we were therefore not able to determine its putative molecular formula. Its absorption spectrum resembles that of pentaenes, but the maxima are slightly lower. This suggests that the compound’s polyketide backbone might be further modified in an atypical way.
Streptomycetes undergo a complex life cycle that requires highly coordinated gene expression responsive to environmental changes [119,120,121]. Various small-molecular-weight signaling molecules participate in gene expression control. In this manner, GBLs stimulate metabolic production and act as natural auto-regulators [122]. They mediate intra- and/or interspecies communication via the so-called quorum sensing system. They also help them react effectively to competitive organisms as they may accelerate the transition from the exponential growth phase to sporulation, which includes the coordinated production of bioactive compounds [123]. Here we showed a positive effect of GBL on the production of hemolytic metabolites in several strains (Figure S2).
Co-cultures have been used successfully between various streptomycete species to activate secondary metabolism [124]. Likewise, complex hemolytic properties can be changed (increased or decreased) as a result of interaction among different species in one habitat [125]. In this work we applied the co-culture technique to those strains for which we had not been able to detect any hemolytic compound in a standard one-strain cultivation. Despite probable considerable losses of hemolytic compounds from samples when using the SPE “miniprep” technique, we were able to detect new compounds in the paired co-cultures of [BCCO 10_1331+BCCO 10_2309] and [BCCO 10_2259+BCCO 10_2295] strains. The first pair revealed the novel polyene C compound (mentioned above). The second pair revealed a known non-polyenic hemolytic compound actiphenol, a phenol metabolite with antifungal effects [126].

4. Materials and Methods

4.1. Strains

In total, 23 β-hemolytic isolates (besides one Nocardioides spp. they were exclusively strains of Streptomyces) were obtained from the Collection of Actinomycetes (Biology Centre Collection of Organisms, BCCO, České Budějovice, the Czech Republic, www.actinomycetes.bcco.cz, 9 November 2022) to compare the hemolytic activities of streptomycete isolates originating from various niches in different regions around the world (the strains are listed in Table S2). Of these, 12 originated from soil, 10 were arthropod-associated, and 1 was plant-associated. One additional strain is the human-associated Streptomyces spp. TR1341, a filipin producer that has already been analyzed [89]. This strain was used here as a positive control for the LC-MS analyses.

4.2. 16S rRNA-encoding Gene-Based Phylogeny

We constructed a phylogenetic tree in which we indicated the origin of the strain, the source of isolation, and the type of substance produced to determine the relatedness of the strains in this study and to compare the studied strains with known polyene antibiotic producers. The strains belonging to the BCCO collection are characterized using a combination of morphological features and basic molecular identification. The morphological characterization was performed according to the protocols of the International Streptomyces Project [127]. The molecular identification was performed using 16S rRNA gene sequencing and an identity cut-off of 98.7% for classification as a known species [128]. Data are provided in the catalogue of the BCCO web pages (see above, Section 4.1) under the respective strain number. Overall, 113 sequences were obtained and used for the construction of the tree as described elsewhere [129]; 32 sequences belong to strains in the BCCO collection and 24 sequences belong to known polyene producers that are listed in Table S1. The remaining ones are reference strains used for phylogenetic placement. Of the 24 sequences of known polyene producers, 15 were downloaded from the EzTaxon (access on 4 April 2022) [130]. The 57 sequences used for phylogenetic placement were obtained from the NCBI database (access 04/04/2022) [131]. The sequences were aligned using Muscle (v3.8.425) [132], and the alignment was manually checked and edited to 1522 final, informative columns. The alignment was then used to produce a 1000-replicated bootstrap consensus maximum likelihood tree using RAxML (v8.2.11) [133]. The 16S rRNA gene sequence of the Amycolatopsis alba DSM44262 was used as outgroup. The tree was edited for publication using iTOL [134].

4.3. Culture Media and Cultivation Conditions

For liquid cultures, Streptomyces strains were cultivated in 5 mL of GYM medium [135] at 28 °C for 96 h if not stated otherwise. If required, the medium was supplemented with defibrinated rabbit’s blood (7%). Paired co-cultures, i.e., two Streptomyces strains inoculated equally into one medium, were performed for those strains in which hemolytic SMs had not been identified from previous one-strain cultures. For hemolytic assays, Columbia sheep blood agar (OXOID CZ s.r.o., Thermo Fisher Scientific, Brno, Czechia) was used.

4.4. Ethyl Acetate Extraction and Hemolytic Activity Testing

After centrifugation at 10,000× g at 4 °C for 10 min, NaCl was added to the supernatant, up to a concentration of 5 M. Subsequently, 1 mL of ethyl acetate was added to each vial and incubated on a shaker at 250 rpm and 4 °C for 30 min. Samples were centrifuged at 5000× g for 10 min, and the upper layer of ethyl acetate was transferred to a rotary vacuum evaporator until the sample was completely dry, followed by the addition of 10 μL of chloroform to each sample. Subsequently, 5 μL of each sample was dropped on a blood agar plate and incubated for 3 days with 5 μL of pure chloroform in the middle as a negative control.

4.5. Solid-Phase Extraction

The solid-phase extraction (SPE) procedure was performed as described elsewhere [136]. Briefly, each strain’s supernatant was isolated by centrifugation at 10,000× g at 4 °C for 10 min, and the pH was adjusted to 3–4 using formic acid (Merck, Darmstadt, Germany). An Oasis HLB 3cc 60 mg cartridge (hydrophilic-lipophilic balanced sorbent, Waters, Milford, MA, USA) was conditioned with 3 mL methanol, equilibrated with 3 mL Milli-Q water (Sigma-Aldrich Co. St. Louis, MO, USA), and subsequently, 3 mL of culture supernatant was loaded. The cartridge was then washed with 3 mL of water, and the absorbed substances were eluted with 1.5 mL of methanol.

4.6. LC-MS Analysis

The eluent of each strain was evaporated to dryness (Concentrator plus/Vacufuge® plus, Eppendorf AG, Hamburg, Germany), and 150 μL of 50% methanol was added to each vial. The vials were centrifuged for 5 min at 5000 rpm (Centrifuge MiniSpin, Rotor F-45-12-11, Eppendorf AG, Hamburg, Germany) and 50 µL of each sample was then loaded onto the LC-MS.
The analyses were performed on the Acquity UPLC system with a 2996 PDA detection system (194–600 nm), connected to an LCT premier XE time-of-flight mass spectrometer (Waters, Milford, MA, USA). A 5 μL aliquot of each sample was loaded onto the Acquity UPLC BEH C18 LC column (50 mm × 2.1 mm I.D., particle size 1.7 μm, Waters, Milford, MA, USA), kept at 40 °C, and eluted with a two-component mobile phase, A and B, consisting of 0.1% formic acid and acetonitrile, respectively, at a flow rate of 0.4 mL min−1. The analyses were performed under a linear gradient program (min/%B) 0/5; 1.5/5; 15/70; 18/99 followed by a 1.0-min column clean-up (99% B) and 1.5-min equilibration (5% B). The mass spectrometer operated in the positive “W” mode with capillary voltage set at +2800 V, cone voltage +40 V, dissolving gas temperature: 350 °C, ion source block temperature, 120 °C, cone gas flow 50 L h−1, dissolving gas flow 800 L h−1, scan time of 0.15 s, and an inter-scan delay of 0.01 s. The mass accuracy was kept below 6 ppm using the lock spray technology with leucine enkephalin as the reference compound (2 ng μL−1, 5 μL min−1). The MS chromatograms were extracted for [M+H]+ ions with a tolerance window of 0.05 Da, smoothed with the mean smoothing method (window size; four scans, number of smooths, two). The data were processed by MassLynx V4.1 (Waters, Milford, MA, USA). The original method is described elsewhere [137].

4.7. Testing for the Presence of Extracellular Hemolytic Proteins

To test for the presence of extracellular hemolytic proteins [138], the supernatant collected (cultured 72 h, 28 °C, 200 rpm, centrifuged at 4000× g, 10 min) was filtered (pore size 5 microns) and divided into thirds. The first third of the supernatant was incubated with proteinase K (Carl Roth, Karlsruhe, Germany) to a final concentration of 1 μg/mL at 37 °C for 60 min. The second third of the supernatant was incubated under the same conditions but without the enzyme (this sample served as a positive control as it was expected to maintain its hemolytic activity). The third part was incubated at 100 °C for 5 min (a negative control as we expected a loss of any hemolytic activity). We applied 5 μL of each sample to a paper disc placed on the blood agar. The samples were cultured overnight at 28 °C.

4.8. Antifungal Activity of Streptomyces

Selected Streptomyces strains were inoculated into a 1cm2 square shape in the middle of a blood agar Petri dish and cultivated at 28° C. After 0, 24 h, and 72 h, clinical isolates of Aspergillus niger, Aspergillus fumigatus, Fusarium spp., and Paecilomyces spp. were added in lines leading to the Streptomyces square and co-cultured at the same conditions for another 72 h. The growth inhibitory zones of the fungi were then measured.

5. Conclusions

An extensive literature review on polyenes and non-polyene hemolytic compounds produced by streptomycetes is presented. Among the secondary metabolites produced by a set of 23 β-hemolytic Streptomyces strains, known—candicidins, filipins, strevertene A, tetrafungin, and tetrin A—and novel—polyene AD—polyenic compounds were found. The new compound producers were incorporated into a streptomycete phylogenetic tree among known polyene producers. No clear relation between phylogeny and production of specific polyene types could be revealed. The obtained results suggested that SMs are responsible for hemolytic activities in Streptomyces spp. Their production was in some cases inducible by GBL or by co-cultivation with other Streptomyces strains. Our data suggest that streptomycetes may still serve as a promising source of novel SMs with antifungal and/or hemolytic activities.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232315045/s1.

Author Contributions

The manuscript is based on E.F., N.B. and T.W.’s bachelor theses; supervision, writing and editing, experimental design, and co-culture experiments, J.B.; investigation: cultures, sample extraction, hemolysis test experiments, E.F. and N.B.; LC-MS and data evaluation, M.Č. and M.P.; strain collection management, β-hemolytic Streptomyces strain selection, supervision, pilot experiments, inhibitory activity screening, isolation of strains, and selection of strains for final dataset, A.C., V.K., M.M. and T.W.; identification of strains, 16S rRNA gene phylogeny analysis, literature review, A.C.L. and A.C.; conceptualization and methodology, literature review, interpretation of the results, and project supervision, K.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Czech Health Research Council project No. 17-30091A (K.P., A.C., A.C.L., and T.W.), the Czech Science Fund project No. 17-22572S (A.C.), and the Research Programme of the Academy of Sciences of the Czech Republic Strategy AV21 “Diversity of life and health of ecosystems” and “Land conservation and restoration” to A.C., A.C.L., V.K., M.M., and T.W. as well as projects of Charles University (SVV 260520 and Cooperatio 207032 Immunity and Infection) to J.B., K.P., M.Č. Project No.: UJEP-SGS-2020-53-002-2 was supported by grant within student grant competition at UJEP, Jan Evangelista Purkyně University in Ústí nad Labem (J.B.). M.M. was supported by the Erasmus Traineeship Programme (Erasmus Student Mobility Program, contract ID: 18/1/KA103/047073/SMP).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Miroslav Kolařík (Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague) for providing cultures isolated from ambrosia beetles in Papua-New Guinea to the BCCO Collection of Actinomycetes. Anna Koubová and Terézia Horváthová (Biology Centre of the Academy of Sciences of the Czech Republic, České Budějovice) are thanked for strain isolations from millipedes. Martina Petrlíková and Nikola Fuková are thanked for laboratory analyses.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Procopio, R.E.; Silva, I.R.; Martins, M.K.; Azevedo, J.L.; Araujo, J.M. Antibiotics produced by streptomyces. Braz. J. Infect. Dis. 2012, 16, 466–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Gross, H. Strategies to unravel the function of orphan biosynthesis pathways: Recent examples and future prospects. Appl. Microbiol. Biotechnol. 2007, 75, 267–277. [Google Scholar] [CrossRef] [PubMed]
  3. Wanner, L.A. A new strain of streptomyces causing common scab in potato. Plant. Dis. 2007, 91, 352–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. el Hassan, A.M.; Fahal, A.H.; Ahmed, A.O.; Ismail, A.; Veress, B. The immunopathology of actinomycetoma lesions caused by streptomyces somaliensis. Trans. R. Soc. Trop. Med. Hyg. 2001, 95, 89–92. [Google Scholar] [CrossRef]
  5. Seipke, R.F.; Kaltenpoth, M.; Hutchings, M.I. Streptomyces as symbionts: An emerging and widespread theme? FEMS Microbiol. Rev. 2012, 36, 862–876. [Google Scholar] [CrossRef] [Green Version]
  6. Zhang, X.H.; Austin, B. Haemolysins in vibrio species. J. Appl. Microbiol. 2005, 98, 1011–1019. [Google Scholar] [CrossRef]
  7. Vesper, S.J.; Vesper, M.J. Possible role of fungal hemolysins in sick building syndrome. Adv. Appl. Microbiol. 2004, 55, 191–213. [Google Scholar]
  8. Bhakdi, S.; Tranum-Jensen, J. Complement lysis: A hole is a hole. Immunol. Today 1991, 12, 318–320; discussion 321. [Google Scholar] [CrossRef]
  9. Dixon, D.M.; Walsh, T.J. Antifungal agents. In Medical. Microbiology; Baron, S., Ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  10. Dinos, G.P. The macrolide antibiotic renaissance. Br. J. Pharmacol. 2017, 174, 2967–2983. [Google Scholar] [CrossRef] [Green Version]
  11. Kristanc, L.; Bozic, B.; Jokhadar, S.Z.; Dolenc, M.S.; Gomiscek, G. The pore-forming action of polyenes: From model membranes to living organisms. Biochim. Biophys. Acta Biomembr. 2019, 1861, 418–430. [Google Scholar] [CrossRef]
  12. Guo, X.; Zhang, J.; Li, X.; Xiao, E.; Lange, J.D.; Rienstra, C.M.; Burke, M.D.; Mitchell, D.A. Sterol sponge mechanism is conserved for glycosylated polyene macrolides. ACS Cent. Sci. 2021, 7, 781–791. [Google Scholar] [CrossRef] [PubMed]
  13. Trejo, W.H.; Bennett, R.E. Streptomyces nodosus sp. N., the amphotericin-producing organism. J. Bacteriol. 1963, 85, 436–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Xiong, Z.Q.; Tu, X.R.; Wei, S.J.; Huang, L.; Li, X.H.; Lu, H.; Tu, G.Q. The mechanism of antifungal action of a new polyene macrolide antibiotic antifungalmycin 702 from streptomyces padanus jau4234 on the rice sheath blight pathogen rhizoctonia solani. PLoS ONE 2013, 8, e73884. [Google Scholar] [CrossRef]
  15. Shenin, I.; Solov’ev, V.N.; Nvinskii, A.A. [structure of the polyenic antibiotic, arenomycin b]. Antibiotiki 1973, 18, 872–876. [Google Scholar]
  16. Yang, J.; Xu, D.; Yu, W.; Hao, R.; Wei, J. Regulation of aureofuscin production by the PAS-LuxR family regulator AurJ3M. Enzym. Microb. Technol. 2020, 137, 109532. [Google Scholar] [CrossRef]
  17. Graessle, O.E.; Phares, H.F.; Robinson, H.J. In vitro studies with a new antimycotic agent: Lucensomycin. Antibiot Chemother 1962, 12, 608–617. [Google Scholar]
  18. Kim, H.J.; Kim, M.K.; Lee, M.J.; Won, H.J.; Choi, S.S.; Kim, E.S. Post-pks tailoring steps of a disaccharide-containing polyene npp in pseudonocardia autotrophica. PLoS ONE 2015, 10, e0123270. [Google Scholar] [CrossRef]
  19. Hazen, E.L.; Brown, R. Fungicidin, an antibiotic produced by a soil actinomycete. Proc. Soc. Exp. Biol. Med. 1951, 76, 93–97. [Google Scholar] [CrossRef] [PubMed]
  20. Struyk, A.P.; Hoette, I.; Drost, G.; Waisvisz, J.M.; Van Eek, T.; Hoogerheide, J.C. Pimaricin, a new antifungal antibiotic. Antibiot Annu. 1957, 5, 878–885. [Google Scholar]
  21. Ulrych, A.; Derrick, P.J.; Adamek, F.; Novak, P.; Lemr, K.; Havlicek, V. Dissociation of nystatin and amphotericin analogues: Characterisation of minor anti-fungal macrolides. Eur. J. Mass. Spectrom. 2010, 16, 73–80. [Google Scholar] [CrossRef]
  22. Hamilton-Miller, J.M. Chemistry and biology of the polyene macrolide antibiotics. Bacteriol. Rev. 1973, 37, 166–196. [Google Scholar] [CrossRef] [PubMed]
  23. Davisson, J.W.; Tanner, F.W., Jr.; Finlay, A.C.; Solomons, I.A. Rimocidin, a new antibiotic. Antibiot Chemother 1951, 1, 289–290. [Google Scholar] [PubMed]
  24. Veiga, M.; Fabregas, J. Tetrafungin, a new polyene macrolide antibiotic. I. Fermentation, isolation, characterization, and biological properties. J. Antibiot. 1983, 36, 770–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Dornberger, K.; Fugner, R.; Bradler, G.; Thrum, H. Tetramycin, a new polyene antibiotic. J. Antibiot. 1971, 24, 172–177. [Google Scholar] [CrossRef]
  26. Pandey, R.C.; German, V.F.; Nishikawa, Y.; Rinehart, K.L., Jr. Polyene antibiotics. Ii. The strucutre of tetrin a. J. Am. Chem. Soc. 1971, 93, 3738–3747. [Google Scholar]
  27. Ryu, G.; Choi, W.C.; Hwang, S.; Yeo, W.H.; Lee, C.S.; Kim, S.K. Tetrin c, a new glycosylated polyene macrolide antibiotic produced by streptomyces sp. Gk9244. J. Nat. Prod. 1999, 62, 917–919. [Google Scholar] [CrossRef]
  28. English, A.R.; Mc, B.T. Pa 150, pa 153, and pa 166: New polyene antifungal antibiotics: Biological studies. Antibiot Annu. 1957, 5, 893–896. [Google Scholar]
  29. Taig, M.M.; Solov’eva, N.K.; Braginskaia, P.S. [characteristics of the culture-producer of aurenin]. Antibiotiki 1969, 14, 873–876. [Google Scholar] [PubMed]
  30. Caltrider, P.; Gottlieb, D. Capacidin: A new member of the polyene antibiotic group. Antibiot Chemother 1961, 10, 702–708. [Google Scholar]
  31. Pandey, R.C.; Narasimhachari, N.; Rinehart, K.L., Jr.; Millington, D.S. Polyene antibiotics. Iv. Structure of chainin. J. Am. Chem. Soc. 1972, 94, 4306–4310. [Google Scholar] [CrossRef]
  32. Trinci, A.; Chalkley, L.J. Comparison of the effects of the polyenes amphotericin b and elizabethin on candida pseudotropicalis. Trans. Br. Mycol. Soc. 1983, 80, 491–495. [Google Scholar] [CrossRef]
  33. Cirillo, V.P.; Harsch, M.; Lampen, J.O. Action of the polyene antibiotics filipin, nystatin and n-acetylcandidin on the yeast cell membrane. J. Gen. Microbiol. 1964, 35, 249–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Umezawa, S.; Tanaka, Y.; Ooka, M.; Shiotsu, S. A new antifungal antibiotic, pentamycin. J. Antibiot. 1958, 11, 26–29. [Google Scholar] [PubMed]
  35. Li, Z.; Rawlings, B.J.; Harrison, P.H.; Vederas, J.C. Production of new polyene antibiotics by streptomyces cellulosae after addition of ethyl (z)-16-phenylhexadec-9-enoate. J. Antibiot. 1989, 42, 577–584. [Google Scholar] [CrossRef] [PubMed]
  36. Endo, M.; Hayashida, T.; Tsunematsu, Y. [application of an antifungal antibiotic (kabicidin) in cultures of cells]. Jpn. J. Med. Sci. Biol. 1959, 12, 173–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Brazhnikova, M.G.; Kudinova, M.K.; Lavrova, M.F.; Borisova, V.N.; Krugliak, E.B. [isolation, purification and physico-chemical properties of antibiotic lienomycin]. Antibiotiki 1971, 16, 483–486. [Google Scholar] [PubMed]
  38. Bycroft, B.W.; Payne, D.J. Dictionary of Antibiotics and Related Substances: With CD-ROM, 2nd ed.; CRC Press: Boca Raton, FL, USA, 2014. [Google Scholar]
  39. Berdy, J.; Horváth, I. Antibiotics produced by streptomyces vi. Pentafungin, a new antifungal antibiotic. J. Basic Microbiol. 2007, 5, 345–350. [Google Scholar]
  40. Yao, T.; Liu, Z.; Li, T.; Zhang, H.; Liu, J.; Li, H.; Che, Q.; Zhu, T.; Li, D.; Li, W. Characterization of the biosynthetic gene cluster of the polyene macrolide antibiotic reedsmycins from a marine-derived streptomyces strain. Microb. Cell Fact 2018, 17, 98. [Google Scholar] [CrossRef]
  41. Van Arnam, E.B.; Ruzzini, A.C.; Sit, C.S.; Horn, H.; Pinto-Tomas, A.A.; Currie, C.R.; Clardy, J. Selvamicin, an atypical antifungal polyene from two alternative genomic contexts. Proc. Natl. Acad. Sci. USA 2016, 113, 12940–12945. [Google Scholar] [CrossRef] [Green Version]
  42. Guo, J.; Schlingmann, G.; Carter, G.T.; Nakanishi, K.; Berova, N. Absolute stereochemistry of the strevertenes. Chirality 2000, 12, 43–51. [Google Scholar] [CrossRef]
  43. Kim, Y.P.; Tomoda, H.; Iizima, K.; Fukuda, T.; Matsumoto, A.; Takahashi, Y.; Omura, S. Takanawaenes, novel antifungal antibiotics produced by streptomyces sp. K99-5278. I. Taxonomy, fermentation, isolation and biological properties. J. Antibiot. 2003, 56, 448–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Greule, A.; Intra, B.; Flemming, S.; Rommel, M.G.; Panbangred, W.; Bechthold, A. The draft genome sequence of actinokineospora bangkokensis 44ehw(t) reveals the biosynthetic pathway of the antifungal thailandin compounds with unusual butylmalonyl-coa extender units. Molecules 2016, 21, 1607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Martin, J.F.; McDaniel, L.E. Isolation, purification and properties of the hexaene macrolides candihexin i and candihexin ii. J. Antibiot. 1974, 27, 610–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sakamoto, J.M. [study on antifungal antibiotics. Ii. Cryptocidin, a new antibiotic produced by streptomycetes]. J. Antibiot. 1959, 12, 21–23. [Google Scholar]
  47. Barashkova, N.P.; Shenin Iu, D.; Omel’chenko, V.N.; Konev Iu, E. [actinomyces chromogenes var. Graecus var. Nov., a producer of a new hexaene antibiotic]. Antibiotiki 1977, 22, 832–837. [Google Scholar]
  48. Chakrabarti, S.; Chandra, A.L. A new streptomycete and a new polyene antibiotic, acmycin. Folia Microbiol. (Praha) 1982, 27, 167–172. [Google Scholar] [CrossRef]
  49. Dutcher, J.D. The discovery and development of amphotericin b. Dis. Chest 1968, 54 (Suppl. S1), 296–298. [Google Scholar] [CrossRef]
  50. Kotler-Brajtburg, J.; Medoff, G.; Kobayashi, G.S.; Boggs, S.; Schlessinger, D.; Pandey, R.C.; Rinehart, K.L., Jr. Classification of polyene antibiotics according to chemical structure and biological effects. Antimicrob. Agents Chemother. 1979, 15, 716–722. [Google Scholar] [CrossRef] [Green Version]
  51. Kligman, A.M.; Lewis, F.S. In vitro and in vivo activity of candicidin on pathogenic fungi. Proc. Soc. Exp. Biol. Med. 1953, 82, 399–404. [Google Scholar] [CrossRef]
  52. Taber, W.A.; Vining, L.C.; Waksman, S.A. Candidin, a new antifungal antibiotic produced by streptomyces viridoflavus. Antibiot Chemother 1954, 4, 455–461. [Google Scholar]
  53. Kruglikova, L.F.; Shenin Iu, D. [characteristics of a new aromatic heptaenic antibiotic, flavumycin a]. Antibiotiki 1976, 21, 407–411. [Google Scholar] [PubMed]
  54. Mohan, R.R.; Pianotti, R.S.; Martin, J.F.; Ringel, S.M.; Schwartz, B.S.; Bailey, E.G.; McDaniel, L.E.; Schaffner, C.P. Fungimycin production, isolation, biosynthesis, and in vitro antifungal activity. Antimicrob. Agents Chemother. 1963, 161, 462–470. [Google Scholar] [PubMed]
  55. Gokhale, B.B. Hamycin--A new antifungal antibiotic. Arch. Dermatol. 1963, 88, 558–560. [Google Scholar] [CrossRef] [PubMed]
  56. Filippova, A.I.; Solov’ev, S.N.; Vasil’eva, N.G.; Dumova, A.M.; Egorenkova, A.N. [isomerization of polyene antibiotics with characteristics of isolevorin a2]. Antibiotiki 1972, 17, 932–937. [Google Scholar]
  57. Filippova, A.I.; Lashkov, G.I.; Kozel, S.P.; Shenin Iu, D. [spectrophotometric research on the photochemical isomerization of the polyene macrolide antibiotic levorin a2]. Antibiot Med. Biotekhnol. 1987, 32, 749–754. [Google Scholar]
  58. Borowski, E.; Malyshkina, M.; Soloviev, S.; Ziminski, T. Isolation and characterization of levorin a and b, the heptaene macrolide antifungal antibiotics of “aromatic” subgroup. Chemotherapy 1965, 10, 176–194. [Google Scholar] [CrossRef]
  59. Martinez-Quesada, J.; Torres-Rodriguez, J.M.; Roses-Codinachs, M.; Amaral-Olivera, M. Minimal inhibitory concentrations of lucknomycin, a new polyenic derivative, for candida and aspergillus spp. Antimicrob. Agents Chemother. 1983, 24, 132–133. [Google Scholar] [CrossRef] [Green Version]
  60. Luna, T.; Mazzolla, R.; Romano, G.; Blasi, E. Potent antifungal effects of a new derivative of partricin a in a murine model of cerebral cryptococcosis. Antimicrob. Agents Chemother. 1997, 41, 706–708. [Google Scholar] [CrossRef] [Green Version]
  61. Pawlak, J.; Sowinski, P.; Borowski, E.; Gariboldi, P. Stereostructure of perimycin a. J. Antibiot. 1995, 48, 1034–1038. [Google Scholar] [CrossRef] [Green Version]
  62. Mendizabal, A.F.; Inza, R.; Salaber, J.A. [trichomycin: A new antibiotic for the treatment of colpitis caused by trichomonas & candida albicans]. Rev. Asoc. Med. Argent. 1957, 71, 48–50. [Google Scholar]
  63. Bortolo, R.; Spera, S.; Gugliemetti, G.; Cassani, G. Ab023, novel polyene antibiotics. Ii. Isolation and structure determination. J. Antibiot. 1993, 46, 255–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cai, P.; Kong, F.; Fink, P.; Ruppen, M.E.; Williamson, R.T.; Keiko, T. Polyene antibiotics from streptomyces mediocidicus. J. Nat. Prod. 2007, 70, 215–219. [Google Scholar] [CrossRef] [PubMed]
  65. McAlpine, J.B.; Bachmann, B.O.; Piraee, M.; Tremblay, S.; Alarco, A.M.; Zazopoulos, E.; Farnet, C.M. Microbial genomics as a guide to drug discovery and structural elucidation: Eco-02301, a novel antifungal agent, as an example. J. Nat. Prod. 2005, 68, 493–496. [Google Scholar] [CrossRef] [PubMed]
  66. Irschik, H.; Schummer, D.; Hofle, G.; Reichenbach, H.; Steinmetz, H.; Jansen, R. Etnangien, a macrolide-polyene antibiotic from sorangium cellulosum that inhibits nucleic acid polymerases. J. Nat. Prod. 2007, 70, 1060–1063. [Google Scholar] [CrossRef]
  67. Park, H.S.; Nah, H.J.; Kang, S.H.; Choi, S.S.; Kim, E.S. Screening and isolation of a novel polyene-producing streptomyces strain inhibiting phytopathogenic fungi in the soil environment. Front. Bioeng. Biotechnol. 2021, 9, 692340. [Google Scholar] [CrossRef] [PubMed]
  68. Almeida, E.L.; Kaur, N.; Jennings, L.K.; Carrillo Rincon, A.F.; Jackson, S.A.; Thomas, O.P.; Dobson, A.D.W. Genome mining coupled with osmac-based cultivation reveal differential production of surugamide a by the marine sponge isolate streptomyces sp. Sm17 when compared to its terrestrial relative s. Albidoflavus j1074. Microorganisms 2019, 7, 394. [Google Scholar] [CrossRef] [Green Version]
  69. Chater, K.F.; Biro, S.; Lee, K.J.; Palmer, T.; Schrempf, H. The complex extracellular biology of streptomyces. FEMS Microbiol. Rev. 2010, 34, 171–198. [Google Scholar] [CrossRef] [Green Version]
  70. Matsuda, K.; Kuranaga, T.; Sano, A.; Ninomiya, A.; Takada, K.; Wakimoto, T. The revised structure of the cyclic octapeptide surugamide a. Chem. Pharm. Bull. 2019, 67, 476–480. [Google Scholar] [CrossRef] [Green Version]
  71. Umezawa, K.; Nakazawa, K.; Ikeda, Y.; Naganawa, H.; Kondo, S. Polyoxypeptins a and b produced by streptomyces: Apoptosis-inducing cyclic depsipeptides containing the novel amino acid (2s,3r)-3-hydroxy-3-methylproline. J. Org. Chem. 1999, 64, 3034–3038. [Google Scholar] [CrossRef]
  72. Umezawa, H. Bleomycin and other antitumor antibiotics of high molecular weight. Antimicrob. Agents Chemother. (Bethesda) 1965, 5, 1079–1085. [Google Scholar]
  73. Johnson, A.W. The chemistry of actinomycin d and related compounds. Ann. N. Y. Acad. Sci. 1960, 89, 336–341. [Google Scholar] [CrossRef] [PubMed]
  74. Giessen, T.W.; Franke, K.B.; Knappe, T.A.; Kraas, F.I.; Bosello, M.; Xie, X.; Linne, U.; Marahiel, M.A. Isolation, structure elucidation, and biosynthesis of an unusual hydroxamic acid ester-containing siderophore from actinosynnema mirum. J. Nat. Prod. 2012, 75, 905–914. [Google Scholar] [CrossRef] [PubMed]
  75. Kido, G.S.; Spyhalski, E. Antimycin a, an antibiotic with insecticidal and miticidal properties. Science 1950, 112, 172–173. [Google Scholar] [CrossRef] [PubMed]
  76. Hatanaka, H.; Iwami, M.; Kino, T.; Goto, T.; Okuhara, M. Fr-900520 and fr-900523, novel immunosuppressants isolated from a streptomyces. I. Taxonomy of the producing strain. J. Antibiot. (Tokyo) 1988, 41, 1586–1591. [Google Scholar] [CrossRef] [PubMed]
  77. Andexer, J.N.; Kendrew, S.G.; Nur-e-Alam, M.; Lazos, O.; Foster, T.A.; Zimmermann, A.S.; Warneck, T.D.; Suthar, D.; Coates, N.J.; Koehn, F.E.; et al. Biosynthesis of the immunosuppressants fk506, fk520, and rapamycin involves a previously undescribed family of enzymes acting on chorismate. Proc. Natl. Acad. Sci. USA 2011, 108, 4776–4781. [Google Scholar] [CrossRef]
  78. He, M.; Haltli, B.; Summers, M.; Feng, X.; Hucul, J. Isolation and characterization of meridamycin biosynthetic gene cluster from streptomyces sp. Nrrl 30748. Gene 2006, 377, 109–118. [Google Scholar] [CrossRef]
  79. Doscher, M.E.; Wood, I.B.; Pankavich, J.A.; Ricks, C.A. Efficacy of nemadectin, a new broad-spectrum endectocide, against natural infections of canine gastrointestinal helminths. Vet. Parasitol. 1989, 34, 255–259. [Google Scholar] [CrossRef]
  80. Seto, B. Rapamycin and mtor: A serendipitous discovery and implications for breast cancer. Clin. Transl. Med. 2012, 1, 29. [Google Scholar] [CrossRef] [Green Version]
  81. Rhodes, A.; Fantes, K.H.; Boothroyd, B.; McGonagle, M.P.; Crosse, R. Venturicidin: A new antifungal antibiotic of potential use in agriculture. Nature 1961, 192, 952–954. [Google Scholar] [CrossRef]
  82. Spizek, J.; Malek, I.; Suchy, J.; Vondracek, M.; Vanek, Z. Metabolites of streptomyces noursei. V. Relation of the production of cycloheximide and actiphenol to the production of fungicidin. Folia Microbiol. 1965, 10, 263–266. [Google Scholar]
  83. Seaton, P.J.; Gould, S.J. New products related to kinamycin from streptomyces murayamaensis. Ii. Structures of pre-kinamycin, keto-anhydrokinamycin, and kinamycins e and f. J. Antibiot. 1989, 42, 189–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhang, J.; Li, S.; Wu, X.; Guo, Z.; Lu, C.; Shen, Y. Nam7 hydroxylase is responsible for the formation of the naphthalenic ring in the biosynthesis of neoansamycins. Org. Lett. 2017, 19, 2442–2445. [Google Scholar] [CrossRef] [PubMed]
  85. Bhuyan, B.K.; Smith, C.G. Differential interaction of nogalamycin with DNA of varying base composition. Proc. Natl. Acad. Sci. USA 1965, 54, 566–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Koshino, H.; Takahashi, H.; Osada, H.; Isono, K. Reveromycins, new inhibitors of eukaryotic cell growth. Iii. Structures of reveromycins a, b, c and d. J. Antibiot. 1992, 45, 1420–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Liu, W.; Nonaka, K.; Nie, L.; Zhang, J.; Christenson, S.D.; Bae, J.; Van Lanen, S.G.; Zazopoulos, E.; Farnet, C.M.; Yang, C.F.; et al. The neocarzinostatin biosynthetic gene cluster from streptomyces carzinostaticus atcc 15944 involving two iterative type i polyketide synthases. Chem. Biol. 2005, 12, 293–302. [Google Scholar] [CrossRef] [Green Version]
  88. BeBoer, C.; Dietz, A. The description and antibiotic production of streptomyces hygroscopicus var. Geldanus. J. Antibiot. 1976, 29, 1182–1188. [Google Scholar]
  89. Herbrik, A.; Corretto, E.; Chronakova, A.; Langhansova, H.; Petraskova, P.; Hrdy, J.; Cihak, M.; Kristufek, V.; Bobek, J.; Petricek, M.; et al. A human lung-associated streptomyces sp. Tr1341 produces various secondary metabolites responsible for virulence, cytotoxicity and modulation of immune response. Front. Microbiol. 2019, 10, 3028. [Google Scholar] [CrossRef]
  90. Knopik-Skrocka, A.; Bielawski, J. The mechanism of the hemolytic activity of polyene antibiotics. Cell Mol. Biol. Lett. 2002, 7, 31–48. [Google Scholar]
  91. te Welscher, Y.M.; Jones, L.; van Leeuwen, M.R.; Dijksterhuis, J.; de Kruijff, B.; Eitzen, G.; Breukink, E. Natamycin inhibits vacuole fusion at the priming phase via a specific interaction with ergosterol. Antimicrob. Agents Chemother. 2010, 54, 2618–2625. [Google Scholar] [CrossRef] [Green Version]
  92. Gray, K.C.; Palacios, D.S.; Dailey, I.; Endo, M.M.; Uno, B.E.; Wilcock, B.C.; Burke, M.D. Amphotericin primarily kills yeast by simply binding ergosterol. Proc. Natl. Acad. Sci. USA 2012, 109, 2234–2239. [Google Scholar] [CrossRef] [Green Version]
  93. Elliott, S.J.; Srinivas, S.; Albert, M.J.; Alam, K.; Robins-Browne, R.M.; Gunzburg, S.T.; Mee, B.J.; Chang, B.J. Characterization of the roles of hemolysin and other toxins in enteropathy caused by alpha-hemolytic escherichia coli linked to human diarrhea. Infect Immun. 1998, 66, 2040–2051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Scherr, N.; Nguyen, L. Mycobacterium versus streptomyces--We are different, we are the same. Curr. Opin. Microbiol. 2009, 12, 699–707. [Google Scholar] [CrossRef] [PubMed]
  95. Wurzer, T. The Interaction of Streptomyces-like Bacteria and Model Microorganisms in Secondary Metabolite Production, Motility and Hemolytic Activities—Experimental. Bachelor’s Thesis, University of South Bohemia in České Budějovice, České Budějovice, Czech Republic, 2019. [Google Scholar]
  96. Labeda, D.P.; Goodfellow, M.; Brown, R.; Ward, A.C.; Lanoot, B.; Vanncanneyt, M.; Swings, J.; Kim, S.B.; Liu, Z.; Chun, J.; et al. Phylogenetic study of the species within the family streptomycetaceae. Antonie Van Leeuwenhoek 2012, 101, 73–104. [Google Scholar] [CrossRef] [PubMed]
  97. Rajesh, T.; Jeon, J.M.; Kim, Y.H.; Kim, H.J.; Yi da, H.; Park, S.H.; Choi, K.Y.; Kim, Y.G.; Kim, J.; Jung, S.; et al. Functional analysis of the gene sco1782 encoding streptomyces hemolysin (s-hemolysin) in streptomyces coelicolor m145. Toxicon 2013, 71, 159–165. [Google Scholar] [CrossRef] [PubMed]
  98. Harir, M.; Bendif, H.; Bellahcene, M.; Pogni, Z.F.R. Streptomyces secondary metabolites. In Basic Biology and Applications of Actinobacteria; Enany, S., Ed.; IntechOpen: London, UK, 2018. [Google Scholar]
  99. Bernal, M.G.; Campa-Cordova, A.I.; Saucedo, P.E.; Gonzalez, M.C.; Marrero, R.M.; Mazon-Suastegui, J.M. Isolation and in vitro selection of actinomycetes strains as potential probiotics for aquaculture. Vet. World 2015, 8, 170–176. [Google Scholar] [CrossRef] [Green Version]
  100. Sheng, Y.; Ou, Y.; Hu, X.; Deng, Z.; Bai, L.; Kang, Q. Generation of tetramycin b derivative with improved pharmacological property based on pathway engineering. Appl. Microbiol. Biotechnol. 2020, 104, 2561–2573. [Google Scholar] [CrossRef]
  101. Cinar, B.; Demir, Z.; Tunca, S. Heterologous expression of 8-demethyl-tetracenomycin (8-dmtc) affected streptomyces coelicolor life cycle. Braz. J. Microbiol. 2021, 52, 1107–1118. [Google Scholar] [CrossRef]
  102. Tenconi, E.; Traxler, M.F.; Hoebreck, C.; van Wezel, G.P.; Rigali, S. Production of prodiginines is part of a programmed cell death process in streptomyces coelicolor. Front. Microbiol. 2018, 9, 1742. [Google Scholar] [CrossRef]
  103. Haro-Reyes, T.; Diaz-Peralta, L.; Galvan-Hernandez, A.; Rodriguez-Lopez, A.; Rodriguez-Fragoso, L.; Ortega-Blake, I. Polyene antibiotics physical chemistry and their effect on lipid membranes; impacting biological processes and medical applications. Membranes 2022, 12, 681. [Google Scholar] [CrossRef]
  104. Grubbs, K.J.; Surup, F.; Biedermann, P.H.W.; McDonald, B.R.; Klassen, J.L.; Carlson, C.M.; Clardy, J.; Currie, C.R. Cycloheximide-producing streptomyces associated with xyleborinus saxesenii and xyleborus affinis fungus-farming ambrosia beetles. Front. Microbiol. 2020, 11, 562140. [Google Scholar] [CrossRef]
  105. Skelton, J.; Jusino, M.A.; Li, Y.; Bateman, C.; Thai, P.H.; Wu, C.; Lindner, D.L.; Hulcr, J. Detecting symbioses in complex communities: The fungal symbionts of bark and ambrosia beetles within asian pines. Microb. Ecol. 2018, 76, 839–850. [Google Scholar] [CrossRef] [PubMed]
  106. Goettler, W.; Kaltenpoth, M.; McDonald, S.; Strohm, E. Comparative morphology of the symbiont cultivation glands in the antennae of female digger wasps of the genus philanthus (hymenoptera: Crabronidae). Front. Physiol. 2022, 13, 815494. [Google Scholar] [CrossRef] [PubMed]
  107. Dhodary, B.; Spiteller, D. Ammonia production by streptomyces symbionts of acromyrmex leaf-cutting ants strongly inhibits the fungal pathogen escovopsis. Microorganisms 2021, 9, 1622. [Google Scholar] [CrossRef]
  108. Vergnes, S.; Gayrard, D.; Veyssière, M.; Toulotte, J.; Martinez, Y.; Dumont, V.; Bouchez, O.; Rey, T.; Dumas, B. Phyllosphere colonization by a soil streptomyces sp. Promotes plant defense responses against fungal infection. Mol. Plant-Microbe Interact. MPMI 2020, 33, 223–234. [Google Scholar] [CrossRef] [PubMed]
  109. Kim, D.R.; Cho, G.; Jeon, C.W.; Weller, D.M.; Thomashow, L.S.; Paulitz, T.C.; Kwak, Y.S. A mutualistic interaction between streptomyces bacteria, strawberry plants and pollinating bees. Nat. Commun. 2019, 10, 4802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Dohet, L.; Grégoire, J.C.; Berasategui, A.; Kaltenpoth, M.; Biedermann, P.H. Bacterial and fungal symbionts of parasitic dendroctonus bark beetles. FEMS Microbiol. Ecol. 2016, 92. [Google Scholar] [CrossRef] [Green Version]
  111. Schlosser, E.; Gottlieb, D. Mode of hemolytic action of the antifungal polyene antibiotic filipin. Z. Naturforsch. B 1966, 21, 74–77. [Google Scholar] [CrossRef]
  112. Kim, J.D.; Han, J.W.; Lee, S.C.; Lee, D.; Hwang, I.C.; Kim, B.S. Disease control effect of strevertenes produced by streptomyces psammoticus against tomato fusarium wilt. J. Agric. Food Chem. 2011, 59, 1893–1899. [Google Scholar] [CrossRef]
  113. Shenin, Y.D.; Belakhov, V.V.; Araviiskii, R.A. Nystatin: Methods of preparation, search for derivatives, and prospects for medicinal use (review). Pharm. Chem. J. 1993, 27, 84–92. [Google Scholar] [CrossRef]
  114. Brautaset, T.; Bruheim, P.; Sletta, H.; Hagen, L.; Ellingsen, T.E.; Strom, A.R.; Valla, S.; Zotchev, S.B. Hexaene derivatives of nystatin produced as a result of an induced rearrangement within the nysc polyketide synthase gene in s. Noursei atcc 11455. Chem. Biol. 2002, 9, 367–373. [Google Scholar] [CrossRef] [Green Version]
  115. Asturias, J.A.; Liras, P.; Martin, J.F. Phosphate control of pabs gene transcription during candicidin biosynthesis. Gene 1990, 93, 79–84. [Google Scholar] [CrossRef] [PubMed]
  116. Szczeblewski, P.; Laskowski, T.; Kubacki, B.; Dziergowska, M.; Liczmanska, M.; Grynda, J.; Kubica, P.; Kot-Wasik, A.; Borowski, E. Analytical studies on ascosin, candicidin and levorin multicomponent antifungal antibiotic complexes. The stereostructure of ascosin a2. Sci. Rep. 2017, 7, 40158. [Google Scholar] [CrossRef] [PubMed]
  117. Brajtburg, J.; Medoff, G.; Kobayashi, G.S.; Elberg, S.; Finegold, C. Permeabilizing and hemolytic action of large and small polyene antibiotics on human erythrocytes. Antimicrob. Agents Chemother. 1980, 18, 586–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Hammond, S.M.; Kliger, B.N. Mode of action of the polyene antibiotic candicidin: Binding factors in the wall of candida albicans. Antimicrob. Agents Chemother. 1976, 9, 561–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Mikulik, K.; Bobek, J.; Zidkova, J.; Felsberg, J. 6s rna modulates growth and antibiotic production in streptomyces coelicolor. Appl. Microbiol. Biotechnol. 2014, 98, 7185–7197. [Google Scholar] [CrossRef]
  120. Bobek, J.; Strakova, E.; Zikova, A.; Vohradsky, J. Changes in activity of metabolic and regulatory pathways during germination of s. Coelicolor. BMC Genom. 2014, 15, 1173. [Google Scholar] [CrossRef] [Green Version]
  121. Mikulik, K.; Suchan, P.; Bobek, J. Changes in ribosome function induced by protein kinase associated with ribosomes of streptomyces collinus producing kirromycin. Biochem. Biophys. Res. Commun. 2001, 289, 434–443. [Google Scholar] [CrossRef]
  122. Horinouchi, S. A microbial hormone, a-factor, as a master switch for morphological differentiation and secondary metabolism in streptomyces griseus. Front. Biosci. 2002, 7, d2045–d2057. [Google Scholar]
  123. Takano, E. Gamma-butyrolactones: Streptomyces signalling molecules regulating antibiotic production and differentiation. Curr. Opin. Microbiol. 2006, 9, 287–294. [Google Scholar] [CrossRef]
  124. Yamanaka, K.; Oikawa, H.; Ogawa, H.O.; Hosono, K.; Shinmachi, F.; Takano, H.; Sakuda, S.; Beppu, T.; Ueda, K. Desferrioxamine e produced by streptomyces griseus stimulates growth and development of streptomyces tanashiensis. Microbiology (Reading) 2005, 151, 2899–2905. [Google Scholar] [CrossRef]
  125. Lee, H.J.; Whang, K.S. Streptomyces rhizosphaerihabitans sp. Nov. And streptomyces adustus sp. Nov., isolated from bamboo forest soil. Int. J. Syst. Evol. Microbiol. 2016, 66, 3573–3578. [Google Scholar] [CrossRef] [PubMed]
  126. Human, Z.R.; Moon, K.; Bae, M.; de Beer, Z.W.; Cha, S.; Wingfield, M.J.; Slippers, B.; Oh, D.C.; Venter, S.N. Antifungal streptomyces spp. Associated with the infructescences of protea spp. In south africa. Front. Microbiol. 2016, 7, 1657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Shirling, E.B.; Gottlieb, D. Methods for characterization of streptomyces species1. Int. J. Syst. Evol. Microbiol. 1966, 16, 313–340. [Google Scholar]
  128. Chun, J.; Oren, A.; Ventosa, A.; Christensen, H.; Arahal, D.R.; da Costa, M.S.; Rooney, A.P.; Yi, H.; Xu, X.-W.; De Meyer, S.; et al. Proposed minimal standards for the use of genome data for the taxonomy of prokaryotes. Int. J. Syst. Evol. Microbiol. 2018, 68, 461–466. [Google Scholar] [CrossRef]
  129. Kotrbová, L.; Lara, A.C.; Corretto, E.; Scharfen, J.; Ulmann, V.; Petříčková, K.; Chroňáková, A. Evaluation and comparison of antibiotic susceptibility profiles of streptomyces spp. From clinical specimens revealed common and region-dependent resistance patterns. Sci. Rep. 2022, 12, 1–12. [Google Scholar] [CrossRef]
  130. Chun, J.; Lee, J.H.; Jung, Y.; Kim, M.; Kim, S.; Kim, B.K.; Lim, Y.W. Eztaxon: A web-based tool for the identification of prokaryotes based on 16s ribosomal rna gene sequences. Int. J. Syst. Evol. Microbiol. 2007, 57, 2259–2261. [Google Scholar] [CrossRef] [PubMed]
  131. Schoch, C.L.; Ciufo, S.; Domrachev, M.; Hotton, C.L.; Kannan, S.; Khovanskaya, R.; Leipe, D.; McVeigh, R.; O’Neill, K.; Robbertse, B.; et al. Ncbi taxonomy: A comprehensive update on curation, resources and tools. Database 2020, 2020, baaa062. [Google Scholar] [CrossRef]
  132. Edgar, R.C. Muscle: A multiple sequence alignment method with reduced time and space complexity. BMC Bioinform. 2004, 5, 113. [Google Scholar] [CrossRef] [Green Version]
  133. Stamatakis, A. Raxml version 8: A tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics 2014, 30, 1312–1313. [Google Scholar] [CrossRef] [Green Version]
  134. Letunic, I.; Bork, P. Interactive tree of life (itol) v5: An online tool for phylogenetic tree display and annotation. Nucleic Acids Res. 2021, 49, W293–W296. [Google Scholar] [CrossRef]
  135. Ochi, K. Metabolic initiation of differentiation and secondary metabolism by streptomyces griseus: Significance of the stringent response (ppgpp) and gtp content in relation to a factor. J. Bacteriol. 1987, 169, 3608–3616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kamenik, Z.; Hadacek, F.; Mareckova, M.; Ulanova, D.; Kopecky, J.; Chobot, V.; Plhackova, K.; Olsovska, J. Ultra-high-performance liquid chromatography fingerprinting method for chemical screening of metabolites in cultivation broth. J. Chromatogr. A 2010, 1217, 8016–8025. [Google Scholar] [CrossRef] [PubMed]
  137. Cihak, M.; Kamenik, Z.; Smidova, K.; Bergman, N.; Benada, O.; Kofronova, O.; Petrickova, K.; Bobek, J. Secondary metabolites produced during the germination of streptomyces coelicolor. Front. Microbiol. 2017, 8, 2495. [Google Scholar] [CrossRef] [Green Version]
  138. Guan, S.; Ji, C.; Zhou, T.; Li, J.; Ma, Q.; Niu, T. Aflatoxin b(1) degradation by stenotrophomonas maltophilia and other microbes selected using coumarin medium. Int. J. Mol. Sci. 2008, 9, 1489–1503. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Phylogeny of Streptomyces strains involved in the study inferred from 16S rRNA gene similarity. Color codes represent the type of a polyene produced by the strain, the isolation source, and the country of origin. Only bootstraps above 50 are shown. The question mark indicates metabolites detected in a co-culture. The sequences of type strains closely related to the sequences of known polyene producers and BCCO strains were included to support topography of phylogenetic tree. The strains were selected according to Labeda et al. [96]. Abbreviations: A.–Amycolatopsis, N.–Nocardioides, S.–Streptomycces.
Figure 1. Phylogeny of Streptomyces strains involved in the study inferred from 16S rRNA gene similarity. Color codes represent the type of a polyene produced by the strain, the isolation source, and the country of origin. Only bootstraps above 50 are shown. The question mark indicates metabolites detected in a co-culture. The sequences of type strains closely related to the sequences of known polyene producers and BCCO strains were included to support topography of phylogenetic tree. The strains were selected according to Labeda et al. [96]. Abbreviations: A.–Amycolatopsis, N.–Nocardioides, S.–Streptomycces.
Ijms 23 15045 g001
Table 1. Actinomycete polyene SMs. AB—antibacterial; AF—antifungal; AP/I—antiparasitic, insecticidal; HL/CL—hemo-/cytolytic; AC—anti-cancer. Asterisks indicate compounds with clinical (*) or agricultural (**) application.
Table 1. Actinomycete polyene SMs. AB—antibacterial; AF—antifungal; AP/I—antiparasitic, insecticidal; HL/CL—hemo-/cytolytic; AC—anti-cancer. Asterisks indicate compounds with clinical (*) or agricultural (**) application.
CompoundFormulaCalculated Average MassActivitiesRef.
CYCLIC—TETRAENESABAFAP/IHL/CLAC
Amphotericin A *C47H75NO17926.1090 + + [13]
Antifungalmycin 702C35H60O14704.8530 + [14]
Arenomycin B (Lucensomycin)C36H55NO13709.8316 + [15]
AureofuscinC28H43NO12585.6490 + [16]
Lucimycin (Lucensomycin, Etruscomycin)C36H53NO13707.8158 + + [17]
NPP A1C55H88N2O221129.3040 + [18]
Nystatin A1 (Fungicidin) *C47H75NO17926.1090-+ [19]
Nystatin A2C47H75NO16910.1096
Nystatin A3C53H85NO201056.2526
Pimaricin (Natamycin) */**C33H47NO13665.7351 + - [20]
Polyfungin BC53H85NO191040.2532 + [21]
ProtocidinC29H45NO13615.6752 [22]
RimocidinC39H61NO14767.9117-+ [23]
TetrafunginC47H82NO231029.1610 + [24]
Tetramycin A
Tetramycin B
C35H53NO13
C35H53NO14
695.8048
711.8042
++ [25]
Tetrin A
Tetrin B
Tetrin C
C34H51NO13
C34H51NO14
C34H49NO13
681.7779
697.7773
679.7620
+ [26]
[27]
Toyamycin (Akitamycin)C41H65NO18859.9630 + [28]
PA-166C35H53NO14711.8042 + [28]
CYCLIC—PENTAENES ABAFAP/IHL/CLAC
Aurenin (1’-Hydroxyisochainin)C33H54O11626.7852 + [29]
CapacidinC54H85NO181050.2716 + [30]
ChaininC33H54O10610.7858 + [31]
ElizabethinC35H58O12670.8383 + [32]
Filipin IC35H58O9622.8401 + + [33]
Filipin IIC35H58O10638.8395
Filipin IIIC35H58O11654.8389
Filipin IVC35H58O11654.8389
Fungichromin (Pentamycin) *C35H58O12670.8383++ + [34]
HomochaininC34H56O10624.8126 + [31]
IsochaininC33H54O10610.7858 + [35]
KabicidinC35H60O13688.8536 + [36]
LienomycinC67H107NO181214.5825++ +[37]
Moldicidin AC42H81NO19904.1005 + [16]
NorchaininC32H52O10596.7589 + [31]
Onomycin-I
Onomycin-II
C43H76NO17
C42H67NO17
879.0730
857.9905
+ [38]
PentacidinC31H50O10582.7320 + [16]
PentafunginC41H74NO16837.0357 + [39]
PA-153C37H61NO14743.8897 + [28]
S 728C56H93NO201100.3492 + [16]
Reedsmycin A-E
Reedsmycin F
C36H58O10
C36H58O11
650.8505
666.8499
+ [40]
SelvamicinC47H76O18929,0955 + [41]
Strevertene A
Strevertene B
Strevertene C
Strevertene D
Strevertene E
Strevertene F
Strevertene G
C31H48O10
C32H50O10
C32H50O10
C33H52O10
C33H52O10
C34H54O10
C31H50O9
580.7161
594.7430
594.7430
608.7699
608.7699
622.7968
566.7326
+ [42]
Takanawaene A
Takanawaene B
Takanawaene C
C30H48O8
C32H52O8
C33H51O8
536.7063
564.7601
578.7870
+ [43]
Thailandin A
Thailandin B
C39H62O14
C33H52O10
754.9208
608.7699
+ [44]
CYCLIC—HEXAENES ABAFAP/IHL/CLAC
Candihexin AC48H76NO19/C43H76NO19/C43H77NO19971.1268/911.0718/912.0797 + [45]
Candihexin BC48H90NO21/C48H91NO211017.2367/1018.2447 + [45]
Candihexin EC38H67NO16793.9471 + [45]
CryptocidinC52H84NO17995.2355++ [46]
GrecomycinC38H41O10/C38H38O10657.7375/654.712++ [47]
CYCLIC—HEPTAENES ABAFAP/IHL/CLAC
AcmycinC36H68NO30994.9247 + [48]
Amphotericin BC47H73NO17924.0932 ++ [49]
Aureofungin AC59H86N2O19/C59H88N2O191127.3339/1129.3498 + [50]
Aureofungin BC57H85NO19/C57H87NO191088.2972/1090.3131 + [50]
Candicidin A1 (VI, Levorin A0, Ascosin A1) *
Candicidin A2 (D, A, D1, Levorin A2, Ascosin A2)
Candicidin A3 (V, Levorin A3, Ascosin A3)
C59H84N2O17
C59H84N2O18
C59H86N2O18
1093.3272
1109.3186
1111.3345
+ + [51]
CandidinC47H71NO17922.0773 + [52]
Flavumycin AC60H91N2O17/C54H79NO161112.3858/998.2184 + [53]
FungimycinC59H86N2O171095.3351 +++ [54]
Hamycin AC58H86N2O191115.3229 +++ [55]
Isolevorin A2C60H86N2O181123.3455 + [56]
Levorin A2C59H86N2O18/C59H89N2O181111.3345/1114.3583 + [57]
Levorin BC62H98N2O251271.4586 + [58]
LucknomycinC61H98N2O24/C54H80N2O191243.4482/1061.2313 + [59]
Partricin AC59H86N2O9967.3399 + [60]
Partricin BC55H84N2O191077.2740 + [60]
Perimycin AC59H88N2O171097.3510 + [61]
Trichomycin AC58H84N2O18/C61H86N2O211097.3076/1183.3547 ++ [62]
AF-1231C42H68N2O17873.0052 + [16]
DJ-400 B1
DJ-400 B2
C65H96N2O21
C58H86N2O20
1241.4781
1131.3223
+ [38]
67-121 A
67-121 C
C59H88N2O19
C65H98N2O28
1129.3498
1355.4898
+ [38]
NPP B1C55H86N2O221127.2881 + [18]
LINEAR POLYENESABAFAP/IHL/CLAC
AB023a
AB023b
C31H50O8
C32H52O8
550.7332
564.7601
+ [63]
ClethramycinC63H99N3O18S1218.5545 + [64]
ECO-02301C70H109N2O201298.6369 + [65]
EtnangienC49H76O11841.1358+ [66]
Linearmycin A
Linearmycin B
Linearmycin C
C64H101NO16
C66H103NO16
C67H105NO16
1140.5031
1166.5410
1180.5678
++ + [67]
Mediomycin
Mediomycin A
Mediomycin B
C62H99NO16S
C62H97NO18S
C62H97NO15
1146.5312
1176.5141
1096.4499
+ [64]
MeijiemycinC66H105NO191216.5550 + [68]
MycangimycinC20H24O4328.4082 + [69]
Neotetrafibricin AC67H105NO191228.5660 + [64]
Table 2. Non-polyene SMs of actinomycetes targeting human cells.
Table 2. Non-polyene SMs of actinomycetes targeting human cells.
CompoundFormulaCalculated Average MassActivitiesRef.
PEPTIDES
Surugamide AC48H81N9O8912.21428anticancer, antifungal[70]
PolyoxypeptinC35H60O14704.84403pro-apoptotic[71]
BleomycinC55H84N17O21S3+1415.55415anti-cancer[72]
Actinomycin DC62H86N12O161255.41969anti-cancer[73]
MirubactinC26H32N6O11604.56701siderophore[74]
ANTIMYCINS
Antimycin AC24H40N2O9548.62641inhibitor of respiration[75]
NON-POLYENIC MACROLIDES
FK506 (Tacrolimus)C44H69NO12804.02005immunosuppressive, antifungal[76]
FK520 (Ascomycin)C43H69NO12792.00931immunosuppressive, antifungal[77]
MeridamycinC45H75NO12822.07844neuroprotective[78]
NemadectinC36H52O8612.79481antiparasitic[79]
Sirolimus (Rapamycin)–a cyclic molecule containing conjugated triene. C51H79NO13914.17404immunosuppressive, antifungal[80]
Venturicidin BC40H66O10706.94776antifungal[81]
NON-POLYENIC POLYKETIDES
ActiphenolC15H17NO4275.30042proteosynthesis inhibitor[82]
Kinamycin FC18H14N2O7370.31373anti-cancer[83]
Neoansamycin AC30H37NO7523.61855antibiotic, antiviral[84]
NogalamycinC39H49NO16787.80515anti-cancer[85]
Reveromycin AC36H52O11660.79303EGF inhibitor[86]
OTHER—ACTIVE ON THE HUMAN CELLS
NeocarzinostatinC35H33NO12659.63751anti-cancer[87]
NocardamineC27H48N6O9600.70599anti-cancer siderophore[88]
Table 3. Inhibitory activity of Streptomyces spp. against Candida albicans.
Table 3. Inhibitory activity of Streptomyces spp. against Candida albicans.
BCCO Strain No.Metabolite ProducedStreptomyces sp. (Vertical Line)
Candida albicans (Horizontal Line)
Size of Inhibitory Zone (mm)Size of Hemolytic Zone (mm)
10_1099candicidin A, A1, A3; surugamide AIjms 23 15045 i001107
10_1093tetrafungin, tetrin A, polyene BIjms 23 15045 i00259
10_1094tetrafungin, tetrin A, polyene BIjms 23 15045 i00339
10_1095tetrafungin, tetrin A, polyene BIjms 23 15045 i00437
10_1104tetrafungin, tetrin A, polyene BIjms 23 15045 i00588
10_2282tetrafungin, tetrin A, polyene BIjms 23 15045 i0061216
Table 4. Inhibitory activity of Streptomyces spp. against filamentous fungi. Streptomycetes were inoculated in a square shape and the fungi were inoculated in lines. The size of the observed inhibitory zones is presented in mm above each picture. Pictures of hemolytic zones of streptomycetes were taken from the bottom of the dishes.
Table 4. Inhibitory activity of Streptomyces spp. against filamentous fungi. Streptomycetes were inoculated in a square shape and the fungi were inoculated in lines. The size of the observed inhibitory zones is presented in mm above each picture. Pictures of hemolytic zones of streptomycetes were taken from the bottom of the dishes.
BCCO Strain No./Found MetabolitePaecilomyces spp.Fusarium spp.Aspergillus fumigatusAspergillus nigerCultivation Timing/Streptomycete Hemolysis
10_22596000Streptomyces 144 h; fungus 72 h
actiphenol (?)Ijms 23 15045 i007Ijms 23 15045 i008Ijms 23 15045 i009Ijms 23 15045 i010Ijms 23 15045 i011
10_109910no growth06Streptomyces 96 h; fungus 72h
candicidinIjms 23 15045 i012Ijms 23 15045 i013Ijms 23 15045 i014Ijms 23 15045 i015Ijms 23 15045 i016
10_11068002Streptomyces 96 h; fungus 72 h
polyene AIjms 23 15045 i017Ijms 23 15045 i018Ijms 23 15045 i019Ijms 23 15045 i020Ijms 23 15045 i021
10_109211no growth38Streptomyces 144 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i022Ijms 23 15045 i023Ijms 23 15045 i024Ijms 23 15045 i025Ijms 23 15045 i026
10_10938no growth16Streptomyces 96 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i027Ijms 23 15045 i028Ijms 23 15045 i029Ijms 23 15045 i030Ijms 23 15045 i031
10_109420no growth15Streptomyces 96 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i032Ijms 23 15045 i033Ijms 23 15045 i034Ijms 23 15045 i035Ijms 23 15045 i036
10_10955004Streptomyces 96 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i037Ijms 23 15045 i038Ijms 23 15045 i039Ijms 23 15045 i040Ijms 23 15045 i041
10_109712009Streptomyces 96 h; fungus 72 h
polyene BIjms 23 15045 i042Ijms 23 15045 i043Ijms 23 15045 i044Ijms 23 15045 i045Ijms 23 15045 i046
10_1104243316Streptomyces 144 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i047Ijms 23 15045 i048Ijms 23 15045 i049Ijms 23 15045 i050Ijms 23 15045 i051
10_228220006Streptomyces 144 h; fungus 72 h
tetrafungin, tetrin A, polyene BIjms 23 15045 i052Ijms 23 15045 i053Ijms 23 15045 i054Ijms 23 15045 i055Ijms 23 15045 i056
10_2389no growth0012Streptomyces 144 h; fungus 72 h
polyene BIjms 23 15045 i057Ijms 23 15045 i058Ijms 23 15045 i059Ijms 23 15045 i060Ijms 23 15045 i061
10_230924004Streptomyces 144 h; fungus 72 h
polyene C (?)Ijms 23 15045 i062Ijms 23 15045 i063Ijms 23 15045 i064Ijms 23 15045 i065Ijms 23 15045 i066
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bobek, J.; Filipová, E.; Bergman, N.; Čihák, M.; Petříček, M.; Lara, A.C.; Kristufek, V.; Megyes, M.; Wurzer, T.; Chroňáková, A.; et al. Polyenic Antibiotics and Other Antifungal Compounds Produced by Hemolytic Streptomyces Species. Int. J. Mol. Sci. 2022, 23, 15045. https://doi.org/10.3390/ijms232315045

AMA Style

Bobek J, Filipová E, Bergman N, Čihák M, Petříček M, Lara AC, Kristufek V, Megyes M, Wurzer T, Chroňáková A, et al. Polyenic Antibiotics and Other Antifungal Compounds Produced by Hemolytic Streptomyces Species. International Journal of Molecular Sciences. 2022; 23(23):15045. https://doi.org/10.3390/ijms232315045

Chicago/Turabian Style

Bobek, Jan, Eliška Filipová, Natalie Bergman, Matouš Čihák, Miroslav Petříček, Ana Catalina Lara, Vaclav Kristufek, Melinda Megyes, Theresa Wurzer, Alica Chroňáková, and et al. 2022. "Polyenic Antibiotics and Other Antifungal Compounds Produced by Hemolytic Streptomyces Species" International Journal of Molecular Sciences 23, no. 23: 15045. https://doi.org/10.3390/ijms232315045

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