Next Article in Journal
Garlic Peel-Based Biochar Prepared under Weak Carbonation Conditions for Efficient Removal of Methylene Blue from Wastewater
Previous Article in Journal
A Comprehensive Review on Deep Eutectic Solvents: Their Current Status and Potential for Extracting Active Compounds from Adaptogenic Plants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Phenazine Natural Products: Chemical Structures and Biological Activities

1
School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
2
Shandong Freda Biotech Co., Ltd., Jinan 250101, China
3
CAS Key Lab of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
4
College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
*
Authors to whom correspondence should be addressed.
Molecules 2024, 29(19), 4771; https://doi.org/10.3390/molecules29194771
Submission received: 9 September 2024 / Revised: 3 October 2024 / Accepted: 5 October 2024 / Published: 9 October 2024

Abstract

:
Phenazine natural products are a class of colored nitrogen-containing heterocycles produced by various microorganisms mainly originating from marine and terrestrial sources. The tricyclic ring molecules show various chemical structures and the decorating groups dedicate extensive pharmacological activities, including antimicrobial, anticancer, antiparasitic, anti-inflammatory, and insecticidal. These secondary metabolites provide natural materials for screening and developing medicinal compounds in the field of medicine and agriculture due to biological activities. The review presents a systematic summary of the literature on natural phenazines in the past decade, including over 150 compounds, such as hydroxylated, O-methylated, N-methylated, N-oxide, terpenoid, halogenated, glycosylated phenazines, saphenic acid derivatives, and other phenazine derivatives, along with their characterized antimicrobial and anticancer activities. This review may provide guidance for the investigation of phenazines in the future.

1. Introduction

Natural products are considered to be important sources of drug discovery. As the technologies for isolation, purification, and detection developed, great interest has been shown in underexplored natural products [1,2]. The first phenazine product, “pyocyanine” (now known as pyocyanin, PYO), was determined as a blue pigment from purulent wounds of patients in 1859; however, its structure was not established until nearly 100 years later [3]. Since PYO was identified as a Pseudomonas aeruginosa metabolite, more than 6000 kinds of phenazines have been found, of which there are over 100 phenazine natural products [4,5,6,7]. The skeleton structure of phenazine derivatives is a pyrazine ring (1,4-diazabenzene) coupling two annulated benzenes. The physical and chemical properties of phenazines depend on the types and positions of different substituent functional groups. Their antibiotic, antifungal, antiparasitic, antimalarial, insecticidal, and antitumor biological activities are presumed to be derived from the toxic reactive oxygen species (ROS) formation ability, the facilitation of energy generation, involvement in iron homeostasis via Fe (III) reduction, participation as signal molecules via the activation of the Fe-containing transcription factor SoxR, DNA Π–Π interaction and intercalation, and biofilm morphogenesis [8]. Some natural phenazines might be candidates or prodrugs not only for both bacteria and fungi but also as anticancer or antitumor agents. For example, Clofazimine or Lamprene™, which is a riminophenazine compound, has been successfully applied in clinics for the treatment of leprosy and tuberculosis due to antimicrobial activity and immunosuppressive properties [9]. Fused aryl phenazine derivatives XR11576, XR5944, NC-182, and NC-190 show significant anticancer activity, and some of them are also under clinical studies (Figure 1) [10]. One of the natural phenazine products, phenazine-1-carboxylic acid (PCA, 1), has been developed as a commercialized biopesticide “Shenqinmycin” in China since 2011 and is widely used to treat fungal diseases in crops (Figure 2) [11,12]. Natural phenazines are produced directly from various microbes, such as Pseudomonas spp., Streptomyces spp., Vibrio spp., Burkholderia spp., Brevibacterium spp., Pseudonocardia spp., and archaeal Methanosarcina spp., etc. [13,14]. The biological synthesis of phenazines has been extensively studied for a long time due to their beneficial interactions with plants and animals. Chorismate, an intermediate of the shikimate pathway, is currently known as the common phenazine biosynthetic precursor. The phz-operon, which has been proven to spread in all phenazine-producing Gram-negative and Gram-positive bacteria by horizontal gene transfer [15,16], codes five conserved enzymes: ketosteroid isomerase (PhzB), isochorismatase (PhzD), anthranilate synthase (PhzE), diaminopimelate epimerase similar protein (PhzF), and FMN-dependent enzyme (PhzG). These phz-operon enzymes could convert chorismic acid into PCA and/or phenazine-1,6-dicarboxylic acid (PDC), which are catalyzed to create more complex structures by modification enzymes (proteins for hydroxylation, decarboxylation, methylation, N-oxidation, glycosylation, and prenylation, etc.). The progress regarding biosynthesis and metabolic engineering research of phenazines in the past decades has been summarized in other reviews [13,17,18].
Over the past decade, research reports on natural phenazines isolated and structurally identified have been constantly growing, and phenazines have increasingly attracted people’s attention in the field of medicine and agriculture. This review presents a comprehensive and methodical complication of the chemical structures of phenazines (hydroxylated, O-methylated, N-methylated, N-oxide, terpenoid, halogenated, glycosylated phenazines, saphenic acid derivatives, and other phenazine derivatives), along with their associated biological activities and pharmacological effectiveness. The aim of this work is to elucidate the structure and function to find suitable applications of phenazines.

2. Structural Diversities and Biological Activities

2.1. Basic Phenazine Structures

Phenazine heterocycles contain fused benzene moieties at the carbon positions of a pyrazine nucleus (Figure 2). Within the scope of existing knowledge, all phenazine molecules are derived from two basic carboxylate-bearing phenazines, PCA (1) and PDC (2) (Figure 2). In recent years, the bioactivity of PCA and PDC has been further evaluated. PCA has been isolated from a large number of microbes, such as Streptomyces, Pseudomonas, Lysobacter, Xenorhabdus, Truncatella, Paenibacillus, Rhodococcus, Klebsiella, Kitasatospora, Burkholderia, and Bacillus, etc. It shows broad-spectrum antifungal activity against a series of phytopathogens like Phytophthora apsica, Gibberella zeae, Verticillium dahlia, Phaeoacremonium minimum, Fomitiporia mediterranea, Neofusicoccum parvum, Sclerotinia sclerotiorum, Phyricularia grisea, Streptomyces scabies, and Rhizoctonia solani [19,20,21,22,23,24,25,26]. Based on its outstanding biological activities, PCA was developed as a new registered biopesticide, Shenqinmycin, in 2011 by Xu and colleagues [11,12]. At present, it is widely used in China as a key fungicide to prevent rice sheath blight, pepper blight, downy mildew, leaf spot, fusarium head blight, canker, etc. [27]. PDC, with two carboxyl groups at 1, 6-position of the phenazine ring is commonly isolated from Streptomyces spp., Erwinia herbicola, Pseudomonas spp., and Lysobacter antibioticus. It has lethal activity against Gram-negative (e.g., E. coli) and Gram-positive (e.g., Bacillus subtilis) bacteria at a concentration of 37 mM [11,28,29,30,31,32]. In addition, PDC shows cytotoxicity against MCF7, HeLa, and HT29 cells of breast, cervical, and prostate cancer [30].

2.2. Phenazines with Hydroxyl and Methoxyl Moieties

The two most common types of post-modification in natural phenazines are hydroxylation and hydroxy-methylation. Ten hydroxylated and O-methylated phenazines (310, 03, 08) are summarized in this review (Figure 2). 1-Hydroxyphenazine (3) and 2-hydroxyphenazine (4) with a single hydroxyl, 1,2-dihydroxyphenazine (5), and 1,6-dihydroxyphenazine (6) carry two hydroxyl groups at different positions, and 1-carboxyl-6-formyl-4,7,9-trihydroxy-phenazine (CFTHP, 7) bearing three hydroxyl groups at 4,7,9 position have all been reported as phenazine hydroxylation products [33]. They are mostly present in Streptomyces spp., Pseudomonas spp., Rhodococcus spp., Sphingomonas wittichii, Lysobacter antibioticus, and Xenorhabdus szentirmaii, and can inhibit Aeromonas hydrophila, Fusarium oxysporum, Rhizoctonia solani, Pythiumultimum, Alternaria alternata, Alternaria solani, Colletotrichum acutatum, Curvularia andropogonis, Fusarium moniliforme, Pythium aphanidermatum, and tumor cells [34,35,36,37,38,39]. Compound 4 was reported to possess a better inhibitory effect on the take-all disease of wheat than PCA [40]. Two O-methylated phenazines compounds 1,6-dimethoxyphenazine (8) and 1-hydroxy-6-methoxyphenazine (9) were isolated from Lysobacter antibioticus OH13 with weak antibacterial activity [39]. Further, 1-methoxyphenazine (03) and methyl-6-methoxyphenazine-1-carboxylate (08) were isolated from Streptomyces luteireticuli NIIST-D75, and both compounds were shown explicitly in elaborate in vitro experiments as potentiators of ciprofloxacin for antibacterial activity with highly reduced dosage [41]. Strepphenazine A (10), which contains hydroxyl, carboxyl, and methoxyl groups, has been detected in the culture broth of Streptomyces strain YIM PH20095, showing antifungal activity against Fusarium oxysporum, Plectosphaerella cucumerina, Alternaria panax, and Phoma herbarum, which prevented root-rot disease in Panax notoginseng with minimal inhibitory concentrations of 16–32 μg/mL [42]. This study shows that Streptomyces strain YIM PH20095 provides new sources for the development of biological control agents to prevent the infection of pathogenic fungi of P. notoginseng. Although O-methylated natural products are very common, the O-methylation is important for the antibiotic activity of phenazines. For example, compound 03 has much higher antifungal activity against Bipolaris maydis, Alternaria solani, and Aspergillus flavus than compound 3 [43]. Besides that, the antimicrobial activity of myxin (12) is also much higher than iodinin (11) (Figure 3) [44].

2.3. N-Oxide Phenazines

Molecules with N-oxide can be found in various natural sources including plants, microorganisms, and animals. The N-oxide group may provide water solubility or decrease membrane permeability or immunogenicity and has a special redox reactivity. The level of N-oxide functionality is essential for cytotoxicity. These features render the phenazine N-oxides particularly valuable as starting points for antitumor agents, which selectively promote apoptotic cell death at low levels of oxygen saturation [45,46]. Nine natural phenazine N-oxides (1119) are described in this paper (Figure 3). Compound iodinin (1,6-dihydroxyphenazine-5,10-dioxide, 11), which has been identified in Streptosporangium sp. DSM 45942, and Lysobacter antibioticus OH13, is the N5, N10-dioxide of 1,6-dihydroxyphenazine [47,48]. The compound demonstrates high antimicrobial and cytotoxic activity and is particularly potent against acute myeloid leukemia, acute promyelocytic leukemia cells, and other leukemia cells, with EC50 values for cell death up to 40 times lower for leukemia cells when compared with normal cells. The results of experiments show that 11 represents a promising structure in the development of anticancer therapy. Another well-known compound is myxin (1-hydroxy-6-methoxyphenazine-N5, N10-dioxide, 12) isolated from Lysobacter antibioticus, which has significant antimicrobial activity. DNA intercalation, the production of reactive oxygen species (ROS) and inhibition of topoisomerases and metal chelation are proposed action modes for the biological activity of iodinin and myxin [49]. Other phenazine oxides produced by Lysobacter antibioticus, including 1,6-dihydroxyphenazine-N5-oxide (DHPO, 13), 1,6-dimethoxyphenazine-N5-oxide (14), 1-hydroxy-6-methoxyphenazine-N5-oxide (15), and 1,6-dimethoxyphenazine-N5,N10-dioxide (16) [39]. Izumiphenazine D (17), characterized by Streptomyces sp. IFM 11204, is the first example of quinoline-N-oxide containing phenazine natural product. Compound 17 has a synergistic effect in combination with TRAIL against AGS cells, and does not exhibit inhibition of Wnt signal transcription activity even at 50 μM [50]. Recently, two novel phenazine N-oxides, 1-hydroxyphenazine-N10-oxide (18) and 1-methoxyphenazine-N10-oxide (19) were artificially designed and biosynthesized in Pseudomonas chlororaphis [43,51]. 1-methoxyphenazine-N10-oxide (19) shows better activity than other phenazine products against plant pathogenic fungi, Bipolaris maydis, Alternaria solani, Fusarium graminearum, Rhizoctonia solani, and Aspergillus flavus. NaphzNO1 from Nocardiopsis sp. 13-12-13 and LaPhzNO1 from L. antibioticus OH13 are responsible for adding an oxygen atom to the nitrogen atom in phenazine N-oxides biosynthesis, they are all NADPH-dependent, flavin-containing N-monooxygenases. NaphzNO1 catalyzes the formation of 18 from 3; however, it is unable to convert 03 into 19. Furthermore, LaPhzNO1 is able to convert 9 into 15 but not 12 and cannot convert 8 to any of the N-oxide products in the in vitro assays. These results demonstrate that once the neighboring hydroxyl groups at the C1 or C6 locations are methylated, the N-monooxygenase cannot oxidize the phenazine nitrogen atom and may provide clues on new phenazine derivatives biosynthesis [39].

2.4. Phenazines with N-Methyl Moiety

There are two N-methylated phenazine natural products, pyocyanin (PYO, 20) and 5-methyl phenazine-1-carboxylic acid (5MPCA) (Figure 3). Moreover, 90–95% of Pseudomonas aeruginosa produce one of the most famous phenazines PYO, a redox-active compound that has been heterologously expressed in E. coli [52]. It shows potential against bacterial and fungal strains and has many damaging effects on mammalian cells because of its ability to generate oxidative stress [53,54,55]. It is a fascinating molecule in many fields such as medicine, agriculture, aquaculture, and biosensors [5,56]. The precursor of PYO, 5MPCA, was also characterized as a secondary metabolite from Pseudomonas. 5MPCA exhibits selective cytotoxicity towards lung (A549) and breast (MDA MB-231) cancer cell lines in a dose-dependent manner, with an IC50 value of 488.7 ± 2.52 nM and 458.6 ± 2.48 nM, respectively [57]. It exhibits inhibition of cell viability and DNA synthesis and induces G1 cell-cycle arrest and apoptosis in cancer cells through the mitochondrial intrinsic pathway via the activation of caspase-3 and downregulation of Bcl-2 protein.

2.5. Phenazines with Carboxamide Moiety

Compounds 2125 are carboxamidated phenazines described in this review (Figure 4). Phenazine-1-carboxamide (PCN, 21) is mainly isolated from Pseudomonas, Streptomycetes, Nigrospora oryzae, and Pantoea agglomerans [58,59,60]. PCN exhibits antagonistic activities against a wide range of plant pathogenic fungi Fusarium oxysporum f. sp. niveum (causing fusarium wilt), Fusarium graminearum (causing fusarium head blight), and Rhizoctonia solani (causing rice sheath blight), etc. [61,62], as well as inhibitory effect on different cancer cell lines A549, HeLa, and SW480 between the concentration of 32 and 40 μM [63]. There was research that showed that PCN secreted by a bacterium Pseudomonas piscium ZJU60 from the wheat head directly manipulates the activity of the plant pathogenic fungus Fusarium graminearum histone acetyltransferase domain of FgGcn5 and suppresses fungal virulence, growth, and mycotoxin biosynthesis [64,65]. As a result, it provided a unique example that an antagonistic microbe could inhibit the virulence and growth of a plant pathogenic fungus by affecting its histone modification. PCA amide is a promising skeleton for designing more potent phenazine-based fungicides. 6-Hydroxyphenazine-1-carboxamide (22), methyl 6-carbamoylphenazine-1-carboxylate (23), chromophenazine C (24), and chromophenazine F (25) are four novel phenazine derivatives with carboxamide moiety obtained from Streptomycetes [66,67]. Compounds 22 and 23 exhibit moderate antifungal and antibacterial activities against Fusarium oxysporum (ATCC 7808), Fusarium solani (ATCC 36031), Staphylococcus aureus (ATCC 25923), and Staphylococcus albus (ATCC 10231), respectively [67]. However, the novel complicated compounds 24 and 25 did not show any activity against B. subtilis, E. coli, M. miehei, S. aureus, and C. albicans [66]. Further investigation on the bioactivity of compounds 2225 needs to be undertaken in the future.

2.6. Terpenoid Phenazines

Terpenoid phenazines are a large class of compounds and contain common structural features of isoprenylated C, N, and O side chains or complex structures. In this review, 24 terpenoid phenazines (2649) are summarized (Figure 5). However, most terpenoid phenazines exhibit moderate or weak antibacterial activity. A series of prenylated phenazine compounds have been discovered in Streptomyces. For instance, C-prenylated phenazines, including JBIR-46 (26), JBIR-47 (27), JBIR-48 (28), endophenazine A (29), endophenazine B (30), and a novel phenazine, endophenazine E (31), which represents a glutamine α-amino attached to the carboxyl group of endophenazine A. Compounds 29, 30, and 31 show antimicrobial activity against the Gram-positive Bacillus subtilis and Gram-negative E. coli [68]. Compounds 3235 are O-prenylated phenazines and phenazines 3643 are N-prenylated derivatives [69,70,71]. According to the result of the test, marinophenazine A/C (3233), phenaziterpene A (34), and phenaziterpene B (35) had no discernable biological activity [72]. Only chromophenazine D (42) demonstrated moderate effectiveness against bacteria among the six N-prenylated phenazines, chromophenazines A–F (3641) [66]. Lavanducyanin (42) and its congener 1-hydroxy-7-oxolavanducyanin (43) isolated from Streptomyces show intriguing cytotoxic activities [73]. Antibacterial assays revealed that 43 had significant but reduced anti-Gram-positive bacterial activity compared with 42, and both compounds were all inactive against Gram-negative bacteria [73]. Endophenaside B (44), endophenaside C (45), endophenaside D (46), and endophenazine F1 (47), which possess substantial antimicrobial activities, were isolated from Kitasatospora sp. [68]. Compounds 48 and 49, with terpenoid aliphatic chains at positions 1 and/or 6 of the phenazine ring, were identified in the Pseudomonas aeruginosa strain RRLJ 04 culture [74]. The strain exhibited growth promotion and disease control in pigeon peas, both compounds 48 and 49 showed in vitro antibiosis against Fusarium udum and can be further exploited for growth enhancement and disease control of other crops.

2.7. Glycosylated Phenazines

The increasing bacterial multi-drug resistance necessitates novel drug-discovery efforts. One way of obtaining novel chemistry is the decoration of molecules by glycosylation because the pharmacological properties of the parent scaffold can be dramatically influenced by glycosylation, such as in anthracycline, avermectin, aureolic acid, and enediyne antibiotics [75]. Glycosylation is a common modification of natural phenazine products with high diversity in both the sugar moieties and the targeted aglycones. A total of 28 glycosylated phenazines (5077) and their activities are described in this literature (Figure 6A,B). Compound 4-O-glucosyl-1-carboxyl-phenazine (50) from Streptomyces sp. strain UICC B-92 exhibited inhibitory potency against Gram-positive bacteria B. cereus strain ATCC 10876 and S. aureus strain ATCC 25923 [76]. A number of novel glycosylated phenazine-type antibiotic endophenasides A–E (51, 44, 45, 46, 52) were produced by Kitasatospora sp. MBT66 [68]. Endophenaside A (51) and endophenaside C (45) are phenazines that contain a rare 2′-O-methylation of the sugar moiety. Endophenasides A–E showed antimicrobial activity against the Gram-positive Bacillus subtilis or Gram-negative E. coli [68]. Nannocystis pusilla strain Ari7 has been demonstrated to produce 1,6-dihydroxyphenazine glycosylated derivative, 1-hydroxyphenazine-6-yl-α-D-arabinofuranoside (53) [77]. Novel glycosylated phenazine products solphenazines A–F (5459), along with izuminosides A–C (6062), were isolated from Streptomyces sp. strain DL-93 and IFM 11260, respectively. Compounds 5459 did not show any antifungal or antibacterial activity; however, 54, 55, and 59 displayed some cytotoxicity against HCT-116 cancer cells (Figure 6A), the cytotoxicity was not associated with DNA intercalations and topoisomerase inhibition and the mechanisms of action were uncertain [78]. Compounds 61 (10 μM) and 62 (60 μM) in combination with TRAIL showed synergistic activity in sensitizing TRAIL-resistance AGS cells. 2′-O-methylated/2′-O-unmethylated rhamnose glycosylated phenazines and two tautomeric glyceride naturally occurring phenazine products (6377) were characterized in Kitasatospora sp. MBT66 fermentation broth (Figure 6B) [79]. As described above, most glycosylated phenazine natural products do not have remarkable activity, and the activities of glycosylated phenazines need to be further investigated.
Besides that, unprecedented 5N-glucosylated phenazine derivatives 7-imino-5N-(1′β-D-glucopyranosyl)-5,7-dihydrophenazine1-carboxylic acid and 3-imino-5N-(1′β-D-glucopyranosyl)-3,5-dihydrophenazine-1-carboxylic acid were purified and identified when Bacillus sp. G2112 and Pseudomonas sp. G124 were co-cultivated. And 3-imino-5N-(1′β-D-glucopyranosyl)-3,5-dihydrophenazine-1-carboxylic acid did not inhibit Bacillus sp. G2112 which proved that the observed modification constitutes a resistance mechanism [80]. Caenorhabditis elegans, which is a useful model organism to study the xenobiotic detoxification pathways, can detoxify 3 by adding one, two, or three glucose molecules in N2 worms [81]. The structures of the trisaccharide sugar phenazines made by C. elegans were characterized, and the results showed that one of the sugar modifications contains an N-acetylglucosamine (GlcNAc) in place of glucose [82]. Xenobiotic detoxification can weaken the potential of the producing organism or pesticides against pathogens and should be considered during the development of biocontrol strategies.

2.8. Halogenated Phenazines

Halogenated phenazine natural products are rare in nature, and their biosynthetic pathways have not been discovered. The halogenated phenazines are 2-bromo-1-hydroxyphenazine (78) from Streptomyces sp. CNS284 showed strong cancer cell cytotoxicity (IC50 = 0.1 μM against HCT-116) and weak to moderate activity in the NF-κB-luciferase assay (IC50 = 73 μM) [83]. Six brominated phenazines, marinocyanins A–F (7984), that have strong to weak cytotoxicity against HCT-116 human colon carcinoma and possess modest antimicrobial activities against Staphylococcus aureus and Candida albicans were obtained from Streptomyces with the lavanducyanin chlorinated analog WS-9659B (85) (Figure 7) [84]. Up to now, there have been no reports on fluorinated or iodinated phenazine natural products. But a novel series of halogenated phenazines has been developed by chemical synthesis methods due to their potent antibacterial and biofilm eradication activities against critical Gram-positive pathogens, including Staphylococcus aureus, Staphylococcus epidermidis, and Enterococcus faecium [85,86]. These compounds have the potential to dramatically impact future therapies related to biofilm-associated infections.

2.9. Saphenic Acid Derivatives

Microorganisms are a rich source of structurally unique bioactive metabolites and drug candidates. Saphenic acid (6-(1-hydroxyethyl) phenazine-1-carboxylic acid, 86) isolated from Streptomyces, is a common pharmacophore for many antibiotics and antitumor reagents including DC-86-M and phenazostatins [87]. Moreover, its derivatives such as saphenyl ester (87), saphenamycin (88), esmeraldic acid (89), esmeraldin A (90), and esmeraldin B (91) are present in the metabolites of Streptomyces [87]. In particular, marine microbes from the deep sea are a relatively untapped reservoir of metabolites with structural and biological diversity waiting to be discovered. For example, saphenic amide (92), saphenol (93), 6-[1-(2-aminobenzoyloxy) ethyl]-1-phenazinecarboxylic acid (94), phenazostatins E-J (95100) were isolated from the culture broth of marine yeast-like fungus Cystobasidium laryngis derived from deep-sea sediments of the Indian Ocean Ridge (Figure 8) [31,88,89]. Compounds 92 and 93 showed a nitric oxide (NO) production-inhibitory effect against lipopolysaccharide (LPS)-induced murine macrophage RAW 264.7 cells without cytotoxicity at concentrations up to 30 µg/mL. Moreover, diphenazine phenazostatin J (100) exhibited significant antineuroinflammatory activity with an IC50 value of 0.30 μM and cytotoxicity against the NUGC-3 (stomach) cell line with an IC50 value of 7.7 nM, exhibiting 19-fold stronger activity than adriamycin. Interestingly, 100 demonstrated stronger cytotoxicity than its oxidized form (R)-6-(1-((6-acetylphenazine-1-carbonyl)oxy)ethyl) phenazine-1-carboxylic acid and positive control, indicating that the ester linkage and the hydroxyethyl group attached to the phenazine scaffold are essential for the activities [88]. Compound 100 could be a potential agent for the development of antineuroinflammatory and anticancer leads, and the underlying mechanisms for the biological activities and the structure–activity relationship are needed for further study.

2.10. Phenazines with Sulfur

The four phenazines with sulfur (61, 101103) are introduced in this review (Figure 9). Izuminoside B (61), the phenazine with a C-4 coupled methylsulfanyl group at the para-position to carboxylic acid has been obtained from Streptomyces sp. IFM 11260 [90]. The activity of compound 61 has been described in Section 2.7. Compound dermacozines J (101) isolated from Dermacoccus abyssi has been established to have an N-acetylcysteine moiety attached to C-9, and it demonstrated radical scavenging activity [91]. Panphenazine A (102) and B (103) were characterized by actinomycete Kitasatospora sp. HKI 714 as unusual pantetheine-containing phenazines, which were proposed to be synthesized by radical-induced conjugate addition [92]. This study indicated that phenazines possess the ability to capture biogenic thiols, crosslink proteins, and, ultimately, contribute to protein degeneration/aggregation processes.

2.11. Other Phenazine Derivatives with Special Structures

Recently, phenazine natural products with a wider range of structural variations (104150) have been identified from microorganisms. Red solids phenazines izumiphenazines A–C (104106) were isolated from Streptomyces, and izumiphenazine A (104) is the first instance of tetrahydrofuran ring connected phenazine dimer [93]. Compounds 104106 were evaluated to have a synergistic effect in combination with TRAIL (TNF-related apoptosis-inducing ligand) against AGS cells. Izumiphenazine C (106) analogs strepphenazine B–C (107108) have been characterized by Streptomyces with antifungal activities [42]. A novel dimeric phenazine, diastaphenazine (109), was also obtained from the fermentation broth of Streptomyces (Figure 10). Compound 109 has antibacterial activity against S. aureus but is inert against E. coli and C. albicans, according to the antimicrobial test [94]. Among the seven new phenazine-based metabolites—baraphenazines A–G (110116) (Figure 11)—compounds 104106 represent the first reported examples of fused 5-hydroxyquinoxaline/alpha-keto acid-based moiety [95]. Furthermore, baraphenazines D (113) and E (114) present two new diastaphenazine-type C-C-fused phenazine-based congeners, while baraphenazines F (115) and G (116) are two phenazinolin-type C-O-fused compounds. Only baraphenazines E (114) exhibited significant activity in antibacterial and fungal tests at the doses studied. A tin mine tailings-derived Streptomyces was found to create a novel family of diphenazines known as phenazinolins A–E (117121). Phenazinolins A–C (117119) exhibited amazing violet color and antagonistic activity for Bacillus subtilis, Staphylococcus aureus, Aspergillus niger, and Botrytis cinerea with MIC values in the range of 12–27 μM. The IC50 values of compounds 117119 in vitro cytotoxicity against GLC, P388, XWLC, and H460 human cancer cell lines were in the range of 14–40 μM (Figure 12) [96,97].
A series of new phenazine-type pigments dermacozines A–J (122130, 101) and dermacozines M–P (131134) were discovered in the fermentation media of Dermacoccus abyssi sp. strains isolated from Mariana Trench sediment (Figure 13) [98]. Dermacozine N (132) is the first linear pentacyclic oxazinophenazine natural product discovered [99]. This chemical family of compounds showed a low cytotoxic effect against resistant cancer cell lines. Dermacozines F (127) and G (128) displayed the most potent cytotoxic activity. Dermacozine C (124) had the strongest radical scavenging ability.
A recently discovered phenazine intermediate, 6-formylphenazine-1-carboxylic acid (135) was thought to bridge the pathways encoded by the BGCs in nematodes and the symbiotic entomopathogenic bacterium Xenorhabdus szentirmaii. A variety of non-enzymatic and enzymatic reactions can be used to generate demethoxygriseoluteic acid (136), griseoluteic acid (137), and a number of derivatives such as pelagiomicin B (138), phenaszentines A–C (139141), phenaszentines D (142), and E (143), phenaszenketides A–D (144147), phencomycin (148), phencocins A (149), and B (150) (Figure 14) from 135 [100].
Table 1 lists different types of phenazine natural products with various biological activities in this review.

3. Phenazines of Therapeutic Interest

There have been many biological investigations related to natural and synthtic phenazines that demonstrate therapeutic value related to human health and disease. Phenazinomycin isolated from Streptomyces sp. WK-2057 bears a cyclic terpenoid appendage. The compound shows moderate antibacterial activity against Staphylococcus aureus and direct cytotoxic activities against HeLa S3, P388, and P388 doxorubicin-resistant cells. In addition, phenazinomycin demonstrated antitumor activity against experimental murine tumors (Sarcoma 180 cells) in vivo, resulting in a prolongation of survival time for tumor-bearing mice (up to 140%) [101]. Iodinin (11) demonstrated that hypoxia-selective antileukemia activity (EC50 = 2.0 μM against normoxic cells, EC50 = 0.79 μM against hypoxic cells) has the potential to act on malignant cells in hypoxic bone marrow of AML patients [46]. The halogenated phenazine natural product, 2-bromo-1-hydroxyphenazine (78) with an MIC = 6.25 μM, is significantly more potent than pyocyanin (MIC = 50 μM). Synthetic halogenated phenazine compounds HP 1 (MIC = 1.56 μM) and HP 29 (MIC = 0.08 μM) have potent antibacterial activity and are able to eradicate bacterial biofilms. HP 29 has demonstrated especially good efficacy in dorsal wound infection models in mice [102]. Clofazimine has been received FDA approval to treat lepromatous leprosy (caused by Mycobacterium leprae) in 1986 and is on the World Health Organization’s (WHO) model list of essential medicines [103,104]. XR11576 is an oral topoisomerase I and II inhibitor, and bis-phenazine XR5944 is a novel cytotoxic agent that intercalates into DNA and inhibits transcription [105,106]. These two compounds have been under phase I clinical testing. NC-182 is a novel antitumor compound with a benzo-[a]phenazine ring. It is a potent DNA intercalator with nearly the same binding ability as daunomycin; however, the mode of interaction of NC-182 with DNA depends on the concentration of the drug, where the intercalative and electrostatic bindings are dominant at low and high concentrations of the drug, respectively [107]. NC-190 has been shown to induce topoisomerase II-dependent DNA cleavage and DNA fragmentation. The compound demonstrates anticancer activity against multiple cancer cell lines in vitro and in vivo (tumor models in animals), and the activities against HL-60 cells in a dose-dependent manner were comparable to that of etoposide, which is a clinically used cancer therapy [108,109].

4. Conclusions

In this review, we summarized the research on the structural variety and biological activity of natural phenazine products over the last ten years. Over 150 phenazines with various structures were introduced. To summarize, phenazine products are prevalent in nature; however, currently known natural phenazines are only found in microbes. Nevertheless, there are diverse substituent groups that appear on the phenazine ring including carboxyl, hydroxyl, methyl, hydroxymethyl, carboxamide, terpenoid, glycosyl, halogen, sulfur, and some other complicated derivative groups. It can be concluded that phenazines from Streptomyces are deemed more complex, diversified, and fascinating based on the structures of phenazine natural products derived from different sources. The simple post-modified phenazines are mainly from Gram-negative bacteria, for instance, Pseudomonas and Lysobacter antibioticus. However, in Gram-positive bacteria strains, especially Streptomyces, not only simple phenazine derivatives but also complex phenazines can be obtained, pointing out the direction of the search for novel complex phenazine natural structures. Phenazines with diverse structures exhibit powerful anticancer and antibiotic activities. Hydroxylation and hydroxy-methylation of phenazines are very important for the antibiotic activity of phenazines. For example, PCA, 2-hydroxyphenazine, 1-methoxyphenazine, and 1-methoxyphenazine N’ 10-oxide have great potential in preventing plant diseases and insect pests. Especially, PCA is widely used in southern China as a key fungicide to prevent rice sheath blight. Moreover, more than 100 companies in the world are now producing and selling PCA [110]. P. chlororaphis and P. fluorescens, which produce a wide range of bioactive phenazines, have been commercialized by companies from Europe (Cedomon BioAgri AB, Uppsala, Sweden) and the USA (AtEze, Turf Science Laboratories). PYO and PCN, which have potent antimicrobial activity, are typical representatives of N-methylated and carboxamidated phenazines, respectively. Halogenated phenazines can eradicate surface-attached biofilms with excellent potency, and phenazine di-N-oxides are an interesting class of antitumor agents that selectively kill the hypoxic cells found in solid tumors [49,111]. Most of the terpenoid and glycosylated phenazines do not have remarkable antibacterial activity. The biological activities of phenazines and the structure–activity relationship need further study.
Since PCA has been developed as a registered biopesticide, the application of phenazine derivatives will be promising in the field of agriculture pathogenic fungi control. The biosynthesis studies pave the way to create the green manufacturing approach of diverse phenazines. Although enzymes involved in phenazines, methylation, hydroxylation, glycosylation, and isoprenylation have been elucidated, and novel phenazine products have been artificially designed using these post-modification proteins, and there are still plenty of intriguing hidden biosynthetic pathways to be discovered, such as dimerization, halogenation, saphenic acid pathway derivatization, and methanophenazine biosynthesis. Therefore, there is a need to find pivotal enzyme reactions for the biosynthesis of phenazines with excellent activity. In most cases, the titer of naturally occurring products is insufficient for the application. Efforts should be undertaken to optimize the host strains in order to satisfy the demands of an industrial-scale production.

Author Contributions

Conceptualization, methodology, validation, formal analysis, investigation, data curation, writing—original draft preparation, W.H., Y.W., S.Z., H.S., C.W., Z.Z. and P.X.; writing—review and editing, visualization, supervision, project administration, funding acquisition, F.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Scientific Research Starting Foundation of Shandong University of Technology, grant number 4041/422046.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Melander, R.J.; Basak, A.K.; Melander, C. Natural products as inspiration for the development of bacterial antibiofilm agents. Nat. Prod. Rep. 2020, 37, 1454–1477. [Google Scholar] [CrossRef] [PubMed]
  2. Morán-Diez, M.E.; Glare, T.R. What are Microbial-based Biopesticides? Methods Mol. Biol. 2016, 1477, 1–10. [Google Scholar] [PubMed]
  3. Mentel, M.; Ahuja, E.G.; Mavrodi, D.V.; Breinbauer, R.; Thomashow, L.S.; Blankenfeldt, W. Of two make one: The biosynthesis of phenazines. ChemBioChem 2009, 10, 2295–2304. [Google Scholar] [CrossRef] [PubMed]
  4. Yan, J.; Liu, W.; Cai, J.; Wang, Y.; Li, D.; Hua, H.; Cao, H. Advances in phenazines over the past decade: Review of their pharmacological activities, mechanisms of action, biosynthetic pathways and synthetic strategies. Mar. Drugs 2021, 19, 610. [Google Scholar] [CrossRef] [PubMed]
  5. Jayaseelan, S.; Ramaswamy, D.; Dharmaraj, S. Pyocyanin: Production, applications, challenges and new insights. World J. Microbiol. Biotechnol. 2014, 30, 1159–1168. [Google Scholar] [CrossRef] [PubMed]
  6. Mavrodi, D.V.; Blankenfeldt, W.; Thomashow, L.S. Phenazine compounds in fluorescent Pseudomonas spp. biosynthesis and regulation. Annu. Rev. Phytopathol. 2006, 44, 417–445. [Google Scholar] [CrossRef]
  7. Mavrodi, D.V.; Parejko, J.A.; Mavrodi, O.V.; Kwak, Y.-S.; Weller, D.M.; Blankenfeldt, W.; Thomashow, L.S. Recent insights into the diversity, frequency and ecological roles of phenazines in fluorescent Pseudomonas spp. Environ. Microbiol. 2013, 15, 675–686. [Google Scholar] [CrossRef]
  8. Guttenberger, N.; Blankenfeldt, W.; Breinbauer, R. Recent developments in the isolation, biological function, biosynthesis, and synthesis of phenazine natural products. Bioorgan. Med. Chem. 2017, 25, 6149–6166. [Google Scholar] [CrossRef]
  9. Watanabe, F.; Fujiwara, K.; Furuuchi, K.; Ito, M.; Hanada, K.; Kodama, T.; Aono, A.; Mitarai, S.; Yoshiyama, T.; Kurashima, A.; et al. Clofazimine serum concentration and safety/efficacy in nontuberculous mycobacterial pulmonary disease treatment. Respir. Med. 2024, 231, 107718. [Google Scholar] [CrossRef]
  10. Moorthy, N.S.; Pratheepa, V.; Ramos, M.; Vasconcelos, V.; Fernandes, P. Fused Aryl-Phenazines: Scaffold for the Development of Bioactive Molecules. Curr. Drug Targets 2014, 15, 681–688. [Google Scholar] [CrossRef]
  11. Guo, S.; Wang, Y.; Bilal, M.; Hu, H.; Wang, W.; Zhang, X. Microbial Synthesis of Antibacterial Phenazine-1,6-dicarboxylic Acid and the Role of PhzG in Pseudomonas chlororaphis GP72AN. J. Agric. Food Chem. 2020, 68, 2373–2380. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, Y. Genomic Features and Regulation of Phenazine Biosynthesis in the Rhizosphere Strain Pseudomonas aeruginosa M18. In Microbial Phenazines: Biosynthesis, Agriculture and Health; Chincholkar, S., Thomashow, L., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 177–198. [Google Scholar]
  13. Serafim, B.; Bernardino, A.R.; Freitas, F.; Torres, C.A.V. Recent Developments in the Biological Activities, Bioproduction, and Applications of Pseudomonas spp. Phenazines. Molecules 2023, 28, 1368. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, S.; Cui, J.; Bilal, M.; Hu, H.; Wang, W.; Zhang, X. Pseudomonas spp. as cell factories (MCFs) for value-added products: From rational design to industrial applications. Crit. Rev. Biotechnol. 2020, 40, 1232–1249. [Google Scholar] [CrossRef] [PubMed]
  15. Laursen, J.B.; Nielsen, J. Phenazine Natural Products: Biosynthesis, Synthetic Analogues, and Biological Activity. Chem. Rev. 2004, 104, 1663–1686. [Google Scholar] [CrossRef]
  16. Blankenfeldt, W.; Parsons, J.F. The structural biology of phenazine biosynthesis. Curr. Opin. Struct. Biol. 2014, 29, 26–33. [Google Scholar] [CrossRef]
  17. Bilal, M.; Wang, S.; Iqbal, H.M.N.; Zhao, Y.; Hu, H.; Wang, W.; Zhang, X. Metabolic engineering strategies for enhanced shikimate biosynthesis: Current scenario and future developments. Appl. Microbiol. Biotechnol. 2018, 102, 7759–7773. [Google Scholar] [CrossRef]
  18. Bilal, M.; Guo, S.; Iqbal, H.M.N.; Hu, H.; Wang, W.; Zhang, X. Engineering Pseudomonas for phenazine biosynthesis, regulation, and biotechnological applications: A review. World J. Microb. Biotechnol. 2017, 33, 191. [Google Scholar] [CrossRef]
  19. Padaria, J.C.; Tarafdar, A.; Raipuria, R.; Lone, S.A.; Gahlot, P.; Shakil, N.A.; Kumar, J. Identification of phenazine-1-carboxylic acid gene (phc CD) from Bacillus pumilus MTCC7615 and its role in antagonism against Rhizoctonia solani. J. Basic Microbiol. 2016, 56, 999–1008. [Google Scholar] [CrossRef]
  20. Patel, N.P.; Raju, M.; Haldar, S.; Chatterjee, P.B. Characterization of phenazine-1-carboxylic acid by Klebsiella sp. NP-C49 from the coral environment in Gulf of Kutch, India. Arch. Microbiol. 2020, 202, 351–359. [Google Scholar] [CrossRef]
  21. Radhakrishnan, N.A.; Ravi, A.; Joseph, B.J.; Jose, A.; Jithesh, O.; Krishnankutty, R.E. Phenazine 1-carboxylic acid producing seed harbored endophytic bacteria from cultivated rice variety of Kerala and its broad range antagonism to diverse plant pathogens. Probiotics Antimicrob. Proteins 2023, 15, 516–523. [Google Scholar] [CrossRef]
  22. Hane, M.; Wijaya, H.C.; Nyon, Y.A.; Sakihama, Y.; Hashimoto, M.; Matsuura, H.; Hashidoko, Y. Phenazine-1-carboxylic acid (PCA) produced by Paraburkholderia phenazinium CK-PC1 aids postgermination growth of Xyris complanata seedlings with germination induced by Penicillium rolfsii Y-1. Biosci. Biotechnol. Biochem. 2021, 85, 77–84. [Google Scholar] [CrossRef] [PubMed]
  23. Shah, S.G.; Shier, W.T.; Jamaluddin; Tahir, N.; Hameed, A.; Ahmad, S.; Ali, N. Penicillium verruculosum SG: A source of polyketide and bioactive compounds with varying cytotoxic activities against normal and cancer lines. Arch. Microb. 2014, 196, 267–278. [Google Scholar] [CrossRef] [PubMed]
  24. Cimmino, A.; Bahmani, Z.; Castaldi, S.; Masi, M.; Isticato, R.; Abdollahzadeh, J.; Amini, J.; Evidente, A. Phenazine-1-carboxylic Acid (PCA), produced for the first time as an antifungal metabolite by Truncatella angustata, a causal agent of grapevine trunk diseases (GTDs) in Iran. J. Agric. Food Chem. 2021, 69, 12143–12147. [Google Scholar] [CrossRef] [PubMed]
  25. Pagmadulam, B.; Tserendulam, D.; Rentsenkhand, T.; Igarashi, M.; Sawa, R.; Nihei, C.-i.; Nishikawa, Y. Isolation and characterization of antiprotozoal compound-producing Streptomyces species from Mongolian soils. Parasitol. Int. 2020, 74, 101961. [Google Scholar] [CrossRef]
  26. Liu, H.; He, Y.; Jiang, H.; Peng, H.; Huang, X.; Zhang, X.; Thomashow, L.S.; Xu, Y. Characterization of a phenazine-producing strain Pseudomonas chlororaphis GP72 with broad-spectrum antifungal activity from green pepper rhizosphere. Curr. Microbiol. 2007, 54, 302–306. [Google Scholar] [CrossRef]
  27. Guo, S.; Hu, H.; Wang, W.; Bilal, M.; Zhang, X. Production of antibacterial questiomycin A in metabolically engineered Pseudomonas chlororaphis HT66. J. Agric. Food Chem. 2022, 70, 7742–7750. [Google Scholar] [CrossRef]
  28. Shahid, I.; Han, J.; Hardie, D.; Baig, D.N.; Malik, K.A.; Borchers, C.H.; Mehnaz, S. Profiling of antimicrobial metabolites of plant growth promoting Pseudomonas spp. isolated from different plant hosts. 3 Biotech 2021, 11, 48. [Google Scholar] [CrossRef]
  29. Guo, S.; Wang, Y.; Dai, B.; Wang, W.; Hu, H.; Huang, X.; Zhang, X. PhzA, the shunt switch of phenazine-1,6-dicarboxylic acid biosynthesis in Pseudomonas chlororaphis HT66. Appl. Microbiol. Biotechnol. 2017, 101, 7165–7175. [Google Scholar] [CrossRef]
  30. Dasgupta, D.; Kumar, A.; Mukhopadhyay, B.; Sengupta, T.K. Isolation of phenazine 1,6-di-carboxylic acid from Pseudomonas aeruginosa strain HRW.1-S3 and its role in biofilm-mediated crude oil degradation and cytotoxicity against bacterial and cancer cells. Appl. Microbiol. Biotechnol. 2015, 99, 8653–8665. [Google Scholar] [CrossRef]
  31. S Hifnawy, M.; Hassan, H.M.; Mohammed, R.; M Fouda, M.; Sayed, A.M.; A Hamed, A.; F AbouZid, S.; Rateb, M.E.; Alhadrami, H.A.; Abdelmohsen, U.R. Induction of Antibacterial Metabolites by Co-Cultivation of Two Red-Sea-Sponge-Associated Actinomycetes Micromonospora sp. UR56 and Actinokinespora sp. EG49. Mar. Drugs 2020, 18, 243. [Google Scholar] [CrossRef]
  32. Abdelfattah, M.S.; Kazufumi, T.; Ishibashi, M. New pyranonaphthoquinones and a phenazine alkaloid isolated from Streptomyces sp. IFM 11307 with TRAIL resistance-overcoming activity. J. Antibiot. 2011, 64, 729–734. [Google Scholar] [CrossRef]
  33. Deng, R.-X.; Zhang, Z.; Li, H.-L.; Wang, W.; Hu, H.-B.; Zhang, X.-H. Identification of a novel bioactive phenazine derivative and regulation of phoP on its production in Streptomyces lomondensis S015. J. Agric. Food Chem. 2021, 69, 974–981. [Google Scholar] [CrossRef] [PubMed]
  34. Qi, X.; Xue, M.; Cui, H.; Yang, K.; Song, K.; Zha, J.; Wang, G.; Ling, F. Antimicrobial activity of Pseudomonas monteilii JK-1 isolated from fish gut and its major metabolite, 1-hydroxyphenazine, against Aeromonas hydrophila. Aquaculture 2020, 526, 735366. [Google Scholar] [CrossRef]
  35. Prabhu, M.S.; Walawalkar, Y.D.; Furtado, I. Purification and molecular and biological characterisation of the 1-hydroxyphenazine, produced by an environmental strain of Pseudomonas aeruginosa. World J. Microbiol. Biotechnol. 2014, 30, 3091–3099. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, F.; Yang, S.; Xu, F.; Zhang, Z.; Lu, Y.; Zhang, J.; Wang, G. Characteristics of biological control and mechanisms of Pseudomonas chlororaphis zm-1 against peanut stem rot. BMC Microbiol. 2022, 22, 9. [Google Scholar] [CrossRef]
  37. Wan, Y.; Liu, H.; Xian, M.; Huang, W. Biosynthesis and metabolic engineering of 1-hydroxyphenazine in Pseudomonas chlororaphis H18. Microb. Cell Factories 2021, 20, 235. [Google Scholar] [CrossRef]
  38. Castaldi, S.; Masi, M.; Sautua, F.; Cimmino, A.; Isticato, R.; Carmona, M.; Tuzi, A.; Evidente, A. Pseudomonas fluorescens showing antifungal activity against Macrophomina phaseolina, a severe pathogenic fungus of soybean, produces phenazine as the main active metabolite. Biomolecules 2021, 11, 1728. [Google Scholar] [CrossRef]
  39. Zhao, Y.; Qian, G.; Ye, Y.; Wright, S.; Chen, H.; Shen, Y.; Liu, F.; Du, L. Heterocyclic aromatic N-oxidation in the biosynthesis of phenazine antibiotics from Lysobacter antibioticus. Org. Lett. 2016, 18, 2495–2498. [Google Scholar] [CrossRef]
  40. Liu, W.-H.; Yue, S.-J.; Feng, T.-T.; Li, S.; Huang, P.; Hu, H.-B.; Wang, W.; Zhang, X.-H. Characterization and Engineering of Pseudomonas chlororaphis LX24 with High Production of 2-Hydroxyphenazine. J. Agric. Food Chem. 2021, 69, 4778–4784. [Google Scholar] [CrossRef]
  41. Jesmina, A.R.S.; Induja, D.K.; Drissya, T.; Sruthi, C.R.; Raghu, K.G.; Nelson-Sathi, S.; Kumar, B.N.S.A.D.; Lankalapalli, R.S. In vitro antibacterial effects of combination of ciprofloxacin with compounds isolated from Streptomyces luteireticuli NIIST-D75. J. Antibiot. 2023, 76, 198–210. [Google Scholar] [CrossRef]
  42. Chen, X.; Hu, L.-F.; Huang, X.-S.; Zhao, L.-X.; Miao, C.-P.; Chen, Y.-W.; Xu, L.-H.; Han, L.; Li, Y.-Q. Isolation and characterization of new phenazine metabolites with antifungal activity against root-rot pathogens of panax notoginseng from Streptomyces. J. Agric. Food Chem. 2019, 67, 11403–11407. [Google Scholar] [CrossRef] [PubMed]
  43. Wan, Y.; Liu, H.; Xian, M.; Huang, W. Biosynthetic Pathway Construction and Production Enhancement of 1-Hydroxyphenazine Derivatives in Pseudomonas chlororaphis H18. J. Agric. Food Chem. 2022, 70, 1223–1231. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, J.; Guiza Beltran, D.; Schacht, A.; Wright, S.; Zhang, L.; Du, L. Functional and Structural Analysis of Phenazine O-Methyltransferase LaPhzM from Lysobacter antibioticus OH13 and One-Pot Enzymatic Synthesis of the Antibiotic Myxin. ACS Chem. Biol. 2018, 13, 1003–1012. [Google Scholar] [CrossRef] [PubMed]
  45. Huigens, R.W.; Brummel, B.R.; Tenneti, S.; Garrison, A.T.; Xiao, T. Pyrazine and Phenazine Heterocycles: Platforms for Total Synthesis and Drug Discovery. Molecules 2022, 27, 1112. [Google Scholar] [CrossRef] [PubMed]
  46. Viktorsson, E.Ö.; Melling Grøthe, B.; Aesoy, R.; Sabir, M.; Snellingen, S.; Prandina, A.; Høgmoen, Å.O.A.; Bonge-Hansen, T.; Døskeland, S.O.; Herfindal, L.; et al. Total synthesis and antileukemic evaluations of the phenazine 5,10-dioxide natural products iodinin, myxin and their derivatives. Bioorgani. Med. Chem. 2017, 25, 2285–2293. [Google Scholar] [CrossRef]
  47. Sletta, H.; Degnes, K.F.; Herfindal, L.; Klinkenberg, G.; Fjærvik, E.; Zahlsen, K.; Brunsvik, A.; Nygaard, G.; Aachmann, F.L.; Ellingsen, T.E.; et al. Anti-microbial and cytotoxic 1,6-dihydroxyphenazine-5,10-dioxide (iodinin) produced by Streptosporangium sp. DSM 45942 isolated from the fjord sediment. Appl. Microbiol. Biotechnol. 2014, 98, 603–610. [Google Scholar] [CrossRef]
  48. Myhren, L.E.; Nygaard, G.; Gausdal, G.; Sletta, H.; Teigen, K.; Degnes, K.F.; Zahlsen, K.; Brunsvik, A.; Bruserud, Ø.; Døskeland, S.O.; et al. Iodinin (1,6-dihydroxyphenazine 5,10-dioxide) from Streptosporangium sp. induces apoptosis selectively in myeloid leukemia cell lines and patient cells. Mar. Drugs 2013, 11, 332–349. [Google Scholar] [CrossRef]
  49. Viktorsson, E.Ö.; Aesoy, R.; Støa, S.; Lekve, V.; Døskeland, S.O.; Herfindal, L.; Rongved, P. New prodrugs and analogs of the phenazine 5,10-dioxide natural products iodinin and myxin promote selective cytotoxicity towards human acute myeloid leukemia cells. RSC Med. Chem. 2021, 12, 767–778. [Google Scholar] [CrossRef]
  50. Abdelfattah, M.S.; Toume, K.; Ishibashi, M. Izumiphenazine D, a new phenazoquinoline N-oxide from Streptomyces sp. IFM 11204. Chem. Pharm. Bull. 2011, 59, 508–510. [Google Scholar] [CrossRef]
  51. Guo, S.; Liu, R.; Wang, W.; Hu, H.; Li, Z.; Zhang, X. Designing an artificial pathway for the biosynthesis of a novel phenazine N-oxide in Pseudomonas chlororaphis HT66. ACS Synth. Biol. 2020, 9, 883–892. [Google Scholar] [CrossRef]
  52. Gonçalves, T.; Vasconcelos, U. Colour me blue: The history and the biotechnological potential of pyocyanin. Molecules 2021, 26, 927. [Google Scholar] [CrossRef]
  53. Saleem, H.; Mazhar, S.; Syed, Q.; Javed, M.Q.; Adnan, A. Bio-characterization of food grade pyocyanin bio-pigment extracted from chromogenic Pseudomonas species found in Pakistani native flora. Arab. J. Chem. 2021, 14, 103005. [Google Scholar] [CrossRef]
  54. Shouman, H.; Said, H.S.; Kenawy, H.I.; Hassan, R. Molecular and biological characterization of pyocyanin from clinical and environmental Pseudomonas aeruginosa. Microb. Cell Factories 2023, 22, 166. [Google Scholar] [CrossRef]
  55. Forbes, A.; Davey, A.K.; Perkins, A.V.; Grant, G.D.; McFarland, A.J.; McDermott, C.M.; Anoopkumar-Dukie, S. ERK1/2 activation modulates pyocyanin-induced toxicity in A549 respiratory epithelial cells. Chem.-Biol. Interact. 2014, 208, 58–63. [Google Scholar] [CrossRef]
  56. Abdelaziz, A.A.; Kamer, A.M.A.; Al-Monofy, K.B.; Al-Madboly, L.A. Pseudomonas aeruginosa’s greenish-blue pigment pyocyanin: Its production and biological activities. Microb. Cell Factories 2023, 22, 110. [Google Scholar] [CrossRef]
  57. Kennedy, R.K.; Naik, P.R.; Veena, V.; Lakshmi, B.S.; Lakshmi, P.; Krishna, R.; Sakthivel, N. 5-Methyl phenazine-1-carboxylic acid: A novel bioactive metabolite by a rhizosphere soil bacterium that exhibits potent antimicrobial and anticancer activities. Chem.-Biol. Interact. 2015, 231, 71–82. [Google Scholar] [CrossRef]
  58. Gorantla, J.N.; Nishanth Kumar, S.; Nisha, G.V.; Sumandu, A.S.; Dileep, C.; Sudaresan, A.; Sree Kumar, M.M.; Lankalapalli, R.S.; Dileep Kumar, B.S. Purification and characterization of antifungal phenazines from a fluorescent Pseudomonas strain FPO4 against medically important fungi. J. Mycol. Med. 2014, 24, 185–192. [Google Scholar] [CrossRef]
  59. Zhou, L.; Jiang, H.-X.; Sun, S.; Yang, D.-D.; Jin, K.-M.; Zhang, W.; He, Y.-W. Biotechnological potential of a rhizosphere Pseudomonas aeruginosa strain producing phenazine-1-carboxylic acid and phenazine-1-carboxamide. World J. Microbiol. Biotechnol. 2016, 32, 50. [Google Scholar] [CrossRef]
  60. George, E.; Kumar, S.N.; Jacob, J.; Bommasani, B.; Lankalapalli, R.S.; Morang, P.; Kumar, B.S.D. Characterization of the bioactive metabolites from a plant growth-promoting rhizobacteria and their exploitation as antimicrobial and plant growth-promoting agents. Appl. Biochem. Biotechnol. 2015, 176, 529–546. [Google Scholar] [CrossRef]
  61. Tupe, S.G.; Kulkarni, R.R.; Shirazi, F.; Sant, D.G.; Joshi, S.P.; Deshpande, M.V. Possible mechanism of antifungal phenazine-1-carboxamide from Pseudomonas sp. against dimorphic fungi Benjaminiella poitrasii and human pathogen Candida albicans. J. Appl. Microbiol. 2015, 118, 39–48. [Google Scholar] [CrossRef]
  62. Kennedy, R.K.; Veena, V.; Naik, P.R.; Lakshmi, P.; Krishna, R.; Sudharani, S.; Sakthivel, N. Phenazine-1-carboxamide (PCN) from Pseudomonas sp. strain PUP6 selectively induced apoptosis in lung (A549) and breast (MDA MB-231) cancer cells by inhibition of antiapoptotic Bcl-2 family proteins. Apoptosis 2015, 20, 858–868. [Google Scholar] [CrossRef]
  63. Ali, H.M.; El-Shikh, M.S.; Salem, M.Z.M.; Muzaheed, M. Isolation of bioactive phenazine-1-carboxamide from the soil bacterium Pantoea agglomerans and study of its anticancer potency on different cancer cell lines. J. AOAC Int. 2016, 99, 1233–1239. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, Y.; Wang, J.; Yang, N.; Wen, Z.; Sun, X.; Chai, Y.; Ma, Z. Wheat microbiome bacteria can reduce virulence of a plant pathogenic fungus by altering histone acetylation. Nat. Commun. 2018, 9, 3429. [Google Scholar] [CrossRef] [PubMed]
  65. Li, L.; Ran, T.; Zhu, H.; Yin, M.; Yu, W.; Zou, J.; Li, L.; Ye, Y.; Sun, H.; Wang, W.; et al. Molecular mechanism of Fusarium fungus inhibition by phenazine-1-carboxamide. J. Agric. Food Chem. 2024, 72, 15176–15189. [Google Scholar] [CrossRef] [PubMed]
  66. Zendah, I.; Riaz, N.; Nasr, H.; Frauendorf, H.; Schüffler, A.; Raies, A.; Laatsch, H. Chromophenazines from the terrestrial Streptomyces sp. Ank 315. J. Nat. Prod. 2012, 75, 2–8. [Google Scholar] [CrossRef] [PubMed]
  67. Hu, L.; Chen, X.; Han, L.; Zhao, L.; Miao, C.; Huang, X.; Chen, Y.; Li, P.; Li, Y. Two new phenazine metabolites with antimicrobial activities from soil-derived Streptomyces species. J. Antibiot. 2019, 72, 574–577. [Google Scholar] [CrossRef]
  68. Wu, C.; van Wezel, G.P.; Hae Choi, Y. Identification of novel endophenaside antibiotics produced by Kitasatospora sp. MBT66. J. Antibiot. 2015, 68, 445–452. [Google Scholar] [CrossRef]
  69. Saleh, O.; Flinspach, K.; Westrich, L.; Kulik, A.; Gust, B.; Fiedler, H.-P.; Heide, L. Mutational analysis of a phenazine biosynthetic gene cluster in Streptomyces anulatus 9663. Beilstein J. Org. Chem. 2012, 8, 501–513. [Google Scholar] [CrossRef]
  70. Khan, S.T.; Izumikawa, M.; Motohashi, K.; Mukai, A.; Takagi, M.; Shin-Ya, K. Distribution of the 3-hydroxyl-3-methylglutaryl coenzyme A reductase gene and isoprenoid production in marine-derived Actinobacteria. FEMS Microbiol. Lett. 2010, 304, 89–96. [Google Scholar] [CrossRef]
  71. Izumikawa, M.; Khan, S.T.; Takagi, M.; Shin-ya, K. Sponge-derived Streptomyces producing isoprenoids via the mevalonate pathway. J. Nat. Prod. 2010, 73, 208–212. [Google Scholar] [CrossRef]
  72. Song, Y.; Huang, H.; Chen, Y.; Ding, J.; Zhang, Y.; Sun, A.; Zhang, W.; Ju, J. Cytotoxic and antibacterial marfuraquinocins from the deep south China sea-derived Streptomyces niveus SCSIO 3406. J. Nat. Prod. 2013, 76, 2263–2268. [Google Scholar] [CrossRef]
  73. Li, S.; Hu, X.; Li, L.; Hu, X.; Wang, J.; Hu, X.; Liu, H.; Yu, L.; You, X.; Jiang, B.; et al. 1-Hydroxy-7-oxolavanducyanin and Δ7″,8″-6″-hydroxynaphthomevalin from Streptomyces sp. CPCC 203577. J. Antibiot. 2020, 73, 324–328. [Google Scholar] [CrossRef]
  74. Dutta, S.; Morang, P.; Nishanth Kumar, S.; Dileep Kumar, B.S. Fusarial wilt control and growth promotion of pigeon pea through bioactive metabolites produced by two plant growth promoting rhizobacteria. World J. Microb. Biotechnol. 2014, 30, 1111–1121. [Google Scholar] [CrossRef]
  75. La Ferla, B.; Airoldi, C.; Zona, C.; Orsato, A.; Cardona, F.; Merlo, S.; Sironi, E.; D’Orazio, G.; Nicotra, F. Natural glycoconjugates with antitumor activity. Nat. Prod. Rep. 2011, 28, 630–648. [Google Scholar] [CrossRef]
  76. Pratiwi, R.H.; Hidayat, I.; Hanafi, M.; Mangunwardoyo, W. Isolation and structure elucidation of phenazine derivative from Streptomyces sp. strain UICC B-92 isolated from Neesia altissima (Malvaceae). Iran J. Microbiol. 2020, 12, 127–137. [Google Scholar]
  77. Jansen, R.; Sood, S.; Huch, V.; Kunze, B.; Stadler, M.; Müller, R. Pyrronazols, metabolites from the Myxobacteria Nannocystis pusilla and N. exedens, are unusual chlorinated pyrone-oxazole-pyrroles. J. Nat. Prod. 2014, 77, 320–326. [Google Scholar] [CrossRef]
  78. Rusman, Y.; Oppegard, L.M.; Hiasa, H.; Gelbmann, C.; Salomon, C.E. Solphenazines A-F, glycosylated phenazines from Streptomyces sp. strain DL-93. J. Nat. Prod. 2013, 76, 91–96. [Google Scholar] [CrossRef]
  79. Wu, C.; Medema, M.H.; Läkamp, R.M.; Zhang, L.; Dorrestein, P.C.; Choi, Y.H.; van Wezel, G.P. Leucanicidin and endophenasides result from methyl-rhamnosylation by the same tailoring enzymes in Kitasatospora sp. MBT66. ACS Chem. Biol. 2015, 11, 478–490. [Google Scholar] [CrossRef]
  80. Iloabuchi, K.; Spiteller, D. Bacillus sp. G2112 detoxifies phenazine-1-carboxylic acid by N5 glucosylation. Molecules 2024, 29, 589. [Google Scholar] [CrossRef]
  81. Stupp, G.S.; von Reuss, S.H.; Izrayelit, Y.; Ajredini, R.; Schroeder, F.C.; Edison, A.S. Chemical detoxification of small molecules by Caenorhabditis elegans. ACS Chem. Biol. 2013, 8, 309–313. [Google Scholar] [CrossRef]
  82. Asif, M.Z.; Nocilla, K.A.; Ngo, L.; Shah, M.; Smadi, Y.; Hafeez, Z.; Parnes, M.; Manson, A.; Glushka, J.N.; Leach, F.E.; et al. Role of UDP-glycosyltransferase (ugt) genes in detoxification and glycosylation of 1-hydroxyphenazine (1-HP) in Caenorhabditis elegans. Chem. Res. Toxicol. 2024, 37, 590–599. [Google Scholar] [CrossRef]
  83. Conda-Sheridan, M.; Marler, L.; Park, E.-J.; Kondratyuk, T.P.; Jermihov, K.; Mesecar, A.D.; Pezzuto, J.M.; Asolkar, R.N.; Fenical, W.; Cushman, M. Potential chemopreventive agents based on the structure of the lead compound 2-bromo-1-hydroxyphenazine, isolated from Streptomyces species, strain CNS284. J. Med. Chem. 2010, 53, 8688–8699. [Google Scholar] [CrossRef]
  84. Asolkar, R.N.; Singh, A.; Jensen, P.R.; Aalbersberg, W.; Carté, B.K.; Feussner, K.-D.; Subramani, R.; DiPasquale, A.; Rheingold, A.L.; Fenical, W. Marinocyanins, cytotoxic bromo-phenazinone meroterpenoids from a marine bacterium from the Streptomycete clade MAR4. Tetrahedron 2017, 73, 2234–2241. [Google Scholar] [CrossRef]
  85. Xiao, T.; Liu, K.; Gao, Q.; Chen, M.; Kim, Y.S.; Jin, S.; Ding, Y.; Huigens, R.W. Design, Synthesis, and Evaluation of Carbonate-Linked Halogenated Phenazine-Quinone Prodrugs with Improved Water-Solubility and Potent Antibacterial Profiles. ACS Infect. Dis. 2023, 9, 899–915. [Google Scholar] [CrossRef]
  86. Yang, H.; Liu, K.; Jin, S.; Huigens Iii, R.W. Design, synthesis and biological evaluation of a halogenated phenazine-erythromycin conjugate prodrug for antibacterial applications. Org. Biomol. Chem. 2021, 19, 1483–1487. [Google Scholar] [CrossRef]
  87. Rui, Z.; Ye, M.; Wang, S.; Fujikawa, K.; Akerele, B.; Aung, M.; Floss, H.G.; Zhang, W.; Yu, T.W. Insights into a divergent phenazine biosynthetic pathway governed by a plasmid-born esmeraldin gene cluster. Chem. Biol. 2012, 19, 1116–1125. [Google Scholar] [CrossRef]
  88. Lee, H.-S.; Kang, J.S.; Cho, D.-Y.; Choi, D.-K.; Shin, H.J. Isolation, structure determination, and semisynthesis of diphenazine compounds from a deep-sea-derived strain of the fungus Cystobasidium laryngis and their biological activities. J. Nat. Prod. 2022, 85, 857–865. [Google Scholar] [CrossRef]
  89. Lee, H.-S.; Kang, J.S.; Choi, B.-K.; Lee, H.-S.; Lee, Y.-J.; Lee, J.; Shin, H.J. Phenazine derivatives with anti-inflammatory activity from the deep-Sea sediment-derived yeast-like fungus Cystobasidium laryngis IV17-028. Mar. Drugs 2019, 17, 482. [Google Scholar] [CrossRef]
  90. Abdelfattah, M.S.; Toume, K.; Ishibashi, M. Isolation and structure elucidation of izuminosides A-C: A rare phenazine glycosides from Streptomyces sp. IFM 11260. J. Antibiot. 2011, 64, 271–275. [Google Scholar] [CrossRef]
  91. Wagner, M.; Abdel-Mageed, W.M.; Ebel, R.; Bull, A.T.; Goodfellow, M.; Fiedler, H.-P.; Jaspars, M. Dermacozines H-J isolated from a deep-sea strain of Dermacoccus abyssi from Mariana Trench sediments. J. Nat. Prod. 2014, 77, 416–420. [Google Scholar] [CrossRef]
  92. Heine, D.; Sundaram, S.; Beudert, M.; Martin, K.; Hertweck, C. A widespread bacterial phenazine forms S-conjugates with biogenic thiols and crosslinks proteins. Chem. Sci. 2016, 7, 4848–4855. [Google Scholar] [CrossRef] [PubMed]
  93. Abdelfattah, M.S.; Kazufumi, T.; Ishibashi, M. Izumiphenazines A-C: Isolation and structure elucidation of phenazine derivatives from Streptomyces sp. IFM 11204. J. Nat. Prod. 2010, 73, 1999–2002. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Y.; Han, L.; Rong, H.; Li, L.; Zhao, L.; Wu, L.; Xu, L.; Jiang, Y.; Huang, X. Diastaphenazine, a new dimeric phenazine from an endophytic Streptomyces diastaticus subsp. ardesiacus. J. Antibiot. 2015, 68, 210–212. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, X.; Abbas, M.; Zhang, Y.; Elshahawi, S.I.; Ponomareva, L.V.; Cui, Z.; Van Lanen, S.G.; Sajid, I.; Voss, S.R.; Shaaban, K.A.; et al. Baraphenazines A-G, divergent fused phenazine-based metabolites from a Himalayan Streptomyces. J. Nat. Prod. 2019, 82, 1686–1693. [Google Scholar] [CrossRef] [PubMed]
  96. Ding, Z.-G.; Li, M.-G.; Ren, J.; Zhao, J.-Y.; Huang, R.; Wang, Q.-Z.; Cui, X.-L.; Zhu, H.-J.; Wen, M.-L. Phenazinolins A-E: Novel diphenazines from a tin mine tailings-derived Streptomyces species. Org. Biomol. Chem. 2011, 9, 2771–2776. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, Z.; Tang, W.; Jiang, T.; Xu, X.; Kong, K.; Shi, S.; Zhang, S.; Cao, W.; Zhang, Y. Structural characterization, derivatization and antibacterial activity of secondary metabolites produced by termite-associated Streptomyces showdoensis BYF17. Pest Manag. Sci. 2023, 79, 1800–1808. [Google Scholar] [CrossRef]
  98. Abdel-Mageed, W.M.; Milne, B.F.; Wagner, M.; Schumacher, M.; Sandor, P.; Pathom-aree, W.; Goodfellow, M.; Bull, A.T.; Horikoshi, K.; Ebel, R.; et al. Dermacozines, a new phenazine family from deep-sea dermacocci isolated from a Mariana Trench sediment. Org. Biomol. Chem. 2010, 8, 2352–2362. [Google Scholar] [CrossRef]
  99. Juhasz, B.; Pech-Puch, D.; Tabudravu, J.N.; Cautain, B.; Reyes, F.; Jiménez, C.; Kyeremeh, K.; Jaspars, M. Dermacozine N, the first natural linear pentacyclic oxazinophenazine with UV-Vis absorption maxima in the near infrared region, along with dermacozines O and P isolated from the Mariana Trench sediment strain Dermacoccus abyssi MT 1.1T. Mar. Drugs 2021, 19, 325. [Google Scholar] [CrossRef]
  100. Shi, Y.-M.; Brachmann, A.O.; Westphalen, M.A.; Neubacher, N.; Tobias, N.J.; Bode, H.B. Dual phenazine gene clusters enable diversification during biosynthesis. Nat. Chem. Biol. 2019, 15, 331–339. [Google Scholar] [CrossRef]
  101. Omura, S.; Eda, S.; Funayama, S.; Komiyama, K.; Takahashi, Y.; Woodruff, H.B. Studies on a novel antitumor antibiotic, phenazinomycin: Taxonomy, fermentation, isolation, and physicochemical and biological characteristics. J. Antibiot. 1989, 42, 1037–1042. [Google Scholar] [CrossRef]
  102. Yang, H.; Kundra, S.; Chojnacki, M.; Liu, K.; Fuse, M.A.; Abouelhassan, Y.; Kallifidas, D.; Zhang, P.; Huang, G.; Jin, S.; et al. A Modular Synthetic Route Involving N-Aryl-2-nitrosoaniline Intermediates Leads to a New Series of 3-Substituted Halogenated Phenazine Antibacterial Agents. J. Med. Chem. 2021, 64, 7275–7295. [Google Scholar] [CrossRef]
  103. Serafini, M.; Cargnin, S.; Massarotti, A.; Pirali, T.; Genazzani, A.A. Essential Medicinal Chemistry of Essential Medicines. J. Med. Chem. 2020, 63, 10170–10187. [Google Scholar] [CrossRef]
  104. Gopal, M.; Padayatchi, N.; Metcalfe, J.Z.; O’Donnell, M.R. Systematic review of clofazimine for the treatment of drug-resistant tuberculosis [Review article]. Int. J. Tuberc. Lung Dis. 2013, 17, 1001–1007. [Google Scholar] [CrossRef]
  105. Verborg, W.; Thomas, H.; Bissett, D.; Waterfall, J.; Steiner, J.; Cooper, M.; Rankin, E.M. First-into-man phase I and pharmacokinetic study of XR5944.14, a novel agent with a unique mechanism of action. Br. J. Cancer 2007, 97, 844–850. [Google Scholar] [CrossRef]
  106. de Jonge, M.J.A.; Kaye, S.; Verweij, J.; Brock, C.; Reade, S.; Scurr, M.; van Doorn, L.; Verheij, C.; Loos, W.; Brindley, C.; et al. Phase I and pharmacokinetic study of XR11576, an oral topoisomerase I and II inhibitor, administered on days 1–5 of a 3-weekly cycle in patients with advanced solid tumours. Br. J. Cancer 2004, 91, 1459–1465. [Google Scholar] [CrossRef]
  107. Tarui, M.; Doi, M.; Ishida, T.; Inoue, M.; Nakaike, S.; Kitamura, K. DNA-binding characterization of a novel anti-tumour benzo[a]phenazine derivative NC-182: Spectroscopic and viscometric studies. Biochem. J. 1994, 304, 271–279. [Google Scholar] [CrossRef]
  108. Samata, K.; Yamagishi, T.; Ichihara, T.; Nanaumi, K.; Ikeda, T.; Ikeya, H.; Kuraishi, A.; Nakaike, S.; Kashiwagi, K.; Igarashi, K. Establishment and characterization of a mouse FM3A cell mutant resistant to topoisomerase II-inhibitor NC-190. Cancer Chemother. Pharmacol. 2002, 50, 367–372. [Google Scholar] [CrossRef]
  109. Yamagishi, T.; Nakaike, S.; Ikeda, T.; Ikeya, H.; Otomo, S. A novel antitumor compound, NC-190, induces topoisomerase II-dependent DNA cleavage and DNA fragmentation. Cancer Chemother. Pharmacol. 1996, 38, 29–34. [Google Scholar] [CrossRef]
  110. Huang, W.; Wan, Y.; Su, H.; Zhang, Z.; Liu, Y.; Sadeeq, M.; Xian, M.; Feng, X.; Xiong, P.; Hou, F. Recent Advances in Phenazine Natural Products: Biosynthesis and Metabolic Engineering. J. Agric. Food Chem. 2024, 72, 21364–21379. [Google Scholar] [CrossRef]
  111. Valentine-King, M.A.; Cisneros, K.; James, M.O.; Huigens, R.W., 3rd; Brown, M.B. Efficacy data of halogenated phenazine and quinoline agents and an NH125 analogue to veterinary mycoplasmas. BMC Vet. Res. 2020, 16, 107. [Google Scholar] [CrossRef]
Figure 1. The chemical structures of clofazimine, XR5944, XR11576, NC-190, and NC-182.
Figure 1. The chemical structures of clofazimine, XR5944, XR11576, NC-190, and NC-182.
Molecules 29 04771 g001
Figure 2. The chemical structures of compounds phenazine, 18, 03, and 08.
Figure 2. The chemical structures of compounds phenazine, 18, 03, and 08.
Molecules 29 04771 g002
Figure 3. The chemical structures of N-oxide and N-methylated phenazines.
Figure 3. The chemical structures of N-oxide and N-methylated phenazines.
Molecules 29 04771 g003
Figure 4. The chemical structures of compounds 2125.
Figure 4. The chemical structures of compounds 2125.
Molecules 29 04771 g004
Figure 5. The chemical structures of terpenoid phenazines.
Figure 5. The chemical structures of terpenoid phenazines.
Molecules 29 04771 g005
Figure 6. The chemical structures of glycosylated phenazines: (A) novel glycosylated phenazine natural products. (B) 2′-O-methylated/2′-O-unmethylated rhamnose glycosylated phenazines and two tautomeric glycerides naturally occurring phenazines.
Figure 6. The chemical structures of glycosylated phenazines: (A) novel glycosylated phenazine natural products. (B) 2′-O-methylated/2′-O-unmethylated rhamnose glycosylated phenazines and two tautomeric glycerides naturally occurring phenazines.
Molecules 29 04771 g006
Figure 7. The chemical structures of halogenated phenazine natural products.
Figure 7. The chemical structures of halogenated phenazine natural products.
Molecules 29 04771 g007
Figure 8. The chemical structures of saphenic acid derivatives.
Figure 8. The chemical structures of saphenic acid derivatives.
Molecules 29 04771 g008
Figure 9. The chemical structures of phenazines with sulfur.
Figure 9. The chemical structures of phenazines with sulfur.
Molecules 29 04771 g009
Figure 10. The chemical structures of izumiphenazines and analogs.
Figure 10. The chemical structures of izumiphenazines and analogs.
Molecules 29 04771 g010
Figure 11. The chemical structures of baraphenazines.
Figure 11. The chemical structures of baraphenazines.
Molecules 29 04771 g011
Figure 12. The chemical structures of phenazinolins.
Figure 12. The chemical structures of phenazinolins.
Molecules 29 04771 g012
Figure 13. The chemical structures of dermacozines.
Figure 13. The chemical structures of dermacozines.
Molecules 29 04771 g013
Figure 14. The chemical structures of compounds 135147.
Figure 14. The chemical structures of compounds 135147.
Molecules 29 04771 g014
Table 1. Biological activities of phenazines in this review.
Table 1. Biological activities of phenazines in this review.
Substance Class Biological Activities Compounds (Refs)
Simple derivatives Antifungal activity1 [19,20,21,22,23,24,25,26]
Antibacterial activity2 [28,29,30,31,32]
Cytotoxic activity2 [30]
Phenazines with hydroxyl and methoxyl moietiesAntibacterial activity39, 03, 08 [34,35,36,37,38,39,41]
Antifungal activity37, 10 [34,35,36,37,38,39,42]
Antitumor activity37 [34,35,36,37,38,39]
N-oxide phenazinesAntimicrobial activity11, 12 [41,42,43]
Antifungal activity19 [45,46]
Cytotoxic activity11, 12, 17 [47,48,49,50]
N-methylated phenazinesAntibacterial activity20 [53,54,55]
Antitumor activity5MPCA, 20 [57]
Antifungal activity20 [53,54,55]
Carboxamidated phenazinesAntifungal activity2123 [61,62,64,65]
Antitumor activity 21 [63]
Antibacterial activity22, 23 [67]
Terpenoid phenazinesAntibacterial activity2931, 42, 43 [68,73]
Cytotoxic activity42, 43 [73]
Antifungal activity48, 49 [74]
Glycosylated phenazinesAntibacterial activity4446 [68], 5052 [76]
Cytotoxic activity54, 55, 59 [78]
Halogenated phenazinesCytotoxic activity7884 [83,84]
Antimicrobial activity7984 [84]
Saphenic acid derivativesAntineuroinflammatory activity100 [88]
Cytotoxic activity100 [88]
Phenazines with sulfurRadical scavenging activity101 [91]
Special derivativesAntitumor activity104106 [93]
Antifungal activity107108, 114, 117119 [42,95,96,97]
Antibacterial activity109, 114, 117119 [95,96,97]
Cytotoxic activity117119, 122134 [96,97,98,99]
Radical scavenging activity124 [99]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huang, W.; Wan, Y.; Zhang, S.; Wang, C.; Zhang, Z.; Su, H.; Xiong, P.; Hou, F. Recent Advances in Phenazine Natural Products: Chemical Structures and Biological Activities. Molecules 2024, 29, 4771. https://doi.org/10.3390/molecules29194771

AMA Style

Huang W, Wan Y, Zhang S, Wang C, Zhang Z, Su H, Xiong P, Hou F. Recent Advances in Phenazine Natural Products: Chemical Structures and Biological Activities. Molecules. 2024; 29(19):4771. https://doi.org/10.3390/molecules29194771

Chicago/Turabian Style

Huang, Wei, Yupeng Wan, Shuo Zhang, Chaozhi Wang, Zhe Zhang, Huai Su, Peng Xiong, and Feifei Hou. 2024. "Recent Advances in Phenazine Natural Products: Chemical Structures and Biological Activities" Molecules 29, no. 19: 4771. https://doi.org/10.3390/molecules29194771

APA Style

Huang, W., Wan, Y., Zhang, S., Wang, C., Zhang, Z., Su, H., Xiong, P., & Hou, F. (2024). Recent Advances in Phenazine Natural Products: Chemical Structures and Biological Activities. Molecules, 29(19), 4771. https://doi.org/10.3390/molecules29194771

Article Metrics

Back to TopTop