Send Orders for Reprints to [email protected] 1291

Current Topics in Medicinal Chemistry, 2020, 20, 1291-1299 REVIEW ARTICLE

ISSN: 1568-0266 eISSN: 1873-4294

Impact Factor: The Microbiome: A Reservoir to Discover New Antimicrobials Agents 3.442 The international journal for in-depth reviews on Current Topics in Medicinal Chemistry

BENTHAM SCIENCE

Sébastien Boutin1,2,* and Alexander H. Dalpke3

1Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Hei- delberg, Germany; ²German Center for Lung Research (DZL), TLRC Heidelberg, 69120 Heidelberg, Germany; 3Institute of Medical Microbiology and Hygiene, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Gemany

Abstract: Nature offered mankind the first golden era of discovery of novel antimicrobials based on A R T I C L E H I S T O R Y the ability of eukaryotes or micro-organisms to produce such compounds. The microbial world proved to be a huge reservoir of such antimicrobial compounds which play important functional roles in every

Received: November 11, 2019 environment. However, most of those organisms are still uncultivable in a classical way, and therefore, Revised: February 10, 2020 the use of extended culture or DNA based methods (metagenomics) to discover novel compounds Accepted: February 17, 2020 promises usefulness. In the past decades, the advances in next-generation sequencing and bioinformat- DOI: 10.2174/1568026620666200320112731 ics revealed the enormous diversity of the microbial worlds and the functional repertoire available for studies. Thus, data-mining becomes of particular interest in the context of the increased need for new due to antimicrobial resistance and the rush in antimicrobial discovery. In this review, an overview of principles will be presented to discover new natural compounds from the microbiome. We describe culture-based and culture-independent (metagenomic) approaches that have been developed to identify new antimicrobials and the input of those methods in the field as well as their limitations.

Keywords: Antimicrobial peptides, Metagenomics, Data mining, Bioinformatics, Microbiome, Antimicrobials agents.

1. INTRODUCTION The classical procedure to find antimicrobial compounds is to look for them in nature (Fig. 1). The first accessible From the 1930s to the 1960’s, the golden age of antimi- organisms producing antimicrobials were plants and those crobial discoveries took place. Those discoveries lead to the compounds were used for millennia. However, access to introduction in the clinics of the major classes of drugs cur- fungi and bacteria with microbial culture revolutionized the rently used. After the first discoveries, the research field fo- field [5]. The majority of antibiotics currently implemented cused more on the optimization of those drugs in the follow- in clinic originate from cultivable microorganisms [6]. In the ing 50 years [1]. The phase of optimization was always fol- environment, microbes, however, are never living in a lowed by the emergence of antimicrobial resistance leading monoculture environment and therefore, most of them have to an “arm race” to counteract those resistances [2]. Since the developed ways to communicate and/or defend themselves 2000’s, only a few drugs of entirely new classes were ap- against other bacteria/fungi. The billions of years of evolu- proved and discovered, leading to a threat in health due to tion involve in this arms race created a huge arsenal of those the steady increase of emerging pathogens and multi-drug compounds [7]. Most of them are products of “secondary resistant (MDR) bacteria [3]. The currently used drugs focus metabolism,” which are not required for survival under labo- on two major targets in 5 different modes of action. Some ratory conditions but are most likely increasing the fitness of drugs alter the structure of the cell by inhibiting the cell wall their producer in the native complex biome [5]. Those prod- synthesis or disrupting the cell membrane integrity while ucts are often encoded by a biosynthetic gene cluster (BGC), others target the molecular machinery inhibiting either the which is a physically clustered group of two or more genes DNA or RNA synthesis or the protein synthesis via the bac- as an within the genome. Together the BGCs will terial ribosome or metabolic pathways [4]. The emergence of encode cooperatively a complete biosynthetic pathway lead- MDR bacteria guided the field of research into ing to the production of a specialized metabolite and possible actually two ways; understanding the molecular mechanisms chemical variants. Furthermore, genes involved in the regu- leading to the resistance to manage our use of the affected lation, such as transcription factors and transporters, are usu- drugs and developing new antibiotics targeting new path- ally also present in these clusters [8, 9]. ways or identifying new modes of action in known path- ways. The field of microbiology observed in the last decades a shift in the paradigm of environmental and host-related mi- *Address correspondence to this author at the Department of Infectious crobes. The advances in culture-independent methods Diseases, Medical Microbiology and Hygiene, University Hospital Heidel- showed that microbes are part of a complex ecosystem and berg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; the diversity of this microbiome was by far underestimated Current Topics in Medicinal Chemistry E-mail: [email protected]

1873-4294/20 $65.00+.00 © 2020 Bentham Science Publishers 1292 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 Boutin and Dalpke

Fig. (1). Summary of the pipelines used to discover microbiome derived antibiotic. The classical way consists to culture the organisms di- rectly from the environment and test them for antimicrobial production. An evolution of this way leads to the development of in situ culture where the organisms is grown in isolation in a permeable chamber put in the original environment (displayed as a iChip in the figure). The environmental DNA (eDNA) can be used to construct metagenomic library either via direct fragmentation and ligation to a vector or via a PCR targeted approach using conserved region of the biosynthetic gene clusters (BGCs) to amplify the diversity of BGCs within the envi- ronment. Those PCR products can also be integrated in a vector to construct a metagenomic library. The metagenomic data can also be ex- plore (“data-mining”) via bio-informatic to search for new BGCs and design new primers to isolate them from the metagenome. The pre- pared library can then be transferred into a heterologous host (i.e. Escherischia coli) to perform an antimicrobial test assay. Each part of the different path is explained and discuss in details in the separate sections of the manuscript. by classical culture. It is estimated that classical culture re- capacities as well as annotation of the sequences themselves, trieves around 1% of the microbiome of environmental sam- such approaches appear to be very promising. ples, yet reaches slightly better recovery in human-associated 2. THE WORLD’S MICROBIOME AS A POTENTIAL microbiomes, due to the optimization of growth media for SOURCE FOR NEW ANTIBIOTICS detection of human pathogenic bacteria [10]. Many research- ers are currently working on extending culture methods to The environmental microbiome is a source of new anti- improve culturability with impressive advances, especially in biotics, but unfortunately, culture-based approaches barely humans [11–13]. However, symbiotic microbes will still be scratch the surface of the diversity of those microbiomes difficult to culture and the production of antibiotic com- [14]. The advances in the metagenomic field pave the way pounds will be influenced by the growth conditions as well for the exploration of highly diverse microbiomes such as as other organisms growing in nature together with antimi- the soil and aquatic microbiome. The exploration of those crobial producers. Therefore, the field of drug discovery now two major environments showed that the BGCs involved in focuses on metagenomic approaches: This approach aims at the production of antimicrobial compounds by those mi- using the DNA from the environmental microbiome (eDNA) crobes are geographically heterogeneous and show a strong to discover antimicrobial functions by prediction instead of environmental specificity [15-17]. This work was the basis or prior to phenotypical identification of such compounds. of the exploration of diverse environments and geographi- Given the enormous technical developments in sequencing cally distinct biomes to mine for new antibiotics from a highly diverse environment, allowing a low re-discovery rate The Microbiome: A Reservoir to Discover New Antimicrobials Agents Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 1293

[18]. Naturally, the metagenomic approach can be used with cro-organisms. Those chambers improved the culture of mi- any source material, but the main focus of the research is crobes, which are dependent on environmental/host nutrients currently soil, water and host-associated microbiomes as opening the scope of cultured microbes for antibiotic discov- supported by the major consortium performing metagenom- ery. ics and offering a huge database for data-mining [19, 20]. An updated version of the diffusion chamber is the iChip. Soil is of high interest because it is one of the most diverse The principle is the same; bacteria are serially diluted using environments and easily accessible. Many studies are cur- microfluidic diffusion chambers. However, the device pre- rently done to improve both culture and data-mining to find sents many holes containing agar where one microbial cell new pharmaceutical compounds from the soil [21]. The ma- can be isolated, then offering a high-throughput screening rine and clearwater environment is not as easy to explore, but cultivation method [36]. After isolation in the well, top and the high diversity of this environment is reflecting the bio- bottom sites of the chip are sealed with a semi-permeable synthetic potential of these biomes [22]. Finally, host- membrane and the chip is placed in the source environment associated microbiomes might be good candidates for antim- or a simulated environment allowing the nutrient and growth icrobial compounds which are harmless for the eukaryotic factors to diffuse in the agar favoring the growth of previ- hosts. This point is crucial for the translational aspect of the ously uncultivable organisms. This device allowed the isola- approach of drug discovery. The human microbiome project tion of soil Betaproteobacteria Eleftheria terrae, which pro- offers a good database for data mining and a study based on duces a new class of antibiotics called Teixobactin [37]. This the available metagenome showed that 3,118 biosynthetic antibiotic proved highly efficient against gram-positive bac- gene clusters were identified in the human microbiome and teria by binding to the lipid II and then inhibiting the produc- discovered a previously unknown antibiotic, lactocillin, pro- tion of the peptidoglycan layer. However, due to this mode duced by a vaginal commensal [23]. While human is the of action, this antimicrobial compound is inactive against target of choice, other eukaryotic hosts like sea sponges and gram-negative. coral already proved to be a good source for the identifica- tion of natural products, which may serve beneficial roles in In situ cultivation assays were modified and optimized in human health [24-26] various different ways and can be placed in solid or liquid media. Any bacteria depending only on nutrient diffusion 3. ANTIBIOTIC DISCOVERY FROM CULTURED from the native environment may grow, yet limitations have ORGANISMS to be considered [30]. Contamination from the in situ media to the chambers is a major problem as well as the moisture of The classical way to discover antimicrobial compounds solid media like soil to allow the diffusion of the nutrient used since the 1930’s consists in the screening of bacteria into the agar. The permeability of liquid or solid media to from natural sources, such as soil and water for bioactive gaseous exchange also has to be optimized to allow the or- compounds of pharmacological interest. This methodology ganisms to access the necessary gas. However, the in-situ brought a lot of promises and new drugs, however, the lack cultivation shows promises in cultivating new species and of cultivability of the microbiome leads to a low amount of therefore allowing the discovery of new compounds. As a species studied. Most of the antimicrobials currently discov- limitation, some organisms are not only dependent on the ered or under use are produced by organisms belonging to environment or host nutrients but the interactions with other myxobacteria, cyanobacteria, actinomycetes, Pseudomonas, microbes. Co-culture is currently not allowed by the in situ and Bacillus species [27]. Furthermore, secondary metabo- assays, but many studies showed that secondary metabolic lites are clades specific, and therefore limiting our focus on 5 production is influenced by the presence of other microbes clades explains the high rediscovery rates of known antibiot- and that systems must be developed to study those interac- ics [28,29]. The extension of culture conditions and media tions [38, 39]. Some researchers proposed to modify the can be laborious and has been mostly employed for the hu- iChip system to allow the diffusion within the chip of the man microbiome [11]. For environmental samples, another metabolite, allowing a full screening of the microbiome methodology is currently in use, which is in situ cultivation. while keeping the species isolated [40]. This concept basically involves the cultivation of the organ- isms in its natural niche [30-32]. 4. SEQUENCE-BASED METAGENOMICS TO DIS- COVER NEW BIOSYNTHETIC GENE CLUSTER One approach consists of capturing bacterial cells in a diffusion chamber which has access to a flow of nutrients Extended culture will still face issues of unreliable identi- simulating the natural environment of their origin. The sam- fication due to the absence of known close relative organ- ples are serially diluted to minimize the amount of cells per isms helping to identify the cultured one. Therefore, identifi- chambers and inoculated in agar. The agar is then covered by cation will rely on genome sequencing or phenotypic charac- permeable membranes allowing the passage of nutrients terization by an expert. Furthermore, culture in a simulated from the simulated environment. This technique allows to environment or medium remains an artificial model that culture more microorganisms than the classical culture-based might lead to irreproducibility or loss of function in a non- techniques [33, 34]. The development of diffusion chambers selective medium [41]. Therefore, a phenotype observed in was particularly helpful in growing species from the phylum the natural environment cannot be recapitulated in the lab for Verrucomicrobia which grow after subsequent incubation in production. To be able to avoid such non-transferability to the diffusion chambers [34]. Diffusion chambers were also the lab, working with the DNA seems to be a good option. improving the culture of symbiotic organisms from hosts like The advances in genomics and bioinformatics associated sponges [35]. The growth chambers can be implanted in the with the development of cheaper and faster sequencing tech- tissue of the host allowing the growth of host-dependent mi- nologies allow now research to explore the full gene reper- 1294 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 Boutin and Dalpke toire from any microbiome without culture [42]. Metage- the extract of condensation and ketosynthase domains pre- nomics can be applied in two different approaches to study sent in respectively NRPS and PKS genes from the sequence the diversity of the microbiome: targeted (16S, ITS,…) or data. The sequence is then queried to a set of manually cu- not targeted (Whole genome sequencing)[43]. In the field of rated reference genes from well-characterized chemical antibiotic discovery, the term metagenomic is slightly differ- pathways to identify potential secondary metabolite do- ent than the one used in microbial ecology. The term de- mains. Then a phylogeny is inferred to predict if the product scribed the exploration of the full content of the genome is similar to or different from previously known biosynthetic within the microbial community in the ecology field. In con- pathways [51]. eSNaPD is designed in a same way but works trast, in the antibiotic discovery field, it means the functional mostly with PCR-generated sequence tags using degenerate screening of antibiotic production via bioinformatic predic- primers targeting conserved biosynthetic genes. This relies tion or the creation of clones containing a fragment of the on the high similarities seen in natural product biosynthetic metagenome. However, both fields are using either the full genes which allow the design of primers targeting those genomic repertoire or the targeted region of the metagenome. genes to explore the structure of the biosynthetic domains. Advances in metagenomic sequencing, library preparation, Those genes clusters are then identified as potential candi- screening and engineering, which allow the study of more dates with a curated database as well as a phylogeny to infer organisms, will be the focus of the following section. potential new functionality [18]. Targeted methods, often named sequence-based methods The use of degenerate primers to amplify a conserved re- are relying on the premise that homologous sequences will gion of the biosynthetic genes of interest allows to create produce a similar protein and, therefore, phenotype. There- libraries for amplicon sequencing to identify new variants of fore, it becomes easy to screen metagenomic data for homol- the genes which might lead to a new antimicrobial product. ogy with known biosynthetic pathways in a high-throughput The PCR-based sequence has the advantage of being much manner. The challenging step is to identify the homologous more sensitive in a high diverse microbiome; estimates are sequences and predict structural proteins during the data- by 10 to 100 fold increase in sensitivity compared to shotgun mining of the metagenome. However, advances in computa- sequencing [52]. In most of the cases, the gene of interest is tional biology result in the development of several good an NRPS or a PKS gene. One of the most recent examples of tools to identify and annotate potential natural products. sequence targeted discovery is the identification of malacid- Those tools are based on the idea that the natural product ins [53]. This new natural product was found in soil micro- results from biosynthetic gene clusters that follow a highly biomes and is active against multidrug-resistant pathogens conserved logic. Therefore, the algorithms are looking for including methicillin-resistant . The core enzyme/gene-based sequences similarities within the authors used the targeted method to screen several soil mi- databases [44]. One of the most used programs is antiS- crobiomes and selected an environment rich in a new clade MASH, which uses a Hidden Markov Models (HMM) based of malacidins to follow up with the functional metagenomic algorithm named ClusterFinder to identify the BGCs [45, approach that will be reviewed in the next section. 46]. The advantage of an HMM-based algorithm is that it The main disadvantage of sequence based approaches is will identify BGCs that lack high similarity in domain struc- that the scope of research is limited to genes we already ture to any known biosynthetic gene clusters, circumventing know due to the design of primers. Furthermore, most of the the problem that had previously been posed in the identifica- candidate identification is depending on either homology to a tion of biosynthetic gene clusters from sequence data. There- database or a ruling system and therefore, the identification fore, by focusing on the active site via an active site finder of completely novel products is merely impossible. We will module, the program is able to identify conserved amino acid always discover related products or derivate from the exist- motifs in the enzymes of interest like non-ribosomal peptide ing product. For example, the main pathways studied are synthetase (NRPS) and polyketide synthase (PKS). The pro- polyketides, or nonribosomally synthesized peptides, there- gram is also able to predict the structure of the natural prod- fore only pathways which are closely related and fall into the uct based on the sequence of the annotated BGCs. However, rules are implemented in the mining software. The im- such accuracy came with the need for a well-annotated and provement in the knowledge of the evolution of BGCs will assembled metagenome. This step might be a high burden help to extend the rules to identify new pathways. That is the and is not a trivial topic. The need for computational power basis of new software like EvoMining to use evolutionary and expertise in the assembly of high-quality genomes knowledge to relax the rules to identify new compounds makes it extremely complex [47, 48]. Many improvements [54]. Basically, it is considered that BGCs often evolved by have been made to make metagenome assembly more “user duplication and divergence of primary metabolism enzymes. friendly” and almost fully automated programs are now Therefore, the software uses the core metabolism pathways available such as MetaSort or MetaWrap [49, 50]. However, shared between many bacterial species to calculate the di- the quality of the assembly and the reliability of the contigs vergence to identify BGCs that have likely been repurposed remain a problem to be solved. Furthermore, it is still diffi- for secondary metabolite biosynthesis [55]. cult to sequence a highly diverse microbiome like the one from the soil, which contains 104-5 unique species with suffi- cient coverage to assemble the reads correctly. Therefore, a 5. FUNCTIONAL METAGENOMICS TO TEST NEW BIOSYNTHETIC GENE CLUSTER FOR ANTIBIOTIC new set of tools was developed to circumvent the need of longer contigs. Those tools like NaPDos or eSNaPD can DISCOVERY identify secondary metabolite genes based on short reads by Finding the sequence of a new potential antibiotic is not focusing on core domains [18, 51]. NaPDos is focusing on an end per se. Those sequences need to be validated in a The Microbiome: A Reservoir to Discover New Antimicrobials Agents Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 1295 functional assay to ensure the expression and antimicrobial RNA-polymerase of E. coli [67]. To overcome that problem, function of the product. Therefore, most of the studies com- some works are done to express other sigma factors in the E. bine sequence-based to functional metagenomics in the dis- coli host [68, 69]. Another option is to use a non-model het- covery process [53]. This technique is at the interface be- erologous host such as Streptomyces, Sacharopolyspora or tween culture-based and sequence-based techniques. The Myxococcus [70–72]. In general, researchers need to con- basic idea of the functional metagenomic pipeline consists of sider whether the codons are optimized to help expression in extracting total DNA from a microbial community and shear the chosen heterologous hosts and therefore use a host which in fragments (1-5 kb) that can be shotgun cloned in an ex- is not too phylogenetically distant from the original source pression vector. The vector is transferred in the appropriate [73]. To obtain a cleaner expression of the BGCs, the ge- heterologous host (e.g. E. coli) and tested for antibiotic pro- nome of the host can also be minimized to the minimum core duction. The main advantage is that it does not requires prior genome to ensure no competition between the pathways. The knowledge of the DNA sequences or function and will allow new secondary metabolite pathway will then be the only user the discovery of completely new antibiotic or divergent of an efficient supply of primary metabolic precursors and pathways which are not in the database yet [56]. While the biochemical energy;, therefore, being optimized for the pro- theoretical concept of the technique seems simple, the reali- duction of the compound [74]. Alternatively, a co-culture of zation relies on major critical steps such as the extraction and two heterologous hosts expressing different complementary fragmentation of the DNA, the library preparation, the parts of the BGCs can be used to optimize the production. It choice of the heterologous host for expression and the func- was previously demonstrated that the combined use of an tional assay to test the natural product [57]. engineered E. coli and S. cerevisiae as a microbial commu- nity will produce a precursor of the anti-cancer drug pacli- The preparation of the DNA for functional metagenomics taxel. The taxadiene is produced by the E. coli and is trans- will struggle with the same problems that all microbiome formed into taxanes by the engineered S. cerevisiae. The studies are facing; the need for representative and pure DNA extraction. The lysis and extraction methods (precipitation or combination of the two strains increased the production of the compound by 1.8-fold [75]. affinity columns) will perform differently in gram-negative and gram-positive bacteria. Mechanical lysis associated with The final step of the functional metagenomics is to per- enzymatic lysis is mandatory to extract DNA from gram- form the culture assay to validate the expression of the an- positive bacteria and fungi [58]. Furthermore, the purity of timicrobial compounds. Antimicrobial production is a more the DNA is crucial to avoid contaminants, which will inhibit complicated phenotype to select for than antimicrobial resis- the shearing and integration into the vector, therefore, a puri- tance and therefore needs the use of appropriate screening fication step should be included [59, 60]. The BGCs are of- strategies to identify and isolate the proper clones [57]. The ten organized as an operon and therefore cloning pieces of most used technique is the double-agar layer [76]. This DNA can result in functional machinery. However, some method consists of growing the heterologous host expressing machinery requires hundreds of genes (~20kb to 100kb) for a the vector on agar plates and then a top agar containing the complex product; therefore, shearing and cloning such a big indicator strain is overlaid and scored for antimicrobial activ- piece of DNA is challenging. The use of cosmids and fos- ity by looking for a zone of inhibition. This is a high- mids helps to integrate large DNA fragments in the host in throughput technique allowing us to screen a large number an efficient way [61] but to integrate fragments of hundreds of clones (>100,000) from a metagenomic derived library of kilobases, the use of Bacterial Artificial Chromosome [77, 78]. A new method was developed where the top layer (BAC) is needed as they can integrate >300 kb fragment is replaced by a “bacteriospray”. The method is based on the [62]. As cosmids and fosmids are more efficient than BACs use of an air brush, which will spray an even mist of the in- for transformation, a recent development consists of reas- dicator bacteria on the bottom agar containing the colonies sembling pieces of overlapping clones into a BAC using of the heterologous host. As the layer is more uniform, the transformation-associated recombination (TAR) in Sac- authors claimed that the technique is more sensitive, allow- charomyces cerevisiae [63]. Modification of the host also ing to test 5–10 times more clones per agar plate [79]. Of allows the enrichment of functionally active BGCs. Most of course, the methods used in the extended culture method, the known antimicrobial compounds (NRPS and PKS) are such as the diffusion chamber assay and the iChip, can be dependent on their own phosphopantetheine transferase used for screening as the membrane is impermeable to bacte- (PPTase) [64]. Therefore the use of a PPTase deficient host ria. The heterologous host can be grown in the chambers will screen vectors containing a PPTase and most likely an while the indicator bacteria grow in the media. However, as active BGCs [65]. discussed previously, this method is difficult to extend in a high-throughput system to test 100,000s clones. Another critical step after the preparation of the vector is to find a suitable host. Indeed, the transcriptional machinery of the host organism has to recognize promoters from the CONCLUSION metagenome to be able to translate metagenomic transcripts. The advances in extended culture and metagenomics The failure of this step is the main reason for the inability to change our way to explore microbial communities and their detect antimicrobial activity from metagenomic constructs potential. However, metagenomics is not yet “user-friendly,” [66]. Most of the studies are using Escherichia coli as a het- and many steps need to be optimized. The virus/phage world erologous host because its genetic content and transcriptional is under-explored for antimicrobial usage, although we can machinery are well understood. However, the expression of access part of their function via their DNA. The technology metagenomic DNA is limited in this model by the recogni- is still not low cost, so the coverage needed to cover func- tion of the promoter by the sigma factor subunits of the tions of the biosphere is rarely reached by the metagenomic 1296 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 Boutin and Dalpke project. However, big consortia like the human microbiome Tsunematsu, Y.; Wiemann, P.; Wyckoff, E.; Yan, X.; Yim, G.; Yu, project or the earth microbiome project will clearly help us F.; Xie, Y.; Aigle, B.; Apel, A.K.; Balibar, C.J.; Balskus, E.P.; Barona-Gómez, F.; Bechthold, A.; Bode, H.B.; Borriss, R.; Brady, access those biospheres [19, 20]. Those consortia will mostly S.F.; Brakhage, A.A.; Caffrey, P.; Cheng, Y-Q.; Clardy, J.; Cox, likely build strong and reliable Standard Operating Proce- R.J.; De Mot, R.; Donadio, S.; Donia, M.S.; van der Donk, W.A.; dure (SOPs) to extract and purify eDNA for classical and Dorrestein, P.C.; Doyle, S.; Driessen, A.J.M.; Ehling-Schulz, M.; functional metagenomic, which are still lacking currently. Entian, K-D.; Fischbach, M.A.; Gerwick, L.; Gerwick, W.H.; The length of the sequencing data is not long enough yet to Gross, H.; Gust, B.; Hertweck, C.; Höfte, M.; Jensen, S.E.; Ju, J.; Katz, L.; Kaysser, L.; Klassen, J.L.; Keller, N.P.; Kormanec, J.; sequence full BGCs in one reads, but the recent development Kuipers, O.P.; Kuzuyama, T.; Kyrpides, N.C.; Kwon, H-J.; Lautru, of long reads technology such as the SMRT sequencing or S.; Lavigne, R.; Lee, C.Y.; Linquan, B.; Liu, X.; Liu, W.; the nanopore technology will help reach this read length and Luzhetskyy, A.; Mahmud, T.; Mast, Y.; Méndez, C.; Metsä-Ketelä, facilitate the construction of active clones [80, 81]. Further- M.; Micklefield, J.; Mitchell, D.A.; Moore, B.S.; Moreira, L.M.; Müller, R.; Neilan, B.A.; Nett, M.; Nielsen, J.; O’Gara, F.; Oikawa, more, the acquisition of knowledge on bacterial interaction H.; Osbourn, A.; Osburne, M.S.; Ostash, B.; Payne, S.M.; will help us to understand the key player in competition and Pernodet, J-L.; Petricek, M.; Piel, J.; Ploux, O.; Raaijmakers, J.M.; synergism and developed more efficient and stable antimi- Salas, J.A.; Schmitt, E.K.; Scott, B.; Seipke, R.F.; Shen, B.; crobials. Sherman, D.H.; Sivonen, K.; Smanski, M.J.; Sosio, M.; Stegmann, E.; Süssmuth, R.D.; Tahlan, K.; Thomas, C.M.; Tang, Y.; Truman, A.W.; Viaud, M.; Walton, J.D.; Walsh, C.T.; Weber, T.; van CONSENT FOR PUBLICATION Wezel, G.P.; Wilkinson, B.; Willey, J.M.; Wohlleben, W.; Wright, G.D.; Ziemert, N.; Zhang, C.; Zotchev, S.B.; Breitling, R.; Takano, Not applicable. E.; Glöckner, F.O. Minimum Information about a Biosynthetic Gene cluster. Nat. Chem. Biol., 2015, 11(9), 625-631. FUNDING http://dx.doi.org/10.1038/nchembio.1890 PMID: 26284661 [9] Keller, N.P.; Turner, G.; Bennett, J.W. Fungal secondary None. metabolism - from biochemistry to genomics. Nat. Rev. Microbiol., 2005, 3(12), 937-947. http://dx.doi.org/10.1038/nrmicro1286 PMID: 16322742 CONFLICT OF INTEREST [10] Rappé, M.S.; Giovannoni, S.J. The uncultured microbial majority. Annu. Rev. Microbiol., 2003, 57, 369-394. The authors declare no conflict of interest, financial or http://dx.doi.org/10.1146/annurev.micro.57.030502.090759 PMID: otherwise. 14527284 [11] Lagier, J-C.; Dubourg, G.; Million, M.; Cadoret, F.; Bilen, M.; ACKNOWLEDGEMENTS Fenollar, F.; Levasseur, A.; Rolain, J-M.; Fournier, P-E.; Raoult, D. Culturing the human microbiota and culturomics. Nat. Rev. Declared none. Microbiol., 2018, 16, 540-550. http://dx.doi.org/10.1038/s41579-018-0041-0 PMID: 29937540 [12] Bilen, M.; Dufour, J-C.; Lagier, J-C.; Cadoret, F.; Daoud, Z.; REFERENCES Dubourg, G.; Raoult, D. The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. [1] Walsh, C.T.; Wencewicz, T.A. Prospects for new antibiotics: a Microbiome, 2018, 6(1), 94. molecule-centered perspective. J. Antibiot. (Tokyo), 2014, 67(1), 7- http://dx.doi.org/10.1186/s40168-018-0485-5 PMID: 29793532 22. [13] Browne, H.P.; Forster, S.C.; Anonye, B.O.; Kumar, N.; Neville, http://dx.doi.org/10.1038/ja.2013.49 PMID: 23756684 B.A.; Stares, M.D.; Goulding, D.; Lawley, T.D. Culturing of [2] Adu-Oppong, B.; Gasparrini, A.J.; Dantas, G. Genomic and ‘unculturable’ human microbiota reveals novel taxa and extensive functional techniques to mine the microbiome for novel sporulation. Nature, 2016, 533(7604), 543-546. antimicrobials and antimicrobial resistance genes. Ann. N. Y. Acad. http://dx.doi.org/10.1038/nature17645 PMID: 27144353 Sci., 2017, 1388(1), 42-58. [14] Hugenholtz, P.; Goebel, B.M.; Pace, N.R. Impact of culture- http://dx.doi.org/10.1111/nyas.13257 PMID: 27768825 independent studies on the emerging phylogenetic view of bacterial [3] Kinch, M.S.; Patridge, E.; Plummer, M.; Hoyer, D. An analysis of diversity. J. Bacteriol., 1998, 180(18), 4765-4774. FDA-approved drugs for infectious disease: antibacterial agents. http://dx.doi.org/10.1128/JB.180.18.4765-4774.1998 PMID: Drug Discov. Today, 2014, 19(9), 1283-1287. 9733676 http://dx.doi.org/10.1016/j.drudis.2014.07.005 PMID: 25043770 [15] Charlop-Powers, Z.; Owen, J.G.; Reddy, B.V.B.; Ternei, M.A.; [4] Tenover, F.C. Mechanisms of antimicrobial resistance in bacteria. Brady, S.F. Chemical-biogeographic survey of secondary Am. J. Infect. Control, 2006, 34(5)(Suppl. 1), S3-S10. metabolism in soil. Proc. Natl. Acad. Sci. USA, 2014, 111(10), http://dx.doi.org/10.1016/j.ajic.2006.05.219 PMID: 16813980 3757-3762. [5] Newman, D.J.; Cragg, G.M. Natural Products as Sources of New http://dx.doi.org/10.1073/pnas.1318021111 PMID: 24550451 Drugs from 1981 to 2014. J. Nat. Prod., 2016, 79(3), 629-661. [16] Morlon, H.; O’Connor, T.K.; Bryant, J.A.; Charkoudian, L.K.; http://dx.doi.org/10.1021/acs.jnatprod.5b01055 PMID: 26852623 Docherty, K.M.; Jones, E.; Kembel, S.W.; Green, J.L.; Bohannan, [6] Katz, L.; Baltz, R.H. Natural product discovery: past, present, and B.J.M. The Biogeography of Putative Microbial Antibiotic future. J. Ind. Microbiol. Biotechnol., 2016, 43(2-3), 155-176. Production. PLoS One, 2015, 10(6), e0130659. http://dx.doi.org/10.1007/s10295-015-1723-5 PMID: 26739136 http://dx.doi.org/10.1371/journal.pone.0130659 PMID: 26102275 [7] Clardy, J.; Fischbach, M.A.; Currie, C.R. The natural history of [17] Edlund, A.; Loesgen, S.; Fenical, W.; Jensen, P.R. Geographic antibiotics. Curr. Biol., 2009, 19(11), R437-R441. distribution of secondary metabolite genes in the marine http://dx.doi.org/10.1016/j.cub.2009.04.001 PMID: 19515346 actinomycete Salinispora arenicola. Appl. Environ. Microbiol., [8] Medema, M.H.; Kottmann, R.; Yilmaz, P.; Cummings, M.; 2011, 77(17), 5916-5925. Biggins, J.B.; Blin, K.; de Bruijn, I.; Chooi, Y.H.; Claesen, J.; http://dx.doi.org/10.1128/AEM.00611-11 PMID: 21724881 Coates, R.C.; Cruz-Morales, P.; Duddela, S.; Düsterhus, S.; [18] Reddy, B.V.B.; Milshteyn, A.; Charlop-Powers, Z.; Brady, S.F. Edwards, D.J.; Fewer, D.P.; Garg, N.; Geiger, C.; Gomez- eSNaPD: a versatile, web-based bioinformatics platform for Escribano, J.P.; Greule, A.; Hadjithomas, M.; Haines, A.S.; surveying and mining natural product biosynthetic diversity from Helfrich, E.J.N.; Hillwig, M.L.; Ishida, K.; Jones, A.C.; Jones, metagenomes. Chem. Biol., 2014, 21(8), 1023-1033. C.S.; Jungmann, K.; Kegler, C.; Kim, H.U.; Kötter, P.; Krug, D.; http://dx.doi.org/10.1016/j.chembiol.2014.06.007 PMID: 25065533 Masschelein, J.; Melnik, A.V.; Mantovani, S.M.; Monroe, E.A.; [19] Thompson, L.R.; Sanders, J.G.; McDonald, D.; Amir, A.; Ladau, Moore, M.; Moss, N.; Nützmann, H-W.; Pan, G.; Pati, A.; Petras, J.; Locey, K.J.; Prill, R.J.; Tripathi, A.; Gibbons, S.M.; Ackermann, D.; Reen, F.J.; Rosconi, F.; Rui, Z.; Tian, Z.; Tobias, N.J.; G.; Navas-Molina, J.A.; Janssen, S.; Kopylova, E.; Vázquez-Baeza, The Microbiome: A Reservoir to Discover New Antimicrobials Agents Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 1297

Y.; González, A.; Morton, J.T.; Mirarab, S.; Zech Xu, Z.; Jiang, L.; [33] Kaeberlein, T.; Lewis, K.; Epstein, S.S. Isolating “uncultivable” Haroon, M.F.; Kanbar, J.; Zhu, Q.; Jin Song, S.; Kosciolek, T.; microorganisms in pure culture in a simulated natural environment. Bokulich, N.A.; Lefler, J.; Brislawn, C.J.; Humphrey, G.; Owens, Science, 2002, 296(5570), 1127-1129. S.M.; Hampton-Marcell, J.; Berg-Lyons, D.; McKenzie, V.; Fierer, http://dx.doi.org/10.1126/science.1070633 PMID: 12004133 N.; Fuhrman, J.A.; Clauset, A.; Stevens, R.L.; Shade, A.; Pollard, [34] Bollmann, A.; Lewis, K.; Epstein, S.S. Incubation of environmental K.S.; Goodwin, K.D.; Jansson, J.K.; Gilbert, J.A.; Knight, R. Earth samples in a diffusion chamber increases the diversity of recovered Microbiome Project Consortium. A communal catalogue reveals isolates. Appl. Environ. Microbiol., 2007, 73(20), 6386-6390. Earth’s multiscale microbial diversity. Nature, 2017, 551(7681), http://dx.doi.org/10.1128/AEM.01309-07 PMID: 17720826 457-463. [35] Steinert, G.; Whitfield, S.; Taylor, M.W.; Thoms, C.; Schupp, P.J. http://dx.doi.org/10.1038/nature24621 PMID: 29088705 Application of diffusion growth chambers for the cultivation of [20] Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; marine sponge-associated bacteria. Mar. Biotechnol. (NY), 2014, Knight, R.; Gordon, J.I. The human microbiome project. Nature, 16(5), 594-603. 2007, 449(7164), 804-810. http://dx.doi.org/10.1007/s10126-014-9575-y PMID: 24838766 http://dx.doi.org/10.1038/nature06244 PMID: 17943116 [36] Nichols, D.; Cahoon, N.; Trakhtenberg, E.M.; Pham, L.; Mehta, A.; [21] Daniel, R. The soil metagenome--a rich resource for the discovery Belanger, A.; Kanigan, T.; Lewis, K.; Epstein, S.S. Use of ichip for of novel natural products. Curr. Opin. Biotechnol., 2004, 15(3), high-throughput in situ cultivation of “uncultivable” microbial 199-204. species. Appl. Environ. Microbiol., 2010, 76(8), 2445-2450. http://dx.doi.org/10.1016/j.copbio.2004.04.005 PMID: 15193327 http://dx.doi.org/10.1128/AEM.01754-09 PMID: 20173072 [22] Adnan, M.; Alshammari, E.; Patel, M.; Amir Ashraf, S.; Khan, S.; [37] Piddock, L.J.V. Teixobactin, the first of a new class of antibiotics Hadi, S. Significance and potential of marine microbial natural discovered by iChip technology? J. Antimicrob. Chemother., 2015, bioactive compounds against biofilms/biofouling: necessity for 70(10), 2679-2680. green chemistry. PeerJ, 2018, 6, e5049. http://dx.doi.org/10.1093/jac/dkv175 PMID: 26089440 http://dx.doi.org/10.7717/peerj.5049 PMID: 29967730 [38] Momeni, B.; Chen, C-C.; Hillesland, K.L.; Waite, A.; Shou, W. [23] Donia, M.S.; Cimermancic, P.; Schulze, C.J.; Wieland Brown, Using artificial systems to explore the ecology and evolution of L.C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R.G.; symbioses. Cell. Mol. Life Sci., 2011, 68(8), 1353-1368. Fischbach, M.A. A systematic analysis of biosynthetic gene http://dx.doi.org/10.1007/s00018-011-0649-y PMID: 21424911 clusters in the human microbiome reveals a common family of [39] Tanouchi, Y.; Smith, R.P.; You, L. Engineering microbial systems antibiotics. Cell, 2014, 158(6), 1402-1414. to explore ecological and evolutionary dynamics. Curr. Opin. http://dx.doi.org/10.1016/j.cell.2014.08.032 PMID: 25215495 Biotechnol., 2012, 23(5), 791-797. [24] Borchert, E.; Jackson, S.A.; O’Gara, F.; Dobson, A.D.W. Diversity http://dx.doi.org/10.1016/j.copbio.2012.01.006 PMID: 22310174 of natural product biosynthetic genes in the microbiome of the deep [40] Lodhi, A.F.; Zhang, Y.; Adil, M.; Deng, Y. Antibiotic discovery: sea sponges inflatella pellicula, poecillastra compressa, and combining isolation chip (iChip) technology and co-culture Stelletta normani. Front. Microbiol., 2016, 7, 1027. technique. Appl. Microbiol. Biotechnol., 2018, 102(17), 7333-7341. http://dx.doi.org/10.3389/fmicb.2016.01027 PMID: 27446062 http://dx.doi.org/10.1007/s00253-018-9193-0 PMID: 29974183 [25] Radjasa, O.; Wiese, J.; Sabdono, A.; Imhoff, J.F. Corals as source [41] Yang, E.; Fan, L.; Yan, J.; Jiang, Y.; Doucette, C.; Fillmore, S.; of bacteria with antimicrobial activity. J. Coast. Develop., 2008, Walker, B. Influence of culture media, pH and temperature on 11, 121-130. growth and bacteriocin production of bacteriocinogenic lactic acid [26] Kelman, D. Antimicrobial Activity of Sponges and Corals.Coral bacteria. AMB Express, 2018, 8(1), 10. Health and Disease; Rosenberg, E.; Loya, Y., Eds.; Springer http://dx.doi.org/10.1186/s13568-018-0536-0 PMID: 29368243 Berlin, 2004, pp. 243-258. [42] Wiedmann, M.; Carroll, L.M. Next-generation sequencing. http://dx.doi.org/10.1007/978-3-662-06414-6_12 Encyclo. Food Chem., 2018, p. 376. [27] Wilson, M.C.; Mori, T.; Rückert, C.; Uria, A.R.; Helf, M.J.; [43] Héry-Arnaud, G.; Boutin, S.; Cuthbertson, L.; Elborn, S.J.; Tunney, Takada, K.; Gernert, C.; Steffens, U.A.E.; Heycke, N.; Schmitt, S.; M.M. The lung and gut microbiome: what has to be taken into Rinke, C.; Helfrich, E.J.N.; Brachmann, A.O.; Gurgui, C.; consideration for cystic fibrosis? J. Cyst. Fibros., 2019, 18(1), 13- Wakimoto, T.; Kracht, M.; Crüsemann, M.; Hentschel, U.; Abe, I.; 21. Matsunaga, S.; Kalinowski, J.; Takeyama, H.; Piel, J. An http://dx.doi.org/10.1016/j.jcf.2018.11.003 PMID: 30487080 environmental bacterial taxon with a large and distinct metabolic [44] Blin, K.; Kim, H.U.; Medema, M.H.; Weber, T. Recent repertoire. Nature, 2014, 506(7486), 58-62. development of antismash and other computational approaches to http://dx.doi.org/10.1038/nature12959 PMID: 24476823 mine secondary metabolite biosynthetic gene clusters. Brief. [28] Charusanti, P.; Fong, N.L.; Nagarajan, H.; Pereira, A.R.; Li, H.J.; Bioinform., 2019, 20(4), 1103-1113. Abate, E.A.; Su, Y.; Gerwick, W.H.; Palsson, B.O. Exploiting PMID: 29112695 adaptive laboratory evolution of Streptomyces clavuligerus for [45] Weber, T.; Blin, K.; Duddela, S.; Krug, D.; Kim, H.U.; Bruccoleri, antibiotic discovery and overproduction. PLoS One, 2012, 7(3), R.; Lee, S.Y.; Fischbach, M.A.; Müller, R.; Wohlleben, W.; e33727. Breitling, R.; Takano, E.; Medema, M.H. antiSMASH 3.0-a http://dx.doi.org/10.1371/journal.pone.0033727 PMID: 22470465 comprehensive resource for the genome mining of biosynthetic [29] Laskaris, P.; Tolba, S.; Calvo-Bado, L.; Wellington, E.M. gene clusters. Nucleic Acids Res., 2015, 43(W1), W237-243. Coevolution of antibiotic production and counter-resistance in soil http://dx.doi.org/10.1093/nar/gkv437 PMID: 25948579 bacteria. Environ. Microbiol., 2010, 12(3), 783-796. [46] Cimermancic, P.; Medema, M.H.; Claesen, J.; Kurita, K.; Wieland http://dx.doi.org/10.1111/j.1462-2920.2009.02125.x PMID: Brown, L.C.; Mavrommatis, K.; Pati, A.; Godfrey, P.A.; Koehrsen, 20067498 M.; Clardy, J.; Birren, B.W.; Takano, E.; Sali, A.; Linington, R.G.; [30] Berdy, B.; Spoering, A.L.; Ling, L.L.; Epstein, S.S. In situ Fischbach, M.A. Insights into secondary metabolism from a global cultivation of previously uncultivable microorganisms using the analysis of prokaryotic biosynthetic gene clusters. Cell, 2014, ichip. Nat. Protoc., 2017, 12(10), 2232-2242. 158(2), 412-421. http://dx.doi.org/10.1038/nprot.2017.074 PMID: 29532802 http://dx.doi.org/10.1016/j.cell.2014.06.034 PMID: 25036635 [31] Jung, D.; Seo, E-Y.; Epstein, S.S.; Joung, Y.; Han, J.; Parfenova, [47] Quince, C.; Walker, A.W.; Simpson, J.T.; Loman, N.J.; Segata, N. V.V.; Belykh, O.I.; Gladkikh, A.S.; Ahn, T.S. Application of a new Shotgun metagenomics, from sampling to analysis. Nat. cultivation technology, I-tip, for studying microbial diversity in Biotechnol., 2017, 35(9), 833-844. freshwater sponges of Lake Baikal, Russia. FEMS Microbiol. http://dx.doi.org/10.1038/nbt.3935 PMID: 28898207 Ecol., 2014, 90(2), 417-423. [48] Sczyrba, A.; Hofmann, P.; Belmann, P.; Koslicki, D.; Janssen, S.; http://dx.doi.org/10.1111/1574-6941.12399 PMID: 25078251 Dröge, J.; Gregor, I.; Majda, S.; Fiedler, J.; Dahms, E.; Bremges, [32] Stevenson, B.S.; Eichorst, S.A.; Wertz, J.T.; Schmidt, T.M.; A.; Fritz, A.; Garrido-Oter, R.; Jørgensen, T.S.; Shapiro, N.; Blood, Breznak, J.A. New strategies for cultivation and detection of P.D.; Gurevich, A.; Bai, Y.; Turaev, D.; DeMaere, M.Z.; Chikhi, previously uncultured microbes. Appl. Environ. Microbiol., 2004, R.; Nagarajan, N.; Quince, C.; Meyer, F.; Balvočiūtė, M.; Hansen, 70(8), 4748-4755. L.H.; Sørensen, S.J.; Chia, B.K.H.; Denis, B.; Froula, J.L.; Wang, http://dx.doi.org/10.1128/AEM.70.8.4748-4755.2004 PMID: Z.; Egan, R.; Don Kang, D.; Cook, J.J.; Deltel, C.; Beckstette, M.; 15294811 Lemaitre, C.; Peterlongo, P.; Rizk, G.; Lavenier, D.; Wu, Y-W.; 1298 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 Boutin and Dalpke

Singer, S.W.; Jain, C.; Strous, M.; Klingenberg, H.; Meinicke, P.; http://dx.doi.org/10.1073/pnas.89.18.8794 PMID: 1528894 Barton, M.D.; Lingner, T.; Lin, H-H.; Liao, Y-C.; Silva, G.G.Z.; [63] Kim, J.H.; Feng, Z.; Bauer, J.D.; Kallifidas, D.; Calle, P.Y.; Brady, Cuevas, D.A.; Edwards, R.A.; Saha, S.; Piro, V.C.; Renard, B.Y.; S.F. Cloning large natural product gene clusters from the Pop, M.; Klenk, H-P.; Göker, M.; Kyrpides, N.C.; Woyke, T.; environment: piecing environmental DNA gene clusters back Vorholt, J.A.; Schulze-Lefert, P.; Rubin, E.M.; Darling, A.E.; together with TAR. Biopolymers, 2010, 93(9), 833-844. Rattei, T.; McHardy, A.C. Critical Assessment of Metagenome http://dx.doi.org/10.1002/bip.21450 PMID: 20577994 Interpretation-a benchmark of metagenomics software. Nat. [64] Lambalot, R.H.; Gehring, A.M.; Flugel, R.S.; Zuber, P.; LaCelle, Methods, 2017, 14(11), 1063-1071. M.; Marahiel, M.A.; Reid, R.; Khosla, C.; Walsh, C.T. A new http://dx.doi.org/10.1038/nmeth.4458 PMID: 28967888 enzyme superfamily - the phosphopantetheinyl transferases. Chem. [49] Ji, P.; Zhang, Y.; Wang, J.; Zhao, F. MetaSort untangles Biol., 1996, 3(11), 923-936. metagenome assembly by reducing microbial community http://dx.doi.org/10.1016/S1074-5521(96)90181-7 PMID: 8939709 complexity. Nat. Commun., 2017, 8, 14306. [65] Charlop-Powers, Z.; Banik, J.J.; Owen, J.G.; Craig, J.W.; Brady, http://dx.doi.org/10.1038/ncomms14306 PMID: 28112173 S.F. Selective enrichment of environmental DNA libraries for [50] Uritskiy, G.V.; DiRuggiero, J.; Taylor, J. MetaWRAP-a flexible genes encoding nonribosomal peptides and polyketides by pipeline for genome-resolved metagenomic data analysis. phosphopantetheine transferase-dependent complementation of Microbiome, 2018, 6(1), 158. siderophore biosynthesis. ACS Chem. Biol., 2013, 8(1), 138-143. http://dx.doi.org/10.1186/s40168-018-0541-1 PMID: 30219103 http://dx.doi.org/10.1021/cb3004918 PMID: 23072412 [51] Ziemert, N.; Podell, S.; Penn, K.; Badger, J.H.; Allen, E.; Jensen, [66] National Research Council. Committee on metagenomics: P.R. The natural product domain seeker NaPDoS: a phylogeny challenges and functional applications. In: New science of based bioinformatic tool to classify secondary metabolite gene metagenomics: Revealing the secrets of our microbial planet; diversity. PLoS One, 2012, 7(3)e34064 National Academies Press (US): Washington, 2007. http://dx.doi.org/10.1371/journal.pone.0034064 PMID: 22479523 [67] Warren, R.L.; Freeman, J.D.; Levesque, R.C.; Smailus, D.E.; [52] Woodhouse, J.N.; Fan, L.; Brown, M.V.; Thomas, T.; Neilan, B.A. Flibotte, S.; Holt, R.A. Transcription of foreign DNA in Deep sequencing of non-ribosomal peptide synthetases and escherichia coli. Genome Res., 2008, 18(11), 1798-1805. polyketide synthases from the microbiomes of Australian marine http://dx.doi.org/10.1101/gr.080358.108 PMID: 18701636 sponges. ISME J., 2013, 7(9), 1842-1851. [68] Gaida, S.M.; Sandoval, N.R.; Nicolaou, S.A.; Chen, Y.; http://dx.doi.org/10.1038/ismej.2013.65 PMID: 23598791 Venkataramanan, K.P.; Papoutsakis, E.T. Expression of [53] Hover, B.M.; Kim, S-H.; Katz, M.; Charlop-Powers, Z.; Owen, heterologous sigma factors enables functional screening of J.G.; Ternei, M.A.; Maniko, J.; Estrela, A.B.; Molina, H.; Park, S.; metagenomic and heterologous genomic libraries. Nat. Commun., Perlin, D.S.; Brady, S.F. Culture-independent discovery of the 2015, 6, 7045. malacidins as calcium-dependent antibiotics with activity against http://dx.doi.org/10.1038/ncomms8045 PMID: 25944046 multidrug-resistant Gram-positive pathogens. Nat. Microbiol., [69] Stevens, D.C.; Conway, K.R.; Pearce, N.; Villegas-Peñaranda, 2018, 3(4), 415-422. L.R.; Garza, A.G.; Boddy, C.N. Alternative sigma factor over- http://dx.doi.org/10.1038/s41564-018-0110-1 PMID: 29434326 expression enables heterologous expression of a type II polyketide [54] Sélem-Mojica, N.; Aguilar, C.; Gutiérrez-García, K.; Martínez- biosynthetic pathway in Escherichia coli. PLoS One, 2013, 8(5), Guerrero, C.E.; Barona-Gómez, F. Evomining reveals the origin e64858. and fate of natural products biosynthetic enzymes. Microb http://dx.doi.org/10.1371/journal.pone.0064858 PMID: 23724102 Genom., 2019, 5(12), e000260. [70] Baltz, R.H. Streptomyces and Saccharopolyspora hosts for DOI: 10.1099/mgen.0.000260 heterologous expression of secondary metabolite gene clusters. J. [55] Cruz-Morales, P.; Kopp, J.F.; Martínez-Guerrero, C.; Yáñez- Ind. Microbiol. Biotechnol., 2010, 37(8), 759-772. Guerra, L.A.; Selem-Mojica, N.; Ramos-Aboites, H.; Feldmann, J.; http://dx.doi.org/10.1007/s10295-010-0730-9 PMID: 20467781 Barona-Gómez, F. Phylogenomic analysis of natural products [71] McMahon, M.D.; Guan, C.; Handelsman, J.; Thomas, M.G. biosynthetic gene clusters allows discovery of arseno-organic Metagenomic analysis of Streptomyces lividans reveals host- metabolites in model streptomycetes. Genome Biol. Evol., 2016, dependent functional expression. Appl. Environ. Microbiol., 2012, 8(6), 1906-1916. 78(10), 3622-3629. http://dx.doi.org/10.1093/gbe/evw125 PMID: 27289100 http://dx.doi.org/10.1128/AEM.00044-12 PMID: 22427497 [56] Banik, J.J.; Brady, S.F. Recent application of metagenomic [72] Stevens, D.C.; Henry, M.R.; Murphy, K.A.; Boddy, C.N. approaches toward the discovery of antimicrobials and other Heterologous expression of the oxytetracycline biosynthetic bioactive small molecules. Curr. Opin. Microbiol., 2010, 13(5), pathway in Myxococcus xanthus. Appl. Environ. Microbiol., 2010, 603-609. 76(8), 2681-2683. http://dx.doi.org/10.1016/j.mib.2010.08.012 PMID: 20884282 http://dx.doi.org/10.1128/AEM.02841-09 PMID: 20208031 [57] Katz, M.; Hover, B.M.; Brady, S.F. Culture-independent discovery [73] Luo, Y.; Enghiad, B.; Zhao, H. New tools for reconstruction and of natural products from soil metagenomes. J. Ind. Microbiol. heterologous expression of natural product biosynthetic gene Biotechnol., 2016, 43(2-3), 129-141. clusters. Nat. Prod. Rep., 2016, 33(2), 174-182. http://dx.doi.org/10.1007/s10295-015-1706-6 PMID: 26586404 http://dx.doi.org/10.1039/C5NP00085H PMID: 26647833 [58] Yuan, S.; Cohen, D.B.; Ravel, J.; Abdo, Z.; Forney, L.J. Evaluation [74] Komatsu, M.; Uchiyama, T.; Ōmura, S.; Cane, D.E.; Ikeda, H. of methods for the extraction and purification of DNA from the Genome-minimized Streptomyces host for the heterologous human microbiome. PLoS One, 2012, 7(3), e33865. expression of secondary metabolism. Proc. Natl. Acad. Sci. USA, http://dx.doi.org/10.1371/journal.pone.0033865 PMID: 22457796 2010, 107(6), 2646-2651. [59] Schloss, P.D.; Handelsman, J. Biotechnological prospects from http://dx.doi.org/10.1073/pnas.0914833107 PMID: 20133795 metagenomics. Curr. Opin. Biotechnol., 2003, 14(3), 303-310. [75] Zhou, K.; Qiao, K.; Edgar, S.; Stephanopoulos, G. Distributing a http://dx.doi.org/10.1016/S0958-1669(03)00067-3 PMID: metabolic pathway among a microbial consortium enhances 12849784 production of natural products. Nat. Biotechnol., 2015, 33(4), 377- [60] Zhou, J.; Bruns, M.A.; Tiedje, J.M. DNA recovery from soils of 383. diverse composition. Appl. Environ. Microbiol., 1996, 62(2), 316- http://dx.doi.org/10.1038/nbt.3095 PMID: 25558867 322. [76] Rondon, M.R.; August, P.R.; Bettermann, A.D.; Brady, S.F.; http://dx.doi.org/10.1128/AEM.62.2.316-322.1996 PMID: 8593035 Grossman, T.H.; Liles, M.R.; Loiacono, K.A.; Lynch, B.A.; [61] Brady, S.F. Construction of soil environmental DNA cosmid MacNeil, I.A.; Minor, C.; Tiong, C.L.; Gilman, M.; Osburne, M.S.; libraries and screening for clones that produce biologically active Clardy, J.; Handelsman, J.; Goodman, R.M. Cloning the soil small molecules. Nat. Protoc., 2007, 2(5), 1297-1305. metagenome: a strategy for accessing the genetic and functional http://dx.doi.org/10.1038/nprot.2007.195 PMID: 17546026 diversity of uncultured microorganisms. Appl. Environ. Microbiol., [62] Shizuya, H.; Birren, B.; Kim, U.J.; Mancino, V.; Slepak, T.; 2000, 66(6), 2541-2547. Tachiiri, Y.; Simon, M. Cloning and stable maintenance of 300- http://dx.doi.org/10.1128/AEM.66.6.2541-2547.2000 PMID: kilobase-pair fragments of human DNA in Escherichia coli using 10831436 an F-factor-based vector. Proc. Natl. Acad. Sci. USA, 1992, 89(18), [77] Lim, H.K.; Chung, E.J.; Kim, J-C.; Choi, G.J.; Jang, K.S.; Chung, 8794-8797. Y.R.; Cho, K.Y.; Lee, S-W. Characterization of a forest soil The Microbiome: A Reservoir to Discover New Antimicrobials Agents Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 14 1299

metagenome clone that confers indirubin and indigo production on [80] Levene, M.J.; Korlach, J.; Turner, S.W.; Foquet, M.; Craighead, Escherichia coli. Appl. Environ. Microbiol., 2005, 71(12), 7768- H.G.; Webb, W.W. Zero-mode waveguides for single-molecule 7777. analysis at high concentrations. Science, 2003, 299(5607), 682-686. http://dx.doi.org/10.1128/AEM.71.12.7768-7777.2005 PMID: http://dx.doi.org/10.1126/science.1079700 PMID: 12560545 16332749 [81] Branton, D.; Deamer, D.W.; Marziali, A.; Bayley, H.; Benner, [78] Brady, S.F.; Clardy, J. Long-chain n-acyl amino acid antibiotics S.A.; Butler, T.; Di Ventra, M.; Garaj, S.; Hibbs, A.; Huang, X.; isolated from heterologously expressed environmental DNA. J. Am. Jovanovich, S.B.; Krstic, P.S.; Lindsay, S.; Ling, X.S.; Chem. Soc., 2000, 122, 12903-12904. Mastrangelo, C.H.; Meller, A.; Oliver, J.S.; Pershin, Y.V.; Ramsey, http://dx.doi.org/10.1021/ja002990u J.M.; Riehn, R.; Soni, G.V.; Tabard-Cossa, V.; Wanunu, M.; [79] Brahami, A.; Castonguay, A.; Déziel, É. Novel ‘bacteriospray’ Wiggin, M.; Schloss, J.A. The potential and challenges of nanopore method facilitates the functional screening of metagenomic sequencing. Nat. Biotechnol., 2008, 26(10), 1146-1153. libraries for antimicrobial activity. Methods Protoc, 2019, 2(1), 4. http://dx.doi.org/10.1038/nbt.1495 PMID: 18846088 http://dx.doi.org/10.3390/mps2010004 PMID: 31164589