Next Article in Journal
Striated Muscle Evaluation Based on Velocity and Amortization Ratio of Mechanical Impulse Propagation in Simulated Microgravity Environment
Next Article in Special Issue
A Review of Pseudomonas aeruginosa Metallophores: Pyoverdine, Pyochelin and Pseudopaline
Previous Article in Journal
Temperature and Humidity Regulate Sporulation of Corynespora cassiicola That Is Associated with Pathogenicity in Cucumber (Cucumis sativus L.)
Previous Article in Special Issue
Osmotic Gradient Is a Factor That Influences the Gill Microbiota Communities in Oryzias melastigma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antibiotics from Insect-Associated Actinobacteria

by
Anna A. Baranova
1,2,
Yuliya V. Zakalyukina
3,
Anna A. Ovcharenko
1,4,
Vladimir A. Korshun
1 and
Anton P. Tyurin
1,*
1
Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
2
Gause Institute of New Antibiotics, Bol’shaya Pirogovskaya 11, 119021 Moscow, Russia
3
Department of Soil Science, Lomonosov Moscow State University, Leninskie Gory 1-12, 119991 Moscow, Russia
4
Higher Chemical College RAS, Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
*
Author to whom correspondence should be addressed.
Biology 2022, 11(11), 1676; https://doi.org/10.3390/biology11111676
Submission received: 26 October 2022 / Revised: 13 November 2022 / Accepted: 16 November 2022 / Published: 18 November 2022

Abstract

:

Simple Summary

Actinobacteria remain a key source for antibiotic discovery, and the current antimicrobial resistance crisis is becoming a driving force for actinobacteria research. Insect-associated actinomycetes are an underexplored ecological niche with prospects for the search for novel antimicrobial compounds. The described associations of leaf-cutter ants and Pseudonocardia bacteria or solitary wasps and Streptomyces bacteria were the first examples of mutually beneficial coexistence of insects with actinobacteria. On the molecular level, these systems are regulated by antibiotics. The complex relationships between insects and actinobacteria mediated by antibiotics could be important for the stability of ecosystems and agricultural production.

Abstract

Actinobacteria are involved into multilateral relationships between insects, their food sources, infectious agents, etc. Antibiotics and related natural products play an essential role in such systems. The literature from the January 2016–August 2022 period devoted to insect-associated actinomycetes with antagonistic and/or enzyme-inhibiting activity was selected. Recent progress in multidisciplinary studies of insect–actinobacterial interactions mediated by antibiotics is summarized and discussed.

Graphical Abstract

1. Introduction

Insects are the most widespread animals on our planet, playing an essential role in different ecosystems. Pollination, plant biomass destruction and the spread of tick-borne encephalitis and malaria—all these processes are impossible without insects. According to a rough evaluation by Bar-On et al. [1], about 50% of animal biomass on Earth is comprised of arthropods. Approximately one sixth of this huge number is insects (0.2 Gt of 1.2 Gt) [2].
In spite of such ubiquity and diversity, a dangerous trend in insect populations has been detected. In the 2010s, numerous studies reporting significant declines in insect populations emerged. This “decline crisis” is associated mostly with a reduction in numbers, although, in some cases, entire species have become extinct [3,4,5,6]. Globally, terrestrial insects are declining by about 11% per decade, while freshwater insects are increasing by 12% per decade, according to a meta-analysis published in Science [7]. However, this relatively fast change may threaten the stability of ecosystems and agricultural production.
The factors influencing insect survival should be carefully analyzed, including their interaction with microorganisms. Symbiotic bacterial species strongly associated with insects, such as Wolbachia sp., have a long history of study and a great potential for the regulation of insect populations [8]. Coexistence of the macro- and microorganisms is possible through close interaction and intensive signal exchange.
Chemical signaling is a very important factor for insects. Low-molecular compounds mediate insect interactions at various levels, from the individual level, where hormones control development and reproduction, to the interspecies level, where specific metabolites act as defense agents against predators [9,10,11,12]. Unsurprisingly, actinomycetes—one of the most fruitful producers of diverse secondary metabolites [13,14,15]—can interact with insects by biosynthesis of small molecules.
For example, Noodwell and co-workers [16] showed that small concentrations of volatile terpenes produced by streptomycetes attract Drosophila melanogaster. Fruit flies preferentially deposit their eggs on bacteria-contaminated food sources. As a result, the larvae are killed by antibiotics (cosmomycin or avermectin), which are produced by streptomycetes. This “toxic snare” could seriously affect insect populations. Nevertheless, this mechanism of ecological interaction has only recently been established. A similar attractive effect of volatile terpenes was observed for red imported fire ants [17], but, in that case, no toxic effects were detected. On the contrary, the authors suggest that the choice of Streptomyces- and Nocardiopsis-rich environments reduces the mortality of young ants from entomopathogenic fungi.
Actinobacteria remain a key source for antibiotic discovery [13], and the current antimicrobial resistance crisis is becoming a driving force for actinobacteria research. However, insect-associated actinomycetes are not just an underexplored ecological niche with prospects for the search for novel antimicrobial scaffolds [18]; they could also be important for understanding the complex relationships between different types of organisms which define the actual state and further development of ecosystems.

2. Methodology

In this account we make an attempt to summarize recent progress in the study of molecular ecology of insect-actinobacterial interactions mediated by antibiotics.
The search and preliminary selection of sources was performed using a Web of Science, Google Scholar, and PubMed search for keyword combinations (“insect-associated”, “actinobacteria/actinomycete”, “antibiotic/antimicrobial/inhibition”). The inclusion criteria were:
  • The actinobacteria (Phylum: Actinomycetota) have been isolated directly from insects (Class: Insecta) (whole animals, cuticle, gut or other internal/external organs) or from freshly sampled secreted substances. Ant nest fragments and other environmental samples were excluded;
  • The antibiotic compounds (i.e., having some antagonistic or enzyme-inhibition activity) produced by actinomycetes have been described or, at least, antagonistic action was confirmed by bioassay;
  • Only regular articles, reviews and book chapters were included. Patents, conference materials and preprints were excluded.
It should be noted that the insect-associated microbiome as a source of novel natural products was a subject of several other reviews. The discussion of insect–actinobacterial symbiotic relationships and their relevance for drug discovery started from comments by M. Kaltenpoth [19] and H. B. Bode [20]. During the next decade (2010s), this topic was highlighted and/or reviewed in several works [3,21,22,23,24,25,26,27].
The latest systematic review on natural products from insect-associated microorganisms was published by C. Beemelmanns et al. [28] in 2016. Despite it not being focused on a specific genus and type of compounds, we could recognize it as a starting point for our literature selection: the current review is covering January 2016–August 2022. From the latest years we should highlight some works on specific types of insects or ecological niches: fungus-growing ants [29], African edible insects [30], neotropical insects [31] and fungus-farming termites [32]. A critical review on microbial symbionts of insects as a source of new antimicrobials from our Belgian colleagues [18] is the most relevant and closest to our work, but it does not have a systematic character and is not focused on actinomycetes as a major source of antimicrobials.
Using the above-mentioned criteria and methodology, we identified 71 research papers from 2016 [26,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102].
A total of 14 other works were excluded due to the source of actinobacteria isolation (insect-associated environments such as dung beetle’s brood ball, wasp nest, termite fungus comb, bee pollen, etc.—see List S2). The most important information from the selected materials is summarized in Table S1. Here, we present our comments and conclusions based on analysis of the data.

3. General Remarks: Data Pre-Processing

All the presented studies have the same experimental design, which can be illustrated by Scheme 1:
First of all, in some works (11 out of 71), this workflow is not complete: no compounds were isolated and characterized, only antagonistic activity of actinomycete strains was detected. In others, we could find repeated data in strain descriptions (marked as “previously described”, 10 out of 60), which means that new compounds were isolated from the same strains. This could be interpreted as evidence for the high biosynthetic potential of actinobacteria. Other missing values (marked as “no data”) are caused by an absence of experimental details in the original works.

4. Data Analysis: Main Trends

The geographical distribution of sample collection locations is depicted on Figure 1. We could see a clustering of sampling locations—samples were often collected in compact regions: Central America, South Brazil, South Africa, East China, North-East China and South Korea. On the contrary, it is easy to see that most of the world area is not involved in insect–actinobacteria symbiosis studies.
The collected insects could be classified into major taxonomic categories (order, family) as shown in Figure 2. The most studied insect phylum, by a wide margin, was ants (Hymenoptera: Formicidae). We also have to note termites (Blattodea: Termitidae and Rhinotermitidae), bees (Hymenoptera: Apidae), Silphidae beetles (Coleoptera: Silphidae) and grasshoppers (Orthoptera: Acrididae). Unfortunately, in three cases [36,61,96], the taxonomic status of the insects was not determined.
Only the classical isolation approach was used in the selected papers. The most used isolation media were chitin agar (C/N-source—chitin) and Gause agar №1 (C-source—soluble starch, N-source—ammonium salts) (see Table S2 and Figure S1). These are well-known selective media for actinobacteria isolation.
Streptomyces was the most isolated genus (see Figure 3); it is typical for all culture-dependent studies of actinobacteria. From non-streptomycete actinobacterial genera, the Pseudonocardia—a well-studied symbiont of Attini ants—was the most mentioned. New species (8, from which 2 are synonymic) were described: 6 Streptomyces, 1 Amycolatopsis and 1 from genus Actinomadura.
Here, we grouped the isolated compounds by their biosynthetic origin: peptides, polyketides, alkaloids and other. Class and structure assignment is summarized in SI, Schemes S1–S4. For all schemes (here and in SI), new elucidated structures are colored black, while known metabolites are colored gray. The main statistics on the isolated metabolites are presented in Figure 4.
Polyketides are the leading class among described antibiotics (about half of the total). However, more importantly, structural novelty is quite high for all main classes of compounds.
Despite a significant number of publications and detected metabolites, the ecology of the isolated compounds is discussed in just 15 out of 71 papers. In all cases, it is protective symbiosis, but these symbiotic relationships could be divided into two main types: protection of food sources (for fungus-growing insects) and direct protection against entomopatogenic fungi and bacteria (Figure 5).
Food source protection is described extensively for fungus-growing termites and ants (Attini tribe). The fungal cultivar is an important food source for these social insects with specialized diets, and the presence of pathogenic fungal species (Escovopsis sp. [103] for ants and Pseudoxylaria sp. for termites [32]) is a critical factor for colony survival. Compounds selectively inhibiting fungal pathogens and other non-specialized pathogens while being neutral to fungal cultivar are widely represented in the biosynthetic arsenal of actinobacteria.
Examples of antibiotics used by insects for food source protection are summarized on Scheme 2. The majority of the structures are associated with leaf-cutter ants (and their actinobacterial symbionts). We should highlight some newly described antibiotics: an unusual non-polyene macrolide [104] cyphomycin (77) [44,69] and peptide antibiotics attinimicin (23) [38] and dentigerumycin F (8) [48].
Another fruitful source for isolation of the above type of antibiotics is termites. Polyketides macrotermicin A (40) and C (47) from termite-associated Amycolatopsis sp. M39 [90] active against parasitic Pseudoxylaria sp. Polyenes (74) and (75) were isolated from termite-associated Streptomyces sp. HF10 [42]. Both compounds have higher activity against pathogenic fungi (Xylaria sp. and Metarhizium anisopliae) than cultivar Termitomyces.
Only one example of food source protection is described for a different type of insects: bacteria Streptomyces griseus XylebKG-1 isolated from bark beetle Xyleborus saxesenii produce cycloheximide (81). This well-known antifungal inhibits the growth of parasitic fungus Nectria sp., but not of mutualistic Raffaelea sulphurea.
However, the list of such substances could not be complete without well-known inorganic compounds: sulphur and ammonia. Elemental sulfur (S8) produced by Streptomyces chartreusis strain ICBG323 [64] had antifungal activity against Escovopsis sp. Streptomyces sp. Av25_4, and other isolated actinobacterial species overproduce ammonia (up to 8 mM), which completely inhibits the growth of Escovopsis weberi due to strong basic pH [39]. These findings clearly indicate that not just complex antibiotics, but also simple inorganics excreted by symbiotic bacteria, are important for insect ecology.
Antibiotics utilized by insects as self-protection agents are shown in Scheme 3.
Well-known cytotoxic dipeptide antimycin A (19) was isolated from ant-associated Streptomyces albidoflavus A10. At low concentrations, it inhibits the growth of entomopathogenic fungi (Conidiobolus coronatus, Beauveria bassiana and others) [51]. Ionophore antibiotics (84–88) isolated from wasp-associated Streptomyces sp. M54 were effective against Hirsutella citriformis. This fungus is a natural enemy of the host—the wasp Polybia plebeja [45]. Peptides meliponamycins (15, 16) [56], glycosylated antibiotics lobophorins (152153) and polycyclic antibiotics (164165) [65] were isolated from bee-associated actinobacteria. All these compounds exhibited high activity against a bee-specialized pathogen—Paenibacillus larvae, the causative agent of American Foulbrood.
Thiopeptide antibiotic GE37468 (17) (Scheme 4) from Pseudonocardia sp., associated with ant Trachymyrmex septentrionalis, was described as a selective inhibitor of other ant-associated Pseudonocardia strains [54]. Its niche-defense role is an interesting phenomenon which opens up a new dimension in insect–actinobacteria symbiosis research.

5. Conclusions and Outlook

In the current review, we identified the main trends and problems of relevance to insect–actinomycete interaction research. Actinobacterial species are likely involved into multilateral relationships between insects, their food sources, infectious agents, predators, etc. Antibiotics and related natural products play an essential role in such systems.
Our brief analysis clearly indicates that we are only at the very beginning of the path leading to an understanding of the actual diversity of the ecological relationships between actinomycetes and insects. To fill the gaps in knowledge, we need more research data. First, efforts should be made to expand the range of studied insect hosts, with particular attention paid to species whose life cycle is associated with soil, plants, decaying organic debris and other substrates rich in microorganisms. It is noteworthy that various stages of the insect life cycle should be considered as distinct objects of study, since the ecology of larvae and imagines in many species is fundamentally different (lifestyle, habitat, diet etc.).
Traditional culture-dependent methods continue to be the main way of mining for active strains. The efficiency of actinobacteria isolation could be improved using innovative isolation techniques such ichip [105]. At the same time, the study of the metagenome of microbial communities associated with insects can provide a significant amount of valuable information. However, active antibiotic producers are often minor constituents of communities and their role may be underestimated.
Understanding insect–actinobacteria systems at the molecular level requires complex, multidisciplinary approaches. Proper chemical characterization of active metabolites and appropriate bioactivity assays should become the base of ecological research in this area. Insect ecology remains unclear without works at the intersection of microbiology, chemistry and animal studies.
We have found that the potential of this area is much greater than just an exotic source of compounds. The creation of new methods and agents for the prevention of insect-borne infectious diseases, pest control and other related areas may be stimulated by the study of insect-associated actinomycetes and their secondary metabolites.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biology11111676/s1, Scheme S1: Peptide antibiotics; Scheme S2: Polyketide antibiotics; Scheme S3: Alkaloid antibiotics; Scheme S4: Other antibiotics; Table S1: Selected publications; Table S2: Media used for actinobacteria isolation; Figure S1: Media used for actinobacteria isolation; List S1: References for Tables S1 and S2; List S2: Excluded publications.

Author Contributions

Conceptualization, Y.V.Z., V.A.K. and A.P.T.; methodology, A.P.T.; data collection, curation and formal analysis, all authors; writing—original draft preparation, A.A.B. and A.P.T.; writing—review and editing, all authors; visualization, A.A.B. and Y.V.Z.; supervision, A.P.T.; project administration and funding acquisition, V.A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Science and Higher Education of the Russian Federation (Agreement No 075–15-2021-1049).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All datasets generated or analyzed during the current study are openly available in the Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Bar-On, Y.M.; Phillips, R.; Milo, R. The Biomass Distribution on Earth. Proc. Natl. Acad. Sci. USA 2018, 115, 6506–6511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Eggleton, P. The State of the World’s Insects. Annu. Rev. Environ. Resour. 2020, 45, 61–82. [Google Scholar] [CrossRef]
  3. Forister, M.L.; Jahner, J.P.; Casner, K.L.; Wilson, J.S.; Shapiro, A.M. The Race Is Not to the Swift: Long-Term Data Reveal Pervasive Declines in California’s Low-Elevation Butterfly Fauna. Ecology 2011, 92, 2222–2235. [Google Scholar] [CrossRef]
  4. Valtonen, A.; Hirka, A.; Szőcs, L.; Ayres, M.P.; Roininen, H.; Csóka, G. Long-Term Species Loss and Homogenization of Moth Communities in Central Europe. J. Anim. Ecol. 2017, 86, 730–738. [Google Scholar] [CrossRef] [Green Version]
  5. Hallmann, C.A.; Sorg, M.; Jongejans, E.; Siepel, H.; Hofland, N.; Schwan, H.; Stenmans, W.; Müller, A.; Sumser, H.; Hörren, T.; et al. More than 75 Percent Decline over 27 Years in Total Flying Insect Biomass in Protected Areas. PLoS ONE 2017, 12, e0185809. [Google Scholar] [CrossRef] [Green Version]
  6. Sánchez-Bayo, F.; Wyckhuys, K.A.G. Worldwide Decline of the Entomofauna: A Review of Its Drivers. Biol. Conserv. 2019, 232, 8–27. [Google Scholar] [CrossRef]
  7. van Klink, R.; Bowler, D.E.; Gongalsky, K.B.; Swengel, A.B.; Gentile, A.; Chase, J.M. Meta-Analysis Reveals Declines in Terrestrial but Increases in Freshwater Insect Abundances. Science 2020, 368, 417–420. [Google Scholar] [CrossRef]
  8. Kaur, R.; Shropshire, J.D.; Cross, K.L.; Leigh, B.; Mansueto, A.J.; Stewart, V.; Bordenstein, S.R.; Bordenstein, S.R. Living in the Endosymbiotic World of Wolbachia: A Centennial Review. Cell Host Microbe 2021, 29, 879–893. [Google Scholar] [CrossRef] [PubMed]
  9. Wilson, E.O. Chemical Communication in the Social Insects: Insect Societies Are Organized Principally by Complex Systems of Chemical Signals. Science 1965, 149, 1064–1071. [Google Scholar] [CrossRef]
  10. Hoffmann, K.H.; Dettner, K.; Tomaschko, K. Chemical Signals in Insects and Other Arthropods: From Molecular Structure to Physiological Functions. Physiol. Biochem. Zool. 2006, 79, 344–356. [Google Scholar] [CrossRef]
  11. de Bruyne, M.; Baker, T.C. Odor Detection in Insects: Volatile Codes. J. Chem. Ecol. 2008, 34, 882–897. [Google Scholar] [CrossRef]
  12. Klowden, M.J.; Palli, S.R. Communication Systems. In Physiological Systems in Insects; Elsevier: Amsterdam, The Netherlands, 2022; pp. 607–653. ISBN 978-0-12-820359-0. [Google Scholar]
  13. Genilloud, O. Actinomycetes: Still a Source of Novel Antibiotics. Nat. Prod. Rep. 2017, 34, 1203–1232. [Google Scholar] [CrossRef] [PubMed]
  14. Hamedi, J.; Poorinmohammad, N.; Wink, J. The Role of Actinobacteria in Biotechnology. In Biology and Biotechnology of Actinobacteria; Wink, J., Mohammadipanah, F., Hamedi, J., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 269–328. ISBN 978-3-319-60338-4. [Google Scholar]
  15. Ossai, J.; Khatabi, B.; Nybo, S.E.; Kharel, M.K. Renewed Interests in the Discovery of Bioactive Actinomycete Metabolites Driven by Emerging Technologies. J. Appl. Microbiol. 2022, 132, 59–77. [Google Scholar] [CrossRef] [PubMed]
  16. Ho, L.K.; Daniel-Ivad, M.; Jeedigunta, S.P.; Li, J.; Iliadi, K.G.; Boulianne, G.L.; Hurd, T.R.; Smibert, C.A.; Nodwell, J.R. Chemical Entrapment and Killing of Insects by Bacteria. Nat. Commun. 2020, 11, 4608. [Google Scholar] [CrossRef]
  17. Huang, H.; Ren, L.; Li, H.; Schmidt, A.; Gershenzon, J.; Lu, Y.; Cheng, D. The Nesting Preference of an Invasive Ant Is Associated with the Cues Produced by Actinobacteria in Soil. PLoS Pathog. 2020, 16, e1008800. [Google Scholar] [CrossRef]
  18. Van Moll, L.; De Smet, J.; Cos, P.; Van Campenhout, L. Microbial Symbionts of Insects as a Source of New Antimicrobials: A Review. Crit. Rev. Microbiol. 2021, 47, 562–579. [Google Scholar] [CrossRef]
  19. Kaltenpoth, M. Actinobacteria as Mutualists: General Healthcare for Insects? Trends Microbiol. 2009, 17, 529–535. [Google Scholar] [CrossRef] [PubMed]
  20. Bode, H.B. Insects: True Pioneers in Anti-Infective Therapy and What We Can Learn from Them. Angew. Chem. Int. Ed. 2009, 48, 6394–6396. [Google Scholar] [CrossRef]
  21. 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]
  22. Klassen, J.L. Microbial Secondary Metabolites and Their Impacts on Insect Symbioses. Curr. Opin. Insect Sci. 2014, 4, 15–22. [Google Scholar] [CrossRef]
  23. Kaltenpoth, M.; Engl, T. Defensive Microbial Symbionts in Hymenoptera. Funct. Ecol. 2014, 28, 315–327. [Google Scholar] [CrossRef]
  24. Kurtböke, D.İ.; French, J.R.J.; Hayes, R.A.; Quinn, R.J. Eco-Taxonomic Insights into Actinomycete Symbionts of Termites for Discovery of Novel Bioactive Compounds. In Biotechnological Applications of Biodiversity; Mukherjee, J., Ed.; Advances in Biochemical Engineering/Biotechnology; Springer: Berlin/Heidelberg, Germany, 2014; Volume 147, pp. 111–135. ISBN 978-3-662-45096-3. [Google Scholar]
  25. Berasategui, A.; Shukla, S.; Salem, H.; Kaltenpoth, M. Potential Applications of Insect Symbionts in Biotechnology. Appl. Microbiol. Biotechnol. 2016, 100, 1567–1577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Van Arnam, E.B.; Ruzzini, A.C.; Sit, C.S.; Horn, H.; Pinto-Tomás, 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] [PubMed] [Green Version]
  27. Worsley, S.F.; Innocent, T.M.; Heine, D.; Murrell, J.C.; Yu, D.W.; Wilkinson, B.; Hutchings, M.I.; Boomsma, J.J.; Holmes, N.A. Symbiotic Partnerships and Their Chemical Interactions in the Leafcutter Ants (Hymenoptera: Formicidae). Myrmecol. News 2018, 27, 59–74. [Google Scholar] [CrossRef]
  28. Beemelmanns, C.; Guo, H.; Rischer, M.; Poulsen, M. Natural Products from Microbes Associated with Insects. Beilstein J. Org. Chem. 2016, 12, 314–327. [Google Scholar] [CrossRef] [Green Version]
  29. Batey, S.F.D.; Greco, C.; Hutchings, M.I.; Wilkinson, B. Chemical Warfare between Fungus-Growing Ants and Their Pathogens. Curr. Opin. Chem. Biol. 2020, 59, 172–181. [Google Scholar] [CrossRef]
  30. Mudalungu, C.M.; Tanga, C.M.; Kelemu, S.; Torto, B. An Overview of Antimicrobial Compounds from African Edible Insects and Their Associated Microbiota. Antibiotics 2021, 10, 621. [Google Scholar] [CrossRef]
  31. Menegatti, C.; Fukuda, T.T.H.; Pupo, M.T. Chemical Ecology in Insect-Microbe Interactions in the Neotropics. Planta Med. 2021, 87, 38–48. [Google Scholar] [CrossRef]
  32. Schmidt, S.; Kildgaard, S.; Guo, H.; Beemelmanns, C.; Poulsen, M. The Chemical Ecology of the Fungus-Farming Termite Symbiosis. Nat. Prod. Rep. 2022, 39, 231–248. [Google Scholar] [CrossRef]
  33. Zakalyukina, Y.V.; Osterman, I.A.; Wolf, J.; Neumann-Schaal, M.; Nouioui, I.; Biryukov, M.V. Amycolatopsis camponoti sp. nov., New Tetracenomycin-Producing Actinomycete Isolated from Carpenter Ant Camponotus vagus. Antonie Leeuwenhoek 2022, 115, 533–544. [Google Scholar] [CrossRef]
  34. An, J.S.; Lim, H.-J.; Lee, J.Y.; Jang, Y.-J.; Nam, S.-J.; Lee, S.K.; Oh, D.-C. Hamuramicin C, a Cytotoxic Bicyclic Macrolide Isolated from a Wasp Gut Bacterium. J. Nat. Prod. 2022, 85, 936–942. [Google Scholar] [CrossRef] [PubMed]
  35. Wu, Y.; Liu, Y.; Yu, J.; Xu, Y.; Chen, S. Observation of the Antimicrobial Activities of Two Actinomycetes in the Harvester Ant Messor orientalis. Insects 2022, 13, 691. [Google Scholar] [CrossRef] [PubMed]
  36. Wu, W.; Chen, B.; Zhang, S.; Guo, Z.; Li, J. Isolation and Identification of Symbiotic Actinomycetes from Termites in Hainan and Their Activity Against Tropical Plants Pathogenic Fungi. Chin. J. Trop. Crops 2022, 43, 1240–1247. [Google Scholar] [CrossRef]
  37. Lee, S.R.; Schalk, F.; Schwitalla, J.W.; Guo, H.; Yu, J.S.; Song, M.; Jung, W.H.; de Beer, Z.W.; Beemelmanns, C.; Kim, K.H. GNPS-Guided Discovery of Madurastatin Siderophores from the Termite-Associated Actinomadura sp. RB99**. Chem. Eur. J. 2022, 28, e202200612. [Google Scholar] [CrossRef] [PubMed]
  38. Promnuan, Y.; Promsai, S.; Pathom-aree, W.; Meelai, S. Apis andreniformis Associated Actinomycetes Show Antimicrobial Activity against Black Rot Pathogen (Xanthomonas campestris pv. Campestris). PeerJ 2021, 9, e12097. [Google Scholar] [CrossRef]
  39. 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]
  40. Shin, Y.-H.; Ban, Y.H.; Shin, J.; Park, I.W.; Yoon, S.; Ko, K.; Shin, J.; Nam, S.-J.; Winter, J.M.; Kim, Y.; et al. Azetidine-Bearing Non-Ribosomal Peptides, Bonnevillamides D and E, Isolated from a Carrion Beetle-Associated Actinomycete. J. Org. Chem. 2021, 86, 11149–11159. [Google Scholar] [CrossRef]
  41. Shin, Y.-H.; Ban, Y.H.; Kim, T.H.; Bae, E.S.; Shin, J.; Lee, S.K.; Jang, J.; Yoon, Y.J.; Oh, D.-C. Structures and Biosynthetic Pathway of Coprisamides C and D, 2-Alkenylcinnamic Acid-Containing Peptides from the Gut Bacterium of the Carrion Beetle Silpha perforata. J. Nat. Prod. 2021, 84, 239–246. [Google Scholar] [CrossRef]
  42. Li, J.; Sang, M.; Jiang, Y.; Wei, J.; Shen, Y.; Huang, Q.; Li, Y.; Ni, J. Polyene-Producing Streptomyces Spp. From the Fungus-Growing Termite Macrotermes barneyi Exhibit High Inhibitory Activity Against the Antagonistic Fungus Xylaria. Front. Microbiol. 2021, 12, 649962. [Google Scholar] [CrossRef]
  43. Xu, Y.; Xie, J.; Wu, W.C.; Chen, B.T.; Zhang, S.Q.; Wang, R.; Huang, J.; Guo, Z.K. Discovery of an Unprecedented Benz[a]Anthraquinone-Type Heterodimer from a Rare Actinomycete Amycolatopsis sp. HCa1. Fitoterapia 2021, 155, 105039. [Google Scholar] [CrossRef]
  44. Ortega, H.E.; Lourenzon, V.B.; Chevrette, M.G.; Ferreira, L.L.G.; Alvarenga, R.F.R.; Melo, W.G.P.; Venâncio, T.; Currie, C.R.; Andricopulo, A.D.; Bugni, T.S.; et al. Antileishmanial Macrolides from Ant-Associated Streptomyces sp. ISID311. Bioorg. Med. Chem. 2021, 32, 116016. [Google Scholar] [CrossRef] [PubMed]
  45. Matarrita-Carranza, B.; Murillo-Cruz, C.; Avendaño, R.; Ríos, M.I.; Chavarría, M.; Gómez-Calvo, M.L.; Tamayo-Castillo, G.; Araya, J.J.; Pinto-Tomás, A.A. Streptomyces sp. M54: An Actinobacteria Associated with a Neotropical Social Wasp with High Potential for Antibiotic Production. Antonie Leeuwenhoek 2021, 114, 379–398. [Google Scholar] [CrossRef] [PubMed]
  46. Zhou, L.-F.; Wu, J.; Li, S.; Li, Q.; Jin, L.-P.; Yin, C.-P.; Zhang, Y.-L. Antibacterial Potential of Termite-Associated Streptomyces spp. ACS Omega 2021, 6, 4329–4334. [Google Scholar] [CrossRef]
  47. Fukuda, T.T.H.; Helfrich, E.J.N.; Mevers, E.; Melo, W.G.P.; Van Arnam, E.B.; Andes, D.R.; Currie, C.R.; Pupo, M.T.; Clardy, J. Specialized Metabolites Reveal Evolutionary History and Geographic Dispersion of a Multilateral Symbiosis. ACS Cent. Sci. 2021, 7, 292–299. [Google Scholar] [CrossRef] [PubMed]
  48. Bae, M.; Mevers, E.; Pishchany, G.; Whaley, S.G.; Rock, C.O.; Andes, D.R.; Currie, C.R.; Pupo, M.T.; Clardy, J. Chemical Exchanges between Multilateral Symbionts. Org. Lett. 2021, 23, 1648–1652. [Google Scholar] [CrossRef]
  49. Du, Y.E.; Byun, W.S.; Lee, S.B.; Hwang, S.; Shin, Y.-H.; Shin, B.; Jang, Y.-J.; Hong, S.; Shin, J.; Lee, S.K.; et al. Formicins, N-Acetylcysteamine-Bearing Indenone Thioesters from a Wood Ant-Associated Bacterium. Org. Lett. 2020, 22, 5337–5341. [Google Scholar] [CrossRef]
  50. Santamaría, R.I.; Martínez-Carrasco, A.; Sánchez de la Nieta, R.; Torres-Vila, L.M.; Bonal, R.; Martín, J.; Tormo, R.; Reyes, F.; Genilloud, O.; Díaz, M. Characterization of Actinomycetes Strains Isolated from the Intestinal Tract and Feces of the Larvae of the Longhorn Beetle Cerambyx welensii. Microorganisms 2020, 8, 2013. [Google Scholar] [CrossRef]
  51. Baranova, A.A.; Chistov, A.A.; Tyurin, A.P.; Prokhorenko, I.A.; Korshun, V.A.; Biryukov, M.V.; Alferova, V.A.; Zakalyukina, Y.V. Chemical Ecology of Streptomyces albidoflavus Strain A10 Associated with Carpenter Ant Camponotus vagus. Microorganisms 2020, 8, 1948. [Google Scholar] [CrossRef]
  52. Hwang, S.; Le, L.T.H.L.; Jo, S.-I.; Shin, J.; Lee, M.J.; Oh, D.-C. Pentaminomycins C–E: Cyclic Pentapeptides as Autophagy Inducers from a Mealworm Beetle Gut Bacterium. Microorganisms 2020, 8, 1390. [Google Scholar] [CrossRef]
  53. Hwang, S.; Shin, D.; Kim, T.H.; An, J.S.; Jo, S.-I.; Jang, J.; Hong, S.; Shin, J.; Oh, D.-C. Structural Revision of Lydiamycin A by Reinvestigation of the Stereochemistry. Org. Lett. 2020, 22, 3855–3859. [Google Scholar] [CrossRef]
  54. Chang, P.T.; Rao, K.; Longo, L.O.; Lawton, E.S.; Scherer, G.; Van Arnam, E.B. Thiopeptide Defense by an Ant’s Bacterial Symbiont. J. Nat. Prod. 2020, 83, 725–729. [Google Scholar] [CrossRef] [PubMed]
  55. An, J.S.; Lee, J.Y.; Kim, E.; Ahn, H.; Jang, Y.-J.; Shin, B.; Hwang, S.; Shin, J.; Yoon, Y.J.; Lee, S.K.; et al. Formicolides A and B, Antioxidative and Antiangiogenic 20-Membered Macrolides from a Wood Ant Gut Bacterium. J. Nat. Prod. 2020, 83, 2776–2784. [Google Scholar] [CrossRef] [PubMed]
  56. Menegatti, C.; Lourenzon, V.B.; Rodríguez-Hernández, D.; da Paixão Melo, W.G.; Ferreira, L.L.G.; Andricopulo, A.D.; do Nascimento, F.S.; Pupo, M.T. Meliponamycins: Antimicrobials from Stingless Bee-Associated Streptomyces sp. J. Nat. Prod. 2020, 83, 610–616. [Google Scholar] [CrossRef]
  57. Zhang, L.; Song, T.; Wu, J.; Zhang, S.; Yin, C.; Huang, F.; Hang, Y.; Abbas, N.; Liu, X.; Zhang, Y. Antibacterial and Cytotoxic Metabolites of Termite-Associated Streptomyces sp. BYF63. J. Antibiot. 2020, 73, 766–771. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, Z.; Yu, Z.; Zhao, J.; Zhuang, X.; Cao, P.; Guo, X.; Liu, C.; Xiang, W. Community Composition, Antifungal Activity and Chemical Analyses of Ant-Derived Actinobacteria. Front. Microbiol. 2020, 11, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Chen, Z.; Ou, P.; Liu, L.; Jin, X. Anti-MRSA Activity of Actinomycin X2 and Collismycin A Produced by Streptomyces globisporus WA5-2-37 From the Intestinal Tract of American Cockroach (Periplaneta americana). Front. Microbiol. 2020, 11, 555. [Google Scholar] [CrossRef] [Green Version]
  60. 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]
  61. Zhang, X.; Peng, A.; Xie, W.; Wang, M.; Zheng, D.; Feng, M. Hexokinase II Inhibitory Effect of Secondary Metabolites Derived from a Streptomyces sp. Associated with Mud Dauber Wasp. Chem. Biodivers. 2020, 17, e2000140. [Google Scholar] [CrossRef]
  62. Vikeli, E.; Widdick, D.A.; Batey, S.F.D.; Heine, D.; Holmes, N.A.; Bibb, M.J.; Martins, D.J.; Pierce, N.E.; Hutchings, M.I.; Wilkinson, B. In Situ Activation and Heterologous Production of a Cryptic Lantibiotic from an African Plant Ant-Derived Saccharopolyspora Species. Appl. Environ. Microbiol. 2020, 86, e01876-19. [Google Scholar] [CrossRef] [Green Version]
  63. Zakalyukina, Y.V.; Birykov, M.V.; Lukianov, D.A.; Shiriaev, D.I.; Komarova, E.S.; Skvortsov, D.A.; Kostyukevich, Y.; Tashlitsky, V.N.; Polshakov, V.I.; Nikolaev, E.; et al. Nybomycin-Producing Streptomyces Isolated from Carpenter Ant Camponotus vagus. Biochimie 2019, 160, 93–99. [Google Scholar] [CrossRef]
  64. Ortega, H.; Batista, J.; Melo, W.; de Paula, G.; Pupo, M. Structure and Absolute Configuration of Secondary Metabolites from Two Strains of Streptomyces chartreusis Associated with Attine Ants. J. Braz. Chem. Soc. 2019, 30, 2672–2680. [Google Scholar] [CrossRef]
  65. Rodríguez-Hernández, D.; Melo, W.G.P.; Menegatti, C.; Lourenzon, V.B.; do Nascimento, F.S.; Pupo, M.T. Actinobacteria Associated with Stingless Bees Biosynthesize Bioactive Polyketides against Bacterial Pathogens. New J. Chem. 2019, 43, 10109–10117. [Google Scholar] [CrossRef]
  66. Ortega, H.E.; Ferreira, L.L.G.; Melo, W.G.P.; Oliveira, A.L.L.; Ramos Alvarenga, R.F.; Lopes, N.P.; Bugni, T.S.; Andricopulo, A.D.; Pupo, M.T. Antifungal Compounds from Streptomyces Associated with Attine Ants Also Inhibit Leishmania donovani. PLoS Negl. Trop. Dis. 2019, 13, e0007643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Shin, Y.-H.; Beom, J.Y.; Chung, B.; Shin, Y.; Byun, W.S.; Moon, K.; Bae, M.; Lee, S.K.; Oh, K.-B.; Shin, J.; et al. Bombyxamycins A and B, Cytotoxic Macrocyclic Lactams from an Intestinal Bacterium of the Silkworm Bombyx mori. Org. Lett. 2019, 21, 1804–1808. [Google Scholar] [CrossRef] [PubMed]
  68. Yoon, S.-Y.; Lee, S.R.; Hwang, J.Y.; Benndorf, R.; Beemelmanns, C.; Chung, S.J.; Kim, K.H. Fridamycin A, a Microbial Natural Product, Stimulates Glucose Uptake without Inducing Adipogenesis. Nutrients 2019, 11, 765. [Google Scholar] [CrossRef] [Green Version]
  69. Chevrette, M.G.; Carlson, C.M.; Ortega, H.E.; Thomas, C.; Ananiev, G.E.; Barns, K.J.; Book, A.J.; Cagnazzo, J.; Carlos, C.; Flanigan, W.; et al. The Antimicrobial Potential of Streptomyces from Insect Microbiomes. Nat. Commun. 2019, 10, 516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Cambronero-Heinrichs, J.C.; Matarrita-Carranza, B.; Murillo-Cruz, C.; Araya-Valverde, E.; Chavarría, M.; Pinto-Tomás, A.A. Phylogenetic Analyses of Antibiotic-Producing Streptomyces sp. Isolates Obtained from the Stingless-Bee Tetragonisca angustula (Apidae: Meliponini). Microbiology 2019, 165, 292–301. [Google Scholar] [CrossRef]
  71. Martinez, A.F.C.; de Almeida, L.G.; Moraes, L.A.B.; Cônsoli, F.L. Microbial Diversity and Chemical Multiplicity of Culturable, Taxonomically Similar Bacterial Symbionts of the Leaf-Cutting Ant Acromyrmex coronatus. Microb. Ecol. 2019, 77, 1067–1081. [Google Scholar] [CrossRef]
  72. Song, Y.-J.; Zheng, H.-B.; Peng, A.-H.; Ma, J.-H.; Lu, D.-D.; Li, X.; Zhang, H.-Y.; Xie, W.-D. Strepantibins A–C: Hexokinase II Inhibitors from a Mud Dauber Wasp Associated Streptomyces sp. J. Nat. Prod. 2019, 82, 1114–1119. [Google Scholar] [CrossRef]
  73. Hong, S.-H.; Ban, Y.H.; Byun, W.S.; Kim, D.; Jang, Y.-J.; An, J.S.; Shin, B.; Lee, S.K.; Shin, J.; Yoon, Y.J.; et al. Camporidines A and B: Antimetastatic and Anti-Inflammatory Polyketide Alkaloids from a Gut Bacterium of Camponotus kiusiuensis. J. Nat. Prod. 2019, 82, 903–910. [Google Scholar] [CrossRef]
  74. Han, H.; Guo, Z.-K.; Zhang, B.; Zhang, M.; Shi, J.; Li, W.; Jiao, R.-H.; Tan, R.-X.; Ge, H.-M. Bioactive Phenazines from an Earwig-Associated Streptomyces sp. Chin. J. Nat. Med. 2019, 17, 475–480. [Google Scholar] [CrossRef]
  75. Lee, S.R.; Lee, D.; Yu, J.S.; Benndorf, R.; Lee, S.; Lee, D.-S.; Huh, J.; de Beer, Z.W.; Kim, Y.H.; Beemelmanns, C.; et al. Natalenamides A–C, Cyclic Tripeptides from the Termite-Associated Actinomadura sp. RB99. Molecules 2018, 23, 3003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Jiang, S.; Piao, C.; Yu, Y.; Cao, P.; Li, C.; Yang, F.; Li, M.; Xiang, W.; Liu, C. Streptomyces capitiformicae sp. nov., a Novel Actinomycete Producing Angucyclinone Antibiotics Isolated from the Head of Camponotus japonicus Mayr. Int. J. Syst. Evol. Microbiol. 2018, 68, 118–124. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, D.; Kang, K.; Lee, H.-J.; Kim, K. Chemical Characterization of a Renoprotective Metabolite from Termite-Associated Streptomyces sp. RB1 against Cisplatin-Induced Cytotoxicity. Int. J. Mol. Sci. 2018, 19, 174. [Google Scholar] [CrossRef] [Green Version]
  78. Guo, Z.K.; Wang, R.; Chen, F.X.; Liu, T.M. Streptoxamine, an Unprecedented Benzoisoindole-Deferoxamine Hybrid from the Locust-Derived Streptomyces sp. HKHCa2. Fitoterapia 2018, 127, 25–28. [Google Scholar] [CrossRef]
  79. Liu, C.; Han, C.; Jiang, S.; Zhao, X.; Tian, Y.; Yan, K.; Wang, X.; Xiang, W. Streptomyces lasii sp. nov., a Novel Actinomycete with Antifungal Activity Isolated from the Head of an Ant (Lasius flavus). Curr. Microbiol. 2018, 75, 353–358. [Google Scholar] [CrossRef]
  80. Guo, H.; Benndorf, R.; König, S.; Leichnitz, D.; Weigel, C.; Peschel, G.; Berthel, P.; Kaiser, M.; Steinbeck, C.; Werz, O.; et al. Expanding the Rubterolone Family: Intrinsic Reactivity and Directed Diversification of PKS-Derived Pyrans. Chem. Eur. J. 2018, 24, 11319–11324. [Google Scholar] [CrossRef]
  81. Benndorf, R.; Guo, H.; Sommerwerk, E.; Weigel, C.; Garcia-Altares, M.; Martin, K.; Hu, H.; Küfner, M.; de Beer, Z.; Poulsen, M.; et al. Natural Products from Actinobacteria Associated with Fungus-Growing Termites. Antibiotics 2018, 7, 83. [Google Scholar] [CrossRef] [Green Version]
  82. Guo, H.; Benndorf, R.; Leichnitz, D.; Klassen, J.L.; Vollmers, J.; Görls, H.; Steinacker, M.; Weigel, C.; Dahse, H.-M.; Kaster, A.-K.; et al. Isolation, Biosynthesis and Chemical Modifications of Rubterolones A-F: Rare Tropolone Alkaloids from Actinomadura sp. 5-2. Chem. Eur. J. 2017, 23, 9338–9345. [Google Scholar] [CrossRef] [Green Version]
  83. Zhang, M.; Yang, C.L.; Xiao, Y.S.; Zhang, B.; Deng, X.Z.; Yang, L.; Shi, J.; Wang, Y.S.; Li, W.; Jiao, R.H.; et al. Aurachin SS, a New Antibiotic from Streptomyces sp. NA04227. J. Antibiot. 2017, 70, 853–855. [Google Scholar] [CrossRef]
  84. Cao, T.; Mu, S.; Lu, C.; Zhao, S.; Li, D.; Yan, K.; Xiang, W.; Liu, C. Streptomyces amphotericinicus sp. nov., an Amphotericin-Producing Actinomycete Isolated from the Head of an Ant (Camponotus japonicus Mayr). Int. J. Syst. Evol. Microbiol. 2017, 67, 4967–4973. [Google Scholar] [CrossRef]
  85. Ye, L.; Zhao, S.; Li, Y.; Jiang, S.; Zhao, Y.; Li, J.; Yan, K.; Wang, X.; Xiang, W.; Liu, C. Streptomyces lasiicapitis sp. nov., an Actinomycete That Produces Kanchanamycin, Isolated from the Head of an Ant (Lasius fuliginosus L.). Int. J. Syst. Evol. Microbiol. 2017, 67, 1529–1534. [Google Scholar] [CrossRef] [PubMed]
  86. Ortega, H.E.; Batista, J.M.; Melo, W.G.P.; Clardy, J.; Pupo, M.T. Absolute Configurations of Griseorhodins A and C. Tetrahedron Lett. 2017, 58, 4721–4723. [Google Scholar] [CrossRef] [PubMed]
  87. Qin, Z.; Munnoch, J.T.; Devine, R.; Holmes, N.A.; Seipke, R.F.; Wilkinson, K.A.; Wilkinson, B.; Hutchings, M.I. Formicamycins, Antibacterial Polyketides Produced by Streptomyces formicae Isolated from African Tetraponera Plant-Ants. Chem. Sci. 2017, 8, 3218–3227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mevers, E.; Chouvenc, T.; Su, N.-Y.; Clardy, J. Chemical Interaction among Termite-Associated Microbes. J. Chem. Ecol. 2017, 43, 1078–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Shin, Y.-H.; Bae, S.; Sim, J.; Hur, J.; Jo, S.-I.; Shin, J.; Suh, Y.-G.; Oh, K.-B.; Oh, D.-C. Nicrophorusamides A and B, Antibacterial Chlorinated Cyclic Peptides from a Gut Bacterium of the Carrion Beetle Nicrophorus concolor. J. Nat. Prod. 2017, 80, 2962–2968. [Google Scholar] [CrossRef]
  90. Beemelmanns, C.; Ramadhar, T.R.; Kim, K.H.; Klassen, J.L.; Cao, S.; Wyche, T.P.; Hou, Y.; Poulsen, M.; Bugni, T.S.; Currie, C.R.; et al. Macrotermycins A–D, Glycosylated Macrolactams from a Termite-Associated Amycolatopsis sp. M39. Org. Lett. 2017, 19, 1000–1003. [Google Scholar] [CrossRef]
  91. Wyche, T.P.; Ruzzini, A.C.; Beemelmanns, C.; Kim, K.H.; Klassen, J.L.; Cao, S.; Poulsen, M.; Bugni, T.S.; Currie, C.R.; Clardy, J. Linear Peptides Are the Major Products of a Biosynthetic Pathway That Encodes for Cyclic Depsipeptides. Org. Lett. 2017, 19, 1772–1775. [Google Scholar] [CrossRef]
  92. Ma, S.Y.; Xiao, Y.S.; Zhang, B.; Shao, F.L.; Guo, Z.K.; Zhang, J.J.; Jiao, R.H.; Sun, Y.; Xu, Q.; Tan, R.X.; et al. Amycolamycins A and B, Two Enediyne-Derived Compounds from a Locust-Associated Actinomycete. Org. Lett. 2017, 19, 6208–6211. [Google Scholar] [CrossRef]
  93. Xiao, Y.S.; Zhang, B.; Zhang, M.; Guo, Z.K.; Deng, X.Z.; Shi, J.; Li, W.; Jiao, R.H.; Tan, R.X.; Ge, H.M. Rifamorpholines A–E, Potential Antibiotics from Locust-Associated Actinobacteria Amycolatopsis sp. Hca4. Org. Biomol. Chem. 2017, 15, 3909–3916. [Google Scholar] [CrossRef]
  94. Human, Z.R.; Slippers, B.; De Beer, Z.W.; Wingfield, M.J.; Venter, S.N. Antifungal Actinomycetes Associated with the Pine Bark Beetle, Orthotomicus erosus, in South Africa. S. Afr. J. Sci. 2017, 113, 1–7. [Google Scholar] [CrossRef] [Green Version]
  95. Arango, R.A.; Carlson, C.M.; Currie, C.R.; McDonald, B.R.; Book, A.J.; Green, F.; Lebow, N.K.; Raffa, K.F. Antimicrobial Activity of Actinobacteria Isolated From the Guts of Subterranean Termites. Environ. Entomol. 2016, 45, 1415–1423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Axenov-Gribanov, D.; Rebets, Y.; Tokovenko, B.; Voytsekhovskaya, I.; Timofeyev, M.; Luzhetskyy, A. The Isolation and Characterization of Actinobacteria from Dominant Benthic Macroinvertebrates Endemic to Lake Baikal. Folia Microbiol. 2016, 61, 159–168. [Google Scholar] [CrossRef] [PubMed]
  97. Holmes, N.A.; Innocent, T.M.; Heine, D.; Bassam, M.A.; Worsley, S.F.; Trottmann, F.; Patrick, E.H.; Yu, D.W.; Murrell, J.C.; Schiøtt, M.; et al. Genome Analysis of Two Pseudonocardia Phylotypes Associated with Acromyrmex Leafcutter Ants Reveals Their Biosynthetic Potential. Front. Microbiol. 2016, 7, 2073. [Google Scholar] [CrossRef] [Green Version]
  98. Liu, S.; Xu, M.; Zhang, H.; Qi, H.; Zhang, J.; Liu, C.; Wang, J.; Xiang, W.; Wang, X. New Cytotoxic Spectinabilin Derivative from Ant-Associated Streptomyces sp. 1H-GS5. J. Antibiot. 2016, 69, 128–131. [Google Scholar] [CrossRef]
  99. Liu, C.-X.; Liu, S.-H.; Zhao, J.-W.; Zhang, J.; Wang, X.-J.; Li, J.-S.; Wang, J.-D.; Xiang, W.-S. A New Spectinabilin Derivative with Cytotoxic Activity from Ant-Derived Streptomyces sp. 1H-GS5. J. As. Nat. Prod. Res. 2017, 19, 924–929. [Google Scholar] [CrossRef]
  100. Kang, H.R.; Lee, D.; Benndorf, R.; Jung, W.H.; Beemelmanns, C.; Kang, K.S.; Kim, K.H. Termisoflavones A–C, Isoflavonoid Glycosides from Termite-Associated Streptomyces sp. RB1. J. Nat. Prod. 2016, 79, 3072–3078. [Google Scholar] [CrossRef]
  101. Um, S.; Bach, D.-H.; Shin, B.; Ahn, C.-H.; Kim, S.-H.; Bang, H.-S.; Oh, K.-B.; Lee, S.K.; Shin, J.; Oh, D.-C. Naphthoquinone–Oxindole Alkaloids, Coprisidins A and B, from a Gut-Associated Bacterium in the Dung Beetle, Copris tripartitus. Org. Lett. 2016, 18, 5792–5795. [Google Scholar] [CrossRef]
  102. Bai, L.; Liu, C.; Guo, L.; Piao, C.; Li, Z.; Li, J.; Jia, F.; Wang, X.; Xiang, W. Streptomyces formicae Sp. Nov., a Novel Actinomycete Isolated from the Head of Camponotus japonicus Mayr. Antonie Leeuwenhoek 2016, 109, 253–261. [Google Scholar] [CrossRef]
  103. The genus Escovopsis was recently separated into 3 distinct genera, for details see: Montoya, Q.V.; Martiarena, M.J.S.; Bizarria, R.; Gerardo, N.M.; Rodrigues, A. Fungi Inhabiting Attine Ant Colonies: Reassessment of the Genus Escovopsis and Description of Luteomyces and Sympodiorosea Gens. Nov. IMA Fungus 2021, 12, 23. [CrossRef]
  104. Alferova, V.A.; Shuvalov, M.V.; Korshun, V.A.; Tyurin, A.P. Naphthoquinone-Derived Polyol Macrolides from Natural Sources. Russ. Chem. Bull. 2019, 68, 955–966. [Google Scholar] [CrossRef]
  105. Berdy, B.; Spoering, A.L.; Ling, L.L.; Epstein, S.S. In Situ Cultivation of Previously Uncultivable Microorganisms Using the Ichip. Nat. Protoc. 2017, 12, 2232–2242. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. The experimental design used in the selected publications.
Scheme 1. The experimental design used in the selected publications.
Biology 11 01676 sch001
Figure 1. Map of sample collection locations (marked with red points; involved countries are colored beige; main location clusters are highlighted with yellow circles).
Figure 1. Map of sample collection locations (marked with red points; involved countries are colored beige; main location clusters are highlighted with yellow circles).
Biology 11 01676 g001
Figure 2. Classification of the studied insects.
Figure 2. Classification of the studied insects.
Biology 11 01676 g002
Figure 3. Isolation of axenic cultures: isolated genera and new described species. * the taxonomic description does not match the International code of nomenclature of prokaryotes, for details see ref. [81]. ** for taxonomic description see ref. [97].
Figure 3. Isolation of axenic cultures: isolated genera and new described species. * the taxonomic description does not match the International code of nomenclature of prokaryotes, for details see ref. [81]. ** for taxonomic description see ref. [97].
Biology 11 01676 g003
Figure 4. The detected antibiotics: main trends.
Figure 4. The detected antibiotics: main trends.
Biology 11 01676 g004
Figure 5. Main types of protective symbiosis described for insect-actinobacteria systems: (A) Food source protection; (B) Direct self-protection.
Figure 5. Main types of protective symbiosis described for insect-actinobacteria systems: (A) Food source protection; (B) Direct self-protection.
Biology 11 01676 g005
Scheme 2. Specialized metabolites for food source protection (new elucidated structures are colored black, known metabolites are colored gray).
Scheme 2. Specialized metabolites for food source protection (new elucidated structures are colored black, known metabolites are colored gray).
Biology 11 01676 sch002
Scheme 3. Specialized metabolites for self-protection (new elucidated structures are colored black, known metabolites are colored gray).
Scheme 3. Specialized metabolites for self-protection (new elucidated structures are colored black, known metabolites are colored gray).
Biology 11 01676 sch003
Scheme 4. A specialized metabolite as a niche-defense compound (colored gray as a known metabolite).
Scheme 4. A specialized metabolite as a niche-defense compound (colored gray as a known metabolite).
Biology 11 01676 sch004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Baranova, A.A.; Zakalyukina, Y.V.; Ovcharenko, A.A.; Korshun, V.A.; Tyurin, A.P. Antibiotics from Insect-Associated Actinobacteria. Biology 2022, 11, 1676. https://doi.org/10.3390/biology11111676

AMA Style

Baranova AA, Zakalyukina YV, Ovcharenko AA, Korshun VA, Tyurin AP. Antibiotics from Insect-Associated Actinobacteria. Biology. 2022; 11(11):1676. https://doi.org/10.3390/biology11111676

Chicago/Turabian Style

Baranova, Anna A., Yuliya V. Zakalyukina, Anna A. Ovcharenko, Vladimir A. Korshun, and Anton P. Tyurin. 2022. "Antibiotics from Insect-Associated Actinobacteria" Biology 11, no. 11: 1676. https://doi.org/10.3390/biology11111676

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