Skip to main content
  • Review Article
  • Published:

Current strategies to induce secondary metabolites from microbial biosynthetic cryptic gene clusters

Abstract

The genome of actinomycetes and several other microorganisms are endowed with many cryptic gene clusters that can code for previously undetected, a plethora of complex secondary metabolites. Under standard laboratory controlled conditions, the genes regulating these biosynthetic clusters are expressed at very low levels or remain phenotypically cryptic (silent). Over the past several decades, multi-drug-resistant bacteria have been observed with increased frequency, posing a significant threat to human health worldwide. The present alarming situation urgently calls for concerted global efforts for the discovery of new antimicrobials. The present situation, if not controlled, will take us again to the pre-antibiotic era. Today, in the post-genomic era, various new strategies such as the activation of cryptic gene clusters in microorganisms rejuvenate a new conviction in the field of natural product research that may lead to the identification of yet-unidentified novel secondary metabolites of therapeutic and other use. Decryptification of this versatile endogenous genetic reservoir may provide in the near future the more concrete rationale for antibiotic discovery. The present review is an attempt to provide a comprehensive detail, outlining current strategies that have been shown successful to activate cryptic biosynthetic gene clusters in microorganisms.

Introduction

Microorganisms as a source of secondary metabolites

The introduction of penicillin antibiotic led researchers to explore microorganisms for the production of secondary metabolites. The parvome (small bioactive molecules) is considered to be an inexhaustible source of secondary metabolites of microbial origin (Davies 2011). To date, various metabolite molecules of therapeutic and industrial use (Fig. 1) have been identified from microbial cell factories (Bentley 1997; Berdy 2012). The intense investigations in the last 80 years have resulted in the screening of many microorganisms particularly the genera of actinomycetes (53%), and fungi (30%) have attracted increased attention in the search for novel secondary metabolites. A substantial number of secondary metabolites (17%) are also produced by Bacillus, Pseudomonas, myxobacteria, and cyanobacteria (Berdy 2005). Interestingly, harnessing the microbial capacity to obtain secondary metabolites has an added advantage because of their distinctive molecular scaffolds that are not commonly found in other chemical libraries (Feher and Schmidt 2003). For decades, the specialized secondary metabolism of these microorganisms has been exploited for the screening and production of metabolites at the commercial level. A sketch has been provided to illustrate the overall screening operation and the different stages of antibiotic discovery (Fig. 2).

Fig. 1
figure 1

The microbial parvome: the world of small bioactive molecules

Fig. 2
figure 2

The different stages of antibiotic discovery from microorganisms

Microbial diversity

The rate of the discovery of novel natural compounds from microorganisms has been diminished dramatically. Moreover, the most of antibiotic screening programs repeatedly rediscover the already known secondary metabolites. For this reason, it is speculated that microorganisms have been exhausted for their specialized metabolic wealth. The rapid increase in multi-drug-resistant pathogens also requires the global attention of researchers for the development of novel antimicrobials, novel treatment options, and alternative antimicrobial therapies. During the million years of evolutionary development, microorganisms acquired the drug resistance via lateral or vertical transmission, but at the same time, these microbes had the equal chance to adapt, counteract, and resist these resistant pathogens by producing more effective, more potent, “anti-resistant” secondary metabolites. The nature remains the richest and versatile source of new antibiotics; therefore, to discover yet to be identified secondary metabolites, genuine efforts are needed to exploit the unexploited and unexplored microorganisms from natural habitats. This hope rejuvenates a renewed curiosity to explore orphan pathways or cryptic gene clusters in microorganisms to search for novel metabolites. Based on the evidence collected from various sources, it has been suggested that ca. 300,000 to 1 million species of bacteria exist on the earth (Allsopp et al. 1995; Tiedje 1994). Around 70,000 fungal strains have been identified and assumed to be present at the rate of 1.5 × 106 species (Hawksworth 1991). It has been estimated that a gram of soil may contain 8.3 million prokaryotic species, while the total number of prokaryotic cells present in the natural habitats has been considered to be 4 to 6 × 1030 cells (Fierer and Jackson 2006; Torsvik and Ovreas 2002; Whitman et al. 1998). The majority of cells obtained from soil samples and visualized by microscopy are viable, but they do not generally form visible colonies on the media that contains agar (Eilers et al. 2000; Xu et al. 1982). Intriguingly, to date, only few thousands of prokaryotic species have been formally described, which means that the vast majority of prokaryotes (90–99%) present in natural habitats have still to be identified (Harwani 2013; Rappe and Giovannoni 2003; Schloss and Handelsman 2004; Zengler et al. 2002).

Need for novel antimicrobials

A major public health concern is the mortality rate due to the multi-drug-resistant bacteria. A total of 1.7 million infections and ca. 99,000 deaths have been calculated to occur each year in the USA itself (Klevens et al. 2007). The strains of Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. (ESKAPE) have been categorized as multi-drug resistant (MDR) pathogens (Rice 2006). In addition to ESKAPE, the emergence of MDR Clostridium difficile and members of Enterobacteriaceae family has also been reported (Peterson 2009). The nosocomial infections caused by methicillin-resistant S. aureus (MRSA) and vancomycin-resistant enterococci (VRE) have also contributed in increased morbidity and mortality. The infections caused by Gram-negative, MDR bacteria, such as Pseudomonas aeruginosa, Acinetobacter baumannii, extended-spectrum 훽-lactamase-producing Enterobacteriaceae, and carbapenem-resistant Enterobacteriaceae have been also found to pose significant threat to human health. The development of MDR in bacterial pathogens may be attributed to the transfer of resistance genes from Klebsiella pneumoniae and New Delhi metallo-훽-lactamase (NDM-1) strains (Kallel et al. 2006; Rice 2008). Therefore, stronger actions are urgently needed to develop new antibiotics and new alternative therapies to effectively treat MDR pathogens.

Strategies for the activation of novel natural products from cryptic gene clusters

One strain many compounds (OSMAC)

The term OSMAC was initially used by Zeeck and co-workers that simply states that a single strain is capable of producing different metabolic compounds under the different fermentation conditions (Bode et al. 2002). The growth parameters that can be manipulated in OSMAC include media composition (incorporating different carbon, phosphate, and nitrogen sources), aeration rate, temperature, pH, inclusion of inhibitors or growth enhancers, etc. It is hypothesized in several reports that the prevailing growth environment has an intense effect on the production of microbial metabolites. A minute change in the growth parameter (such as variable concentrations of phosphate) may induce higher amount of specific secondary metabolite or may result in a completely novel type (Bode et al. 2002; Martin 2004; Miaomiao et al. 2017; Romano et al. 2014). The approach has been successfully applied to induce the cryptic coelichelin biosynthetic gene cluster in S. coelicolor (Ikeda et al. 1999) and to identify the cryptic fungal biosynthetic gene clusters (Daletos et al. 2017).

Culturing the unculturables

The term “unculturable” is used to describe organisms, particularly bacteria that do not grow on artificial media. In other words, we do not have sufficient biological information to culture these bacteria in vitro. It has been proposed that these microscopic cells are dead and therefore would never grow in the laboratory (Spieckermann 1912). In fact, many of these cells have been shown to be metabolically active (Roszak and Colwell 1987). An estimate suggests that the number of antibiotics from these uncultured sources is two to three orders of magnitude, greater than those from the cultured sources. The easiest way to study and exploit the unculturables is through having these available in artificial culture in the laboratory (Nichols et al. 2010). The cryptic antibiotic gene clusters in yet-to-be-characterized microorganisms remain unexplored because the environmental clues that elicit their activation are unknown. The fact that the overwhelming part of microbial diversity remains uncultivated therefore represents an exciting opportunity to make attempts to culture them to search for novel secondary metabolites. This renewed interest in microbial cultivation (Aoi et al. 2009; Connon and Giovannoni 2002; Ferrari and Gillings 2009; Ferrari et al. 2005; Kaeberlein et al. 2002; Stevenson et al. 2004) has led to the development of several innovative approaches to bring new microbial species in the culture. Most of these approaches share one basic strategy that is to mimic the environment of the target microorganism. An ultimate move in this direction is to replace in vitro growth by in vivo cultivation in the natural habitat. Recently, the discovery of a novel antibiotic teixobactin has been announced from a previously uncultured and undescribed soil bacteria belonging to β-proteobacteria, provisionally named as Eleftheria terrae, using isolation chip (iChip) method (Ling et al. 2015; Piddock 2015; Wright 2015).

Overexpression of pathway-specific gene regulators

During transition from late-exponential to stationary growth phase, the overexpression of antibiotic regulatory protein in Streptomyces has been reported to increase the antibiotic yield (Bibb 2005). A unique class of regulatory gene has been identified by Laureti group encoding protein similar to the large ATP-binding regulators of the LuxR family (LAL) (Laureti et al. 2011). The constitutive expression of a pathway-specific, LAL regulator of type-I modular polyketide synthase (PKS) gene cluster in Streptomyces ambofaciens resulted in the production of stambomycins. Similarly, the overexpression of afsS (a 63-amino acid protein) induces selective antibiotic production in S. coelicolor (Matsumoto et al. 1995), S. lividans (Vögtli et al. 1994), S. avermitilis (Lee et al. 2000). The overexpression of afsR has also been reported to induce the antibiotic production in S. coelicolor (Floriano and Bibb 1996), S. peucetius (Parajuli et al. 2005), and S. venezuelae (Maharjan et al. 2009).

Ribosome or RNA polymerase engineering

Ochi group developed a method termed “ribosome engineering” in which the ribosomal protein S12 or RNA polymerase (RNAP) have been targeted to increase the antibiotic production in bacteria (Ochi et al. 2004). The idea was to collect ribosomal mutants and to see whether the gene expression for antibiotic production due to alteration in the transcription or translation processes has been affected or not. The ribosomal mutants, conferring antibiotic resistance, were collected by selecting them on the agar medium containing streptomycin antibiotic. Similarly, RNA polymerase (RNAP) mutants were collected by growing cells on the agar medium containing rifampicin (that binds to RNAP to inhibit RNA synthesis). These mutants have been observed not only for their ability to produce increased amounts of antibiotics but also led to the production of novel antibiotics (Hoshaka et al. 2009). Using this approach, out of “no or sub-optimal” levels of antibiotic-producing 1068 soil bacteria, 6% actinomycetes and 43% Streptomyces species have been identified to induce antibiotic production. The detailed study revealed that augmentation of antibiotic production was due to a mutation at Lys-88 to Glu or Arg in the ribosomal protein S12 that enhances protein synthesis at the stationary-phase growth condition. Consequently, a mutation His 437 to Asp or Leu in the RNAP β-subunit has also been found to increase its promoter binding affinity. Other antibiotics such as erythromycin and gentamicin, both of which target ribosome, have also been demonstrated to exhibit the same level of enhancement in the antibiotic production (Chai et al. 2012; Imai et al. 2012; 2015).

Isotope-guided fractionation

The approach first introduced by Gross and colleague’s uses genome sequence analysis in combination with isotope-guided fractionation to identify unknown compounds coded by cryptic gene clusters (Gross et al. 2007). The strategy has been successfully applied to predict orfamides, the byproduct of cryptic NRPS domain, using bioinformatics analysis of Pseudomonas fluorescens genome (Paulsen et al. 2005). The approach has been also found useful in inferring the amino acid substrate used by the adenylation A domain of bacterial non-ribosomal polyketide synthatase (NRPS) (Rausch et al. 2005). Using the approach, leucine was predicted to be incorporated into four different A domains of the ofa cluster in Pseudomonas fluorescens. Since 15N-L-leucine can be readily detected by 1H-15N Heteronuclear Multiple Bond Correlation (HMBC)-NMR, it was introduced in the exponentially growing cultures, resulting in the induction of orgamide A compound from ofa cluster. The isotope-guided fractionation has the potential to rapidly identify novel metabolites without using genetic manipulations. However, the approach requires optimum levels of expression of the biosynthetic gene cluster under investigation.

Heterologous expression of biosynthetic gene cluster

The introduction of complex PKS and non-ribosomal peptide synthetase (NRPS) gene clusters in heterologous hosts is technically a difficult task. The large size of these megasynthase genes is of major concern. Moreover, it is also difficult to replace a native promoter with a strong promoter that is to be induced in the host for maximizing expression. If successfully expressed, the introduced gene cluster may produce a completely new set of the metabolic products. Using this methodology, Nihira group have engineered A. oryzae strain, by introducing a full citrinin biosynthetic gene cluster from Monascus purpureus that produces citrinin via ctnA activator. This is one of the best examples of cluster-specific activation of heterologous genes that may be used for the other cryptic genetic systems (Sakai et al. 2008).

Transcriptional activators and repressors

Several secondary metabolite gene clusters contain one or more genes that encode transcription factors. A co-occurrence of the LysR-Type transcriptional regulator (LTTR) family with antibiotic synthetic gene clusters has been reported in several articles (Colombo et al. 2001; Rodriguez et al. 2008; Waldron et al. 2001). LTTRs have been found to be the most abundant among Actinobacteria, Proteobacteria, and Firmicutes. These genetic regulators typically have two domains consisted of an N-terminal DNA binding domain and a C-terminal ligand-binding domain (Maddocks and Oyston 2008). In the absence of a signal, they remain on inactive state and upon interaction, they activate the concerned promoters. It is an interesting feature to retain the promoter of cryptic antibiotic gene cluster and shut it down in the absence of a signal. The activation of this type was reported by Rodriguez et al. (Rodriguez et al. 2008) where the synthesis of β-lactam antibiotic “thienamycin” in Streptomyces cattleya was observed to be induced by ThnI protein. Similarly, the LysR protein, designated as ORF-L16, induced spinomycin biosynthesis in Saccharopolyspora spinosa (Waldron et al. 2001). The aur1 polyketide gene cluster led to the biosynthesis of angucycline-like antibiotic auricin in S. aureofaciens (Novakova et al. 2011). The other example includes the cyclic AMP receptor protein (Crp) (catabolite repressor) that has been demonstrated to regulate the antibiotic production in Streptomyces (Gao et al. 2012). The overexpression of crp resulted in the production of antibiotic in Streptomyces species. The pathway-specific regulatory gene scbR2 (encoding c-butyrolactone receptor) in S. coelicolor induced the production of a yellow-pigmented secondary metabolite (yCPK) (Gottelt et al. 2010). On the other hand, deactivation of the suppressor proteins (transcriptional proteins that result in the negative regulation) has been also observed to turn on the silent antibiotic gene clusters. The repression of antibiotic clusters exerts the transcriptional and post-translational control, and the best example in this category is the production of the cryptic orsellinic acid in A. nidulans (Bok et al. 2013).

Synthetic biology

The synthetic manipulations of natural metabolites at the chemical level have marginally decreased its requirement from natural sources. The synthetic biology has developed a global interest in the natural product discovery (Medema et al. 2011a, 2011b; Wright 2014), and the field of “directed evolution” has an immense potential to accelerate this process (Breitling and Takano 2015). At the same time, the genetic manipulation of antibiotic production in heterologous hosts using the tools of synthetic biology has also revealed the utility of priming metabolic machinery for increased production of secondary metabolites (Helfrich et al. 2014). Keasling et al. published an interesting procedure in which plant and bacterial-derived genes were combined and engineered in E. coli and S. cerevisiae to produce amorphadiene and artemisinic acid (precursors of the potent antimalaria drug artemisinin) respectively (Dietrich et al. 2009; Martin et al. 2003; Newman et al. 2006; Ro et al. 2006). The approach has led to the discovery of 74 novel metabolites in yeast (Klein et al. 2014). The technical challenge involved, in generating multiple cluster variants that can express antibiotic gene, is its high cost per compound (Bachmann et al. 2014).

Functional proteomics

At the present time, proteomics tools have been successfully utilized in the identification of orphan gene clusters. By comparing protein profiles of pyomelanin-producing and non-producing strains of A. fumigates, the pyomelanin synthesis has been reported (Schmaler-Ripcke et al. 2009). Proteomic investigation of secondary metabolism (PrISM) has been developed to identify highly expressed megasynthase genes that do not require prior knowledge of genome sequence (Bumpus et al. 2009). The PKS and NRPS are high-molecular-weight enzyme proteins (> 200 kDa), and their proteome can be analyzed by excising their bands from protein gels, using high-resolution tandem mass spectrometry. The ions derived from the phosphopantetheinyl (Ppant) arm in the carrier protein domains of PKS or NRPS may then be tracked by mass spectrometry (Dorrestein et al. 2006). By using the de novo peptide sequencing of the proteome of wild-type Bacillus strain NK2018 using LC-MS, a novel NRPS cluster has been identified. The DNA sequence of this cluster has been observed to be 94% identical to B. cereus AH1134. In addition to the PrISM approach, fluorescent and biotinylated probes, targeting thioesterase domains of PKS and NRPS, have also been developed. Importantly, these probes do not depend on the genetic expression of metabolic clusters (Meier et al. 2008).

Combinatorial chemistry or metabolomics

The modular nature of secondary metabolites makes them an excellent source for their use in combinatorial chemistry. These metabolites are synthesized as polymer backbones and diversified by the actions of tailoring enzymes (Liu et al. 2011). The integration of the natural product synthesis with organic chemistry in combinatorial synthesis has led to engineer a completely new class of compounds, similar to the natural products (Ortholand and Ganesan 2004). In this process, the natural products are used as building blocks upon which new and diverse entities are added. Several combinatorial libraries have been developed covering a huge structural diversity of compounds. To explore natural products for their combinatorial synthesis, Romano group employed ultra-high resolution mass spectrometry to search the exo-metabolome of Pseudovibrio sp., grown under variable phosphate concentrations (Romano et al. 2014). Sarkar and colleagues applied metabolomics-based analysis to screen secondary metabolite production in A. nidulans, grown under various nutrient growth conditions and identified several polyphenolic compounds, including a complete novel prenylated benzophenone preshamixanthone (Sarkar et al. 2012). Forseth et al. identified cryptic products of the gliotoxin gene cluster using NMR-based comparative metabolomics (Forseth et al. 2011). They were able to screen gli-dependent metabolites using differential analysis of 2D-NMR spectra of metabolite extracts derived from A. fumigatus strain, deleted for gli gene. Several other strategies have also been developed for metabolomics-based prioritization of microbes for natural product discovery (Hou et al. 2012). Analytical technologies such as nano-spray desorption electrospray ionization (nanoDESI) mass spectrometry and matrix-assisted laser desorption ionization-time-of-flight-imaging mass spectrometry (MALDI-TOF-IMS) for high-throughput identification of cryptic secondary metabolites have also contributed significantly (Breitling et al. 2013).

Direct cloning using artificial promoters

The insertion of inducible strong promoters has been also reported to lead to the activation of the cryptic antibiotic gene clusters. The biosynthetic genes for secondary metabolites that are usually silent or are expressed minimally can be cloned using strong promoters into suitable plasmid vectors. The silent spectinabilin pathway of S. orinoci and taromycin A pathway of Saccharomonospora sp. have been identified using this system (Shao et al. 2013; Yamanaka et al. 2014). Similarly, a cryptic antibiotic gene cluster SGR810-815 in S. griseus has been also reported to induce three novel polycyclic tetramate macrolactams (Luo et al. 2013). In E. coli, alterochromide lipopeptides of Pseudoalteromonas piscicida have been shown to heterologously expressed using native, E. coli T7 promoter (Ross et al. 2015). The expression of a silent antibiotic gene cluster has also been achieved in Streptomyces using a strong promoter ermE (Baltz 2010). The bbr gene (transcriptional activator) of Amycolatopsis balhimycina also exhibited the induction of balhimycin biosynthesis in the heterologous host A. japonicum (Spohn et al. 2014).

Chromatin remodeling

Modifications that affect gene regulation in higher-order organisms include methylation, acetylation, phosphorylation, ubiquitination, ADP-ribosylation, and glycosylation (Cichewicz 2012). But with respect to the production of antibiotics and their induction from the corresponding genetic clusters, the aspect is relatively less studied. It has been reported that the putative gene clusters for antibiotic synthesis are present in fungal genomes in the distal regions of chromosomes (Shwab et al. 2007). These regions typically exist in a heterochromatin state and gene expression requires epigenetic control here; therefore, there exist a possibility that these antibiotic biosynthetic gene clusters may express. Keller et al. developed a method known as “chromatin remodeling” for the induction of secondary metabolite genes in microorganisms. While investigating the regulatory pathway of sterigmatocystin (a toxin in Aspergillus nidulans), they identified several mutant strains exhibiting the suppressed production of the toxin (Butchko et al. 1999). The mutation was identified in LaeA (methyltransferase) that is known to be involved in the regulation of sterigmatocystin production. In addition, deletion of LaeA blocked the expression of several biosynthetic gene clusters while overexpression of the same triggered penicillin and lovastatin production (Bok and Keller 2004). Since LaeA has the homology with histone methyltransferases, the regulation of this type was proposed as a chromatin-based model of regulation. The deletion of hdaA in Aspergillus histone deacetylase also resulted in the increased production of two telomere-proximal secondary metabolite gene clusters, but the transcription of a telomere-distal cluster remained unchanged (Shwab et al. 2007). Similarly, in Cichewicz’s experiments, 12 fungi were subjected to a library of DNA methyltransferase using variable concentrations of histone deacetylase inhibitors and 11 strains were observed for their increased production of diverse secondary metabolites. Oberlies et al. used the proteasome inhibitor bortezomib to induce a filamentous fungus to produce cryptic metabolite (VanderMolen et al. 2014). The similar method was applied by McArthur et al. in Streptomyces coelicolor (Moore et al. 2012). These observations clearly demonstrate the importance of small molecule epigenetic modifiers in accessing silent gene clusters for the discovery of novel metabolites.

Rare earth elements

Until recently, the rare earth elements were not known for their biological effects in living cells. These elements have been found not only to activate cryptic genes but also in the overproduction of antibiotics. The rare earth elements are consisted of 17 elements, including scandium, yttrium, and lanthanides. Interestingly, the growth medium containing 10–100 μM scandium or lanthanum has been reported to increase antibiotic production in Streptomyces sp. by 25-fold (Kawai et al. 2007). The effect of scandium in the induction of antibiotic in S. coelicolor has been demonstrated to be at the level of transcription of pathway-specific regulatory gene (act II-ORF4). The antibiotic production under low concentrations of scandium indicates that it functions in situ, as an important factor to induce the production of secondary metabolites as well as pigments, toxins, and antibiotics. The addition of scandium to the growth medium of B. subtilis has also been observed to enhance the production of α-amylase and bacilysin (Inaoka and Ochi 2011). The rare earth elements are distributed ubiquitously in the ecosystem at low levels; it is possible that microbes have acquired the ability to react with these low levels over the course of evolution to induce antibiotic biosynthetic gene clusters. The main advantage of using rare earth elements in culture media for augmenting antibiotic production is that the approach does not require prior knowledge of genetic engineering.

High-throughput elicitor screens

The high-throughput elicitor screen method was first used by Nodwell and co-workers to observe the induction of blue polyketide actinorhodin and red prodiginines in S. coelicolor (Craney et al. 2012). A total of approximately 30,500 small molecules were screened from the Canadian Compound Collection (McMaster University, HTS facility) to assess actinorhodin production in S. coelicolor. In this screening process, the upregulation of actinorhodin synthesis as well as the overproduction of germicidins remodeling compounds (ARC2 group; 4 compounds out of the selected 19 actinorhodin-stimulating molecules) was also recorded. The reduction in the production of daptomycin-like CDA and prodiginines was also observed. Intriguingly, ARC2 and triclosan (inducer of actinorhodin synthesis) both displayed the modulated secondary metabolism only at sub-inhibitory concentrations. ARC2 also altered the secondary metabolome of S. pristinaespiralis, S. Peucetius, and Kutzneria sp. by inducing cryptic metabolites. Recently, Cl-ARC (derivative of ARC2) has been also reported to induce a total of 216 cryptic metabolites in 50 different Streptomyces (Pimentel-Elardo et al. 2015). The usefulness of high-throughput elicitor screens has been also demonstrated in Burkholderia thailandensis (Liu and Cheng 2014; Seyedsayamdost 2014).

Co-cultivation

Presently, co-cultivation has turned into one of the main strategies to identify novel secondary metabolites from microorganisms (Brakhage 2013; Moody 2014). The co-cultivation approach or “interspecies crosstalk” has proven to be a success in the de novo production of secondary metabolites (Bertrand et al. 2013a, b; Brakhage and Schroeckh 2011; Marmann et al. 2014; Schroeckh et al. 2014). Several reports on fungal biotic interactions indicated that many cryptic secondary metabolites can be induced by co-culturing fungi with fungi (Bertrand et al. 2013a, b; Cueto et al. 2001; Mela et al. 2011; Wald et al. 2004). Many such studies have also been carried out involving confrontation of fungi and bacteria (Cheng et al. 2013; Ola et al. 2013). Several reports have established that co-culture strategy has a huge potential to activate silent gene clusters by yet unknown mechanisms. A study conducted by Konig et al. clearly demonstrated that co-culture of Aspergillus fumigatus with bacterium led to the activation of an otherwise silent pathway after addition of an epigenetic modulator (Konig et al. 2013). It has been demonstrated that co-cultivation of microbes can induce gene mutation and subsequent expression of silent gene clusters (Charusanti et al. 2012). The exchange of whole gene fragments (horizontal gene transfer) has also been reported to induce the gene expression of previously undetected secondary metabolites (Kurosawa et al. 2008). Pestalone, a potent antibiotic against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecium, is the product of a co-culture of Gram-negative bacterium Thalassopia with marine fungus Pestalotia (Cueto et al. 2001). Gram-positive Bacillus subtilisis has been also shown to induce macrocarpon, 2-carboxymethylamino benzoic acid, and citreoisocoumarinol in Fusarium tricinctum (Ola et al. 2013). Similarly, the activation of silent PKS gene clusters, during co-cultivation, resulted in the induction of a huge number of polyketides in Aspergillus nidulans (Schroeckh et al. 2014).

Environmental signals

The silent antibiotic gene clusters in microorganisms are considered to be a potential source of secondary metabolites, but the environmental clues to induce their expression remain unknown (Abrudan et al. 2015; Zhu et al. 2014). It is highly important to understand the biological role of cryptic antibiotic gene clusters in antibiotic-producing microbes in a given niche before making attempts to activate them. The members of actinomycetes group grow as a branched multicellular network of hyphae and are known to reproduce through spores that are formed by an aerial mycelium. The detailed description of the control of morphological differentiation in actinomycetes is reviewed elsewhere (Chater 2006; Flardh and Buttner 2009; Hopwood 2006). In natural systems, many antibiotics are produced after specific signals are receiving from the surrounding environment. Specialized techniques are required to decode these clues that can activate the production of secondary metabolites in microorganisms. The antibiotics have classically been considered as antimicrobial weapons (Raaijmakers and Mazzola 2012; Ratcliff and Denison 2011). The studies conducted by Abrudan et al. (Abrudan et al. 2015) and Westhoff et al. (Westhoff et al. 2017) in Streptomyces also supported the observation that antibiotics play their major role in environment as defense molecules. The generalization that antibiotics have an antagonistic role in nature similar to its clinical role is a big question (Linares et al. 2006; Ratcliff and Denison 2011; Romero et al. 2011). The reason for this assumption is the low concentrations of antibiotics present in the soil environment that may not exhibit inhibitory effects. Subsequently, sub-inhibitory concentrations of antibiotics can induce a pleiotropic response in microorganisms such as quorum sensing, biofilm formation, and coordinated expression of virulence genes (Hoffman et al. 2005; Stevens et al. 2007; Yim et al. 2006).

Small-molecule chemical elicitors

It is important to understand the regulatory control of cryptic biosynthetic gene clusters (Van Lanen and Shen 2006; Wilkinson and Micklefield 2007). The role of HTH-type transcriptional repressor DasR has been assessed in the control of cryptic type I PKS gene cluster. In S. coelicolor, a signaling cascade consisted of N-acetylglucosamine and the DasR regulon has been shown to activate the antibiotic production. A high concentration of N-acetylglucosamine (bacterial cell-wall component) triggers the antibiotic production under nutrient poor growth conditions (Rigali et al. 2008). N-acetylglucosamine binds to its effector binding site to DasR and acts as a ligand that reduces the protein’s affinity to DNA (Rigali et al. 2006). Another protein, AtrA, appears to counteract DasR, by having opposed effects in the signaling pathway. DasR represses the gene transcription of NagE2 (GlcNAc-specific transporter) and ActII-ORF4, whereas AtrA activates both of these genes at the transcription level (Nothaft et al. 2010). DasR also controls siderophore production (Craig et al. 2012). The enhanced transcription of chromosomally encoded antibiotic biosynthetic clusters (act, cda, red, and cryptic cpk) has been observed in dasR mutants of S. coelicolor (Rigali et al. 2008). Thus, the expression of DasR is linked to the induction of antibiotic genes in many actinomycetes, provided N-acetylglucosamine is used as a carbon source (van Wezel et al. 2009). Other than N-acetylglucosamine, molecules such as nucleotides, oligopeptides, amino acids, polysaccharides, and fatty acids etc. have also been reported to induce the antibiotic production. Triclosan which is known to mediate cell-cell communication has also been observed to involve in quorum sensing and autoinduction mechanisms. When a culture of Pseudomonas aeruginosa is treated with triclosan, it exhibited downregulation of the quorum-sensing genes (Chuanchuen and Schweizer 2012; Craney et al. 2012). The quorum-sensing signaling for activating antibiotic production in streptomycetes is regulated by γ-butyrolactones (Takano 2006).

Genome mining

The abundance of readily available genomic data and advances in sequencing and computational techniques has helped to achieve higher success rate in predicting the novel secondary metabolites from cryptic gene clusters. The following part of the review describes the overall development in the field. At the present time, whole culture collections of microbes have been sequenced and new technologies like single cell genomics and metagenomics have generated massive data to analyze. Previously, conserved genes for PKS or NRPS domains were used as probes in Southern hybridization experiments. DNA primers were prepared from these highly conserved motifs for PCR screenings. Thus, generated results were analyzed using sequence comparison software such as BLAST (Altschul et al. 1990) or DIAMOND (Buchfink et al. 2015) or profile-based tools like HMMer (Finn et al. 2011). Presently, in silico genetic screening of novel NRPS and PKS is performed using NRPS/PKS domain organization in bacteria and from the available metagenomic data. DECEIPHER is the first tool developed by Ecopia Biosciences Inc. for automated cluster mining (Farnet and Zazopoulos 2005). Thereafter, many other tools were developed that include BAGEL (De Jong et al. 2006), CLUSEAN (Weber et al. 2009), antiSMASH3.0 (Blin et al. 2013; Medema et al. 2015; Weber 2015), NRPSpredictor2 ((Röttig et al. 2011)), SBS-PKS (Anand et al. 2010; Ansari et al. 2004), PKS/NRPS (Bachmann and Ravel 2009), NORINE (Caboche et al. 2008), ClustScan (Starcevic et al. 2008), NaPDoS (Ziemert et al. 2012), NRPSsp (Prieto et al. 2012), NRPS/PKS substrate predictor (Khayatt et al. 2013), PRISM (Skinnider et al. 2015), Dynamite (Ogasawara et al. 2015) etc. Using this wealth of information and applied bioinformatics tool, many attempts have been made to identify antibiotics with novel structures and are being submitted to the natural product library at regular basis (Chen et al. 2017; Milshteyn et al. 2014; Ugai et al. 2016; Xu et al. 2016).

Concluding remarks

The discovery and development of antibiotics is one of the greatest achievements in the treatment and prevention of bacterial infections. New infectious diseases and resistant pathogens have been discovered at a rapid rate, but there has been no discovery of new antibiotics that can keep pace with it. It is speculated that the microbial parvome for the discovery of novel metabolic molecules has been exhausted. The increasing rates of antimicrobial resistance urgently necessitate new strategies to stock-up antimicrobial drug pipelines. It is widely accepted that microbes possess a huge potential for the biosynthesis of secondary metabolites of high chemical diversity. It is highly challenging to explore this diversity within the microbial biosynthetic repertoire to drive expression, development, and synthesis of new chemical scaffolds. In addition, several of these biosynthetic pathways are observed to be cryptic or are expressed minimally; therefore, novel strategies are required to identify the environmental clues to activate them. Awakening cryptic gene clusters to produce polymorphism of secondary metabolites is a difficult process, but using the above-described strategies (Fig. 3), the activation of the cryptic genes may lead to the identification of novel compounds of therapeutic and industrial use. Apart from the technicalities of using different strategies for decryptification, it is also important to analyze why biosynthetic genes are cryptic under standard laboratory conditions. Understanding the regulation of these cryptic systems would make possible to explore the full potential of biosynthetic gene clusters in microorganisms for the screening of new secondary metabolic compounds. Nevertheless, additional efforts have to be made in developing more effective strategies to study and understand cryptic genetic systems for the discovery of their yet-unidentified natural products. We do not really need to completely rely on nature as a sole provider of novel natural products; chemical synthesis and modifications of existing moieties also provide potential alternatives. However, it is important to note that nature has unearthed only a tiny fraction of its huge store of metabolic treasure of natural products and it will remain the most preferred source over the chemical synthesis. Upon the discovery and exploration of yet-to-be-discovered secondary metabolites from the hidden sources, we will definitively be having solutions near in the future against the current crises of “slow-moving-drug-development” and multi-drug-resistant pathogens. The consortia of these innovative strategies have strengthened the belief and enlighten the path for the discovery and development of novel chemical scaffolds. With the increasing application of such strategies, it is hoped that novel and effective antibiotics will be identified soon from the microbial parvome. The identification of secondary metabolites from these cryptic genes may facilitate the discovery of much needed novel antimicrobials to effectively resist multiple-drug-resistant pathogens. The burden is on academicians now to conduct research on the hidden aspects of the microbial treasure of secondary metabolism for the identification of yet-to be-identified antibiotics.

Fig. 3
figure 3

Strategies for the induction of secondary metabolites from cryptic gene clusters in bacteria

References

  • Abrudan M, Smakman F, Grimbergen AJ, Westhoff S, Miller EL, van Wezel GP, Rozen DE (2015) Socially mediated induction and suppression of antibiosis during bacterial coexistence. Proc Natl Acad Sci U S A 112:11054–11059

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Allsopp D, Colwell RR, Hawksworth DL (1995) . CAB International, Wallingford

  • Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990) Basic local alignment search tool. J Mol Biol 215:403–410

    Article  PubMed  CAS  Google Scholar 

  • Anand S, Prasad MV, Yadav G, Kumar N, Shehara J, Ansari MZ, Mohanty D (2010) SBSPKS: structure based sequence analysis of polyketide synthases. Nucleic Acids Res 38:W487–W496

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ansari MZ, Yadav G, Gokhale RS, Mohanty D (2004) NRPSPKS: a knowledge-based resource for analysis of NRPS/PKS megasynthases. Nucleic Acids Res 32:W405–W413

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Aoi Y, Kinoshita T, Hata T, Ohta H, Obokata H, Tsuneda S (2009) Hollow fiber membrane chamber as a device for in situ environmental cultivation. Appl Environ Microbiol 75:3826–3833

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Bachmann BO, Ravel J (2009) Methods for in silico prediction of microbial polyketide and nonribosomal peptide biosynthetic pathways from DNA sequence data. Methods in enzymology. Elsevier

  • Bachmann BO, Van Lanen SG, Baltz RH (2014) Microbial genome mining for accelerated natural products discovery: is a renaissance in the making? J Ind Microbiol Biotechnol 41:175–184

    Article  PubMed  CAS  Google Scholar 

  • Baltz RH (2010) Streptomyces and Saccharopolyspora hosts for heterologous expression of secondary metabolite gene clusters. J Ind Microbiol Biotechnol 37:759–772

    Article  PubMed  CAS  Google Scholar 

  • Bentley R (1997) Microbial secondary metabolites play important roles in medicine; prospects for discovery of new drugs. Perspect Biol Med 40:364–394

    Article  PubMed  CAS  Google Scholar 

  • Berdy J (2005) Bioactive microbial metabolites. J Antibiot 58:1–26

    Article  CAS  Google Scholar 

  • Berdy J (2012) Thoughts and facts about antibiotics: where we are now and where we are heading. J Antibiot 65:385–395

    Article  CAS  Google Scholar 

  • Bertrand S, Schumpp O, Bohni N, Bujard A, Azzollini A, Monod M (2013a) Detection of metabolite induction in fungal co-cultures on solid media by high-throughput differential ultra-high pressure liquid chromatography–time-of-flight mass spectrometry fingerprinting. J Chromatogr A1292:219–228

    Article  CAS  Google Scholar 

  • Bertrand S, Schumpp O, Bohni N, Monod M, Gindro K, Wolfender JL (2013b) De novo production of metabolites by fungal co-culture of Trichophyton rubrum and Bionectria ochroleuca. J Nat Prod 76:1157–1165

    Article  PubMed  CAS  Google Scholar 

  • Bibb MJ (2005) Regulation of secondary metabolism in Streptomycetes. Curr Opin Microbiol 8:208–215

    Article  PubMed  CAS  Google Scholar 

  • Blin K, Medema MH, Kazempour D, Fischbach MA, Breitling R, Takano E, Weber T (2013) antiSMASH 2.0 - a versatile platform for genome mining of secondary metabolite producers. Nucleic Acids Res 41:204–212

    Article  Google Scholar 

  • Bode HB, Bethe B, Hofs R, Zeeck A (2002) Big effects from small changes: possible ways to explore nature’s chemical diversity. Chembiochem 3:619–627

    Article  PubMed  CAS  Google Scholar 

  • Bok JW, Keller NP (2004) LaeA, a regulator of secondary metabolism in Aspergillus spp. Eukaryot Cell 3:527–535

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Bok JW, Soukup AA, Chadwick E, Chiang YM, Wang CC, Keller NP (2013) VeA and MvlA repression of the cryptic orsellinic acid gene cluster in Aspergillus nidulans involves histone 3 acetylation. Mol Microbiol 89:963–974

    Article  PubMed  CAS  Google Scholar 

  • Brakhage AA (2013) Regulation of fungal secondary metabolism. Nat Rev Microbiol 11:21–32

    Article  PubMed  CAS  Google Scholar 

  • Brakhage AA, Schroeckh V (2011) Fungal secondary metabolites—strategies to activate silent gene clusters. Fungal Genet Biol 48:15–22

    Article  PubMed  CAS  Google Scholar 

  • Breitling R, Takano E (2015) Synthetic biology advances for pharmaceutical production. Curr Opin Biotechnol 35:46–51

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Breitling R, Ceniceros A, Jankevics A, Takano E (2013) Metabolomics for secondary metabolite research. Metabolites 3:1076–1083

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Buchfink B, Xie C, Huson DH (2015) Fast and sensitive protein alignment using DIAMOND. Nat Methods 12:59–60

    Article  PubMed  CAS  Google Scholar 

  • Bumpus SB, Evans BS, Thomas PM, Ntai I, Kelleher NL (2009) A proteomics approach to discovering natural products and their biosynthetic pathways. Nat Biotechnol 27:951–956

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Butchko RA, Adams TH, Keller NP (1999) Aspergillus nidulans mutants defective in stc gene cluster regulation. Genetics 153:715–720

    PubMed  PubMed Central  CAS  Google Scholar 

  • Caboche S, Pupin M, Leclere V, Fontaine A, Jacques P, Kucherov G (2008) Norine: a database of nonribosomal peptides. Nucleic Acids Res 36:D326–D331

    Article  PubMed  CAS  Google Scholar 

  • Chai YJ, Cui CB, Li CW, Wu CJ, Tian CK, Hua W (2012) Activation of the dormant secondary metabolite production by introducing gentamicin-resistance in a marine-derived Penicillium purpurogenum G59. Mar Drugs 10:559–582

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Charusanti P, Fong NL, Nagarajan H, Pereira AR, Li HJ, Abate EA, Su Y, Gerwick WH, Palsson BO (2012) Exploiting adaptive laboratory evolution of Streptomyces clavuligerus for antibiotic discovery and overproduction. PLoS One 7:e33727

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Chater KF (2006) Streptomyces inside out: a new perspective on the bacteria that provide us with antibiotics. Philos Trans R Soc B 361:761–768

    Article  CAS  Google Scholar 

  • Chen R, Zhang Q, Tan B, Zheng L, Li H, Zhu Y, Zhang C (2017) Genome mining and activation of a silent PKS/NRPS gene cluster direct the production of Totopotensamides. Org Lett 19(20):5697–5700

    Article  PubMed  CAS  Google Scholar 

  • Cheng YF, Jin W, Mao SY, Zhu WY (2013) Production of citrate by anaerobic fungi in the presence of co-culture methanogens as revealed by 1H NMR spectrometry. Asian Australa J Anim Sci 26:1416–1423

    Article  CAS  Google Scholar 

  • Chuanchuen R, Schweizer HP (2012) Global transcriptional responses to triclosan exposure in Pseudomonas aeruginosa. Int J Antimicrob Agents 40:114–122

    Article  PubMed  CAS  Google Scholar 

  • Cichewicz RH (2012) Epigenetic regulation of secondary metabolite biosynthetic genes in fungi. In: Witzany G (ed) Biocommunication of Fungi. Springer: Germany, pp 57–69

  • Colombo V, Fernandez-de-Heredia M, Malpartida F (2001) A polyketide biosynthetic gene cluster from Streptomyces antibioticus includes a LysR-type transcriptional regulator. Microbiology 147:3083–3092

    Article  PubMed  CAS  Google Scholar 

  • Connon SA, Giovannoni SJ (2002) High-throughput methods for culturing microorganisms in very-low-nutrient media yield diverse new marine isolates. Appl Environ Microbiol 68:3878–3885

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Craig M, Lambert S, Jourdan S, Tenconi E, Colson S, Maciejewska M, Ongena M, Martin JF, van Wezel G, Rigali S (2012) Unsuspected control of siderophore production by N-acetylglucosamine in streptomycetes. Environ Microbiol Rep 4:512–521

    Article  PubMed  CAS  Google Scholar 

  • Craney A, Ozimok C, Pimentel-Elardo SM, Capretta A, Nodwell JR (2012) Chemical perturbation of secondary metabolism demonstrates important links to primary metabolism. Chem Biol 19:1020–1027

    Article  PubMed  CAS  Google Scholar 

  • Cueto M, Jensen PR, Kauffman C, Fenical W, Lobkovsky E, Clardy J (2001) Pestalone, a new antibiotic produced by a marine fungus in response to bacterial challenge. J Nat Prod 64:1444–1446

    Article  PubMed  CAS  Google Scholar 

  • Daletos G, Ebrahim W, Ancheeva E, El-Neketi M, Lin WH, Proksch P (2017) Microbial coculture and OSMAC approach as strategies to induce cryptic fungal biogenetic gene clusters. In: Cragg G, Newman DJ (eds) Chemical Biology of Natural Products. CRC Press, pp 233–284

  • Davies J (2011) How to discover new antibiotics: harvesting the Parvome. Curr Opin Chem Biol 15:5–10

    Article  PubMed  CAS  Google Scholar 

  • De Jong A, Hijum V, Bijlsma JE, Kok J, Kuipers PO (2006) Bagel: a web-based bacteriocin genome mining tool. Nucleic Acid Res 34:273–279

    Article  CAS  Google Scholar 

  • Dietrich JA, Yoshikuni Y, Fisher KJ, Woolard FX, Ockey D, McPhee DJ, Renninger NS, Chang MCY, Baker D, Keasling JD (2009) A novel semi-biosynthetic route for artemisinin production using engineered substrate-promiscuous P450(BM3). ACS Chem Biol 4:261–267

    Article  PubMed  CAS  Google Scholar 

  • Dorrestein PC, Bumpus SB, Calderone CT, Garneau-Tsodikova S, Aron ZD, Straight PD, Kolter R, Walsh CT, Kelleher NL (2006) Facile detection of acyl and peptidyl intermediates on thiotemplate carrier domains via phosphopantetheinyl elimination reactions during tandem mass spectrometry. Biochemistry 45:12756–12766

    Article  PubMed  CAS  Google Scholar 

  • Eilers H, Pernthaler J, Glockner FO, Amann R (2000) Culturability and in situ abundance of pelagic bacteria from the North Sea. Appl Environ Microbiol 66:3044–3051

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Farnet CM, Zazopoulos E (2005) Improving drug discovery from microorganisms. In: Zhang L, Demain AL (eds) Natural products: drug discovery and therapeutic medicine. Humana Press, Totowa, pp 95–106

    Chapter  Google Scholar 

  • Feher M, Schmidt JM (2003) Property distributions: differences between drugs, natural products, and molecules from combinatorial chemistry. J Chem Inf Comput Sci 43:218–227

    Article  PubMed  CAS  Google Scholar 

  • Ferrari BC, Gillings MR (2009) Cultivation of fastidious bacteria by viability staining and micromanipulation in a soil substrate membrane system. Appl Environ Microbiol 75:3352–3354

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ferrari BC, Binnerup SJ, Gillings M (2005) Microcolony cultivation on a soil substrate membrane system selects for previously uncultured soil bacteria. Appl Environ Microbiol 71:8714–8720

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Fierer N, Jackson RB (2006) The diversity and biogeography of soil bacterial communities. Proc Natl Acad Sci U S A 103:626–631

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Finn RD, Clements J, Eddy SR (2011) HMMER web server: interactive sequence similarity searching. Nucleic Acids Res 39:W29–W37

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Flardh K, Buttner MJ (2009) Streptomyces morphogenetics: dissecting differentiation in a filamentous bacterium. Nat Rev Microbiol 7:36–49

    Article  PubMed  CAS  Google Scholar 

  • Floriano B, Bibb M (1996) afsR is a pleiotropic but conditionally required regulatory gene for antibiotic production in Streptomyces coelicolor A3(2). Mol Microbiol 21:385–396

    Article  PubMed  CAS  Google Scholar 

  • Forseth RR, Fox EM, Chung D, Howlett BJ, Keller NP, Schroeder FC (2011) Identification of cryptic products of the gliotoxin gene cluster using NMR-based comparative metabolomics and a model for gliotoxin biosynthesis. J Am Chem Soc 133:9678–9681

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Gao C, Hindra Mulder D, Yin C, Elliot MA (2012) Crp is a global regulator of antibiotic production in Streptomyces. MBio 3:e00407–e00412

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Gottelt M, Kol S, Gomez-Escribano JP, Bibb M, Takano E (2010) Deletion of a regulatory gene within the cpk gene cluster reveals novel antibacterial activity in Streptomyces coelicolor A3(2). Microbiology 156:2343–2353

    Article  PubMed  CAS  Google Scholar 

  • Gross H, Stockwell VO, Henkels MD, Nowak-Thompson B, Loper JE, Gerwick WH (2007) The genomisotopic approach: a systematic method to isolate products of orphan biosynthetic gene clusters. Chem Biol 14:53–63

    Article  PubMed  CAS  Google Scholar 

  • Harwani D (2013) The great plate count anomaly and the unculturable bacteria. Int J Sci Res 2:350–351

    Google Scholar 

  • Hawksworth DL (1991) The fungal dimension of biodiversity: magnitude, significance, and conservation. Mycol Res 95:641–655

    Article  Google Scholar 

  • Helfrich EJN, Reiter S, Piel J (2014) Recent advances in genome-based polyketide discovery. Curr Opin Biotechnol 29:107–115

    Article  PubMed  CAS  Google Scholar 

  • Hoffman LR, D'Argenio DA, MacCoss MJ, Zhang Z, Jones RA, Miller SI (2005) Aminoglycoside antibiotics induce bacterial biofilm formation. Nature 436(7054):1171–1175

    Article  PubMed  CAS  Google Scholar 

  • Hopwood DA (2006) Soil to genomics: the Streptomyces chromosome. Annu Rev Genet 40:1–23

    Article  PubMed  CAS  Google Scholar 

  • Hoshaka T, Ohnishi-Kameyama M, Muramatsu H, Murakami K, Tsurumi Y, Kodani S, Yoshida M, Fujie A, Ochi K (2009) Antibacterial discovery in actinomycetes strains with mutations in RNA polymerase or ribosomal protein S12. Nat Biotechnol 27:462–464

    Article  CAS  Google Scholar 

  • Hou Y, Braun DR, Michel CR, Klassen JL, Adnani N, Wyche TP, Bugni TS (2012) Microbial strain prioritization using metabolomics tools for the discovery of natural products. Anal Chem 84:4277–4283

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ikeda H, Nonomiya T, Usami M, Ohta T, Omura S (1999) Organization of the biosynthetic gene cluster for the polyketide anthelmintic macrolide avermectin in Streptomyces avermitilis. Proc Natl Acad Sci U S A 96:9509–9514

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Imai Y, Fujiwara T, Ochi K, Hosaka T (2012) Development of the ability to produce secondary metabolites in Streptomyces through the acquisition of erythromycin resistance. J Antibiot (Tokyo) 65:323–326

    Article  CAS  Google Scholar 

  • Imai Y, Sato S, Tanaka Y, Ochi K, Hosaka T (2015) Lincomycin at subinhibitory concentrations potentiates secondary metabolite production by Streptomyces spp. Appl Environ Microbiol 81:3869–3879

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Inaoka T, Ochi K (2011) Scandium stimulates the production of amylase and bacilysin in Bacillus subtilis. Appl Environ Microbiol 77:8181–8183

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Kaeberlein T, Lewis K, Epstein SS (2002) Isolating “uncultivable” microorganisms in pure culture in a simulated natural environment. Science 296:1127–1129

    Article  PubMed  CAS  Google Scholar 

  • Kallel H, Bahloul M, Hergafietal L, Akrout M, Ketata W, Chelly H, Hamida CB, Rekik N, Hammami A, Bouaziz M (2006) Colistin as a salvage therapy for nosocomial infections caused by multidrug-resistant bacteria in the ICU. Int J Antimicrob Agents 28:366–369

    Article  PubMed  CAS  Google Scholar 

  • Kawai K, Wang G, Okamoto S, Ochi K (2007) The rare earth, scandium, causes antibiotic overproduction in Streptomyces spp. FEMS Microbiol Lett 274:311–315

    Article  PubMed  CAS  Google Scholar 

  • Khayatt BI, Overmars L, Siezen RJ, Francke C (2013) Classification of the adenylation and acyl-transferase activity of NRPS and PKS systems using ensembles of substrate specific hidden Markov models. PLoS One 8:e62136

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Klein J, Heal JR, Hamilton WDO, Boussemghoune T, Tange TO, Delegrange F, Jaeschke G, Hatsch A, Heim J (2014) Yeast synthetic biology platform generates novel chemical structures as scaffolds for drug discovery. ACS Synth Biol 3:314–323

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Klevens RM, Edwards JR, Richards CL Jr, Horan TC, Gaynes RP, Pollock DA, Cardo DM (2007) Estimating health care-associated infections and deaths in U.S hospitals. Public Health Rep 122:160–166

    Article  PubMed  PubMed Central  Google Scholar 

  • Konig CC, Scherlach K, Schroeckh V, Horn F, Nietzsche S (2013) Bacterium induces cryptic meroterpenoid pathway in the pathogenic fungus Aspergillus fumigatus. Chembiochem 14:938–942

    Article  PubMed  CAS  Google Scholar 

  • Kurosawa K, Ghiviriga I, Sambandan TG, Lessard PA, Barbara JE, Rha C (2008) Rhodostreptomycins, antibiotics biosynthesized following horizontal gene transfer from Streptomyces padanus to Rhodococcus fascians. J Am Chem Soc 130:1126–1127

    Article  PubMed  CAS  Google Scholar 

  • Laureti L, Song L, Huang S, Corre C, Leblond P, Challis GL, Aigle B (2011) Identification of a bioactive 51-membered macrolide complex by activation of a silent polyketide synthase in Streptomyces ambofaciens. Proc Natl Acad Sci U S A 108:6258–6263

    Article  PubMed  PubMed Central  Google Scholar 

  • Lee JY, Hwang YS, Kim SS, Kim ES, Choi CY (2000) Effect of a global regulatory gene, afsR2, from Streptomyces lividans on avermectin production in Streptomyces avermitilis. J Biosci Bioeng 89:606–608

    Article  PubMed  CAS  Google Scholar 

  • Linares JF, Gustafsson I, Baquero F, Martinez JL (2006) Antibiotics as intermicrobial signaling agents instead of weapons. Proc Natl Acad Sci U S A 103:19484–19489

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP, Mueller A, Schäberle TF, Hughes DE, Epstein S, Jones M, Lazarides L, Steadman VA, Cohen DR, Felix CR, Fetterman KA, Millett WP, Nitti AG, Zullo AM, Chen C, Lewis K (2015) A new antibiotic kills pathogens without detectable resistance. Nature 517:455–459

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  • Liu X, Cheng YQ (2014) Genome-guided discovery of diverse natural products from Burkholderia sp. J Ind Microbiol Biotechnol 41:275–284

    Article  PubMed  CAS  Google Scholar 

  • Liu T, Chiang YM, Somoza AD, Oakley BR, Wang CC (2011) Engineering of an “unnatural” natural product by swapping polyketide synthase domains in Aspergillus nidulans. J Am Chem Soc 133:13314–13316

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Luo Y, Huang H, Liang J, Wang M, Lu L, Shao Z, Cobb RE, Zhao H (2013) Activation and characterization of a cryptic polycyclic tetramate macrolactam biosynthetic gene cluster. Nat Commun 4:2894

    Article  PubMed  CAS  Google Scholar 

  • Maddocks SE, Oyston PCF (2008) Structure and function of the LysR-type transcriptional regulator (LTTR) family proteins. Microbiology 154:3609–3623

    Article  PubMed  CAS  Google Scholar 

  • Maharjan S, Oh TJ, Lee HC, Sohng JK (2009) Identification and functional characterization of an afsR homolog regulatory gene from Streptomyces venezuelae ATCC 15439. J Microbiol Biotechnol 19:121–127

    Article  PubMed  CAS  Google Scholar 

  • Marmann AH, Aly W, Lin B, Wang P, Proksch P (2014) Co-cultivation-a powerful emerging tool for enhancing the chemical diversity of microorganism. Mar Drugs 12:1043–1065

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Martin JF (2004) Phosphate control of the biosynthesis of antibiotics and other secondary metabolites is mediated by the PhoR-PhoP system: an unfinished story. J Bacteriol 186:5197–5201

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Martin VJJ, Pitera DJ, Withers ST, Newman JD, Keasling JD (2003) Engineering a mevalonate pathway in Escherichia coli for production of terpenoids. Nat Biotechnol 21:796–802

    Article  PubMed  CAS  Google Scholar 

  • Matsumoto A, Ishizuka H, Beppu T, Horinouchi S (1995) Involvement of a small ORF downstream of the afsR gene in the regulation of secondary metabolism in Streptomyces coelicolor A3(2). Actinomycetologica 9:37–43

    Article  Google Scholar 

  • Medema MH, Breitling R, Takano E (2011a) Synthetic biology in Streptomyces bacteria. Methods Enzymol 497:485–502

    Article  PubMed  CAS  Google Scholar 

  • Medema MH, Breitling R, Bovenberg R, Takano E (2011b) Exploiting plug-and-play synthetic biology for drug discovery and production in microorganisms. Nat Rev Microbiol 9:131–137

    Article  PubMed  CAS  Google Scholar 

  • Medema MH, Kottmann R, Yilmaz P, Cummings M, Biggins JB, Blin K, de Bruijn I, Chooi YH, Claesen J, Coates RC (2015) Minimum information about a biosynthetic gene cluster. Nat Chem Biol 11:625–631

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Meier JL, Mercer AC, Burkart MD (2008) Fluorescent profiling of modular biosynthetic enzymes by complementary metabolic and activity based probes. J Am Chem Soc 130:5443–5445

    Article  PubMed  CAS  Google Scholar 

  • Mela F, Fritsche K, de Boer W, van Veen JA, de Graaff LH (2011) Dual transcriptional profiling of a bacterial/fungal confrontation: Collimonas fungivorans versus Aspergillus niger. ISME J 5:1494–1504

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Miaomiao L, Tanja G, Xueting L, Jianying H, Lixin Z, Ronald JQ (2017) A systems approach using OSMAC, log P and NMR fingerprinting: an approach to novelty. Synth Syst Biotechnol 2:276–286

    Article  Google Scholar 

  • Milshteyn A, Schneider JS, Brady SF (2014) Mining the metabiome: identifying novel natural products from microbial communities. Chem Biol 21:1211–1223

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Moody SC (2014) Microbial co-culture: harnessing intermicrobial signaling for the production of novel antimicrobials. Future Microbiol 9:575–578

    Article  PubMed  CAS  Google Scholar 

  • Moore JM, Bradshaw E, Seipke RF, Hutchings MI, McArthur M (2012) Use and discovery of chemical elicitors that stimulate biosynthetic gene clusters in Streptomyces bacteria. Methods Enzymol 517:367–385

    Article  PubMed  CAS  Google Scholar 

  • Newman JD, Marshall J, Chang M, Nowroozi F, Paradise E, Pitera D, Newman KL, Keasling JD (2006) High-level production of amorpha-4,11-diene in a two-phase partitioning bioreactor of metabolically engineered Escherichia coli. Biotechnol Bioeng 95:684–691

    Article  PubMed  CAS  Google Scholar 

  • Nichols D, Cahoon N, Trakhtenberg EM, Pham L, Mehta A, Belanger A, Kanigan T, Lewis K, Epstein SS (2010) Use of ichip for high-throughput in situ cultivation of “uncultivable” microbial species. Appl Environ Microbiol 76:2445–2450

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Nothaft H, Rigali S, Boomsma B, Swiatek M, McDowall KJ, van Wezel GP, Titgemeyer F (2010) The permease gene nagE2 is the key to N-acetylglucosamine sensing and utilization in Streptomyces coelicolor and is subject to multi-level control. Mol Microbiol 75:1133–1144

    Article  PubMed  CAS  Google Scholar 

  • Novakova R, Rehakova A, Kutas P, Feckova L, Kormanec J (2011) The role of two SARP family transcriptional regulators in regulation of the auricin gene cluster in Streptomyces aureofaciens CCM 3239. Microbiology 157:1629–1639

    Article  PubMed  CAS  Google Scholar 

  • Ochi K, Okamoto S, Tozawa Y, Inaoka T, Hosaka T, Xu J, Kurosawa K (2004) Ribosome engineering and secondary metabolite production. Adv Appl Microbiol 56:155–184

    Article  PubMed  CAS  Google Scholar 

  • Ogasawara Y, Benjamin J, Yackley Jacob A, Greenberg, Rogelj S, Charles E, Melançon I (2015) Expanding our understanding of sequence-function relationships of type II polyketide biosynthetic gene clusters: bioinformatics-guided identification of Frankiamicin a from Frankia sp. EAN1pec. PLoS One 10:1–25

    Google Scholar 

  • Ola ARB, Thomy D, Lai D, Brotz-Oesterhelt H, Proksch P (2013) Inducing secondary metabolite production by the endophytic fungus Fusarium tricinctum through coculture with Bacillus subtilis. J Nat Prod 76:2094–2099

    Article  PubMed  CAS  Google Scholar 

  • Ortholand JY, Ganesan A (2004) Natural products and combinatorial chemistry: back to the future. Curr Opin Chem Biol 8:271–280

    Article  PubMed  CAS  Google Scholar 

  • Parajuli N, Viet HT, Ishida K, Tong HT, Lee HC, Liou K, Sohng JK (2005) Identification and characterization of the afsR homologue regulatory gene from Streptomyces peucetius ATCC 27952. Res Microbiol 156:707–712

    Article  PubMed  CAS  Google Scholar 

  • Paulsen IT, Press CM, Ravel J, Kobayashi DY, Myers GS, Mavrodi D, DeBoy RT, Seshadri R, Ren Q, Madupu R, Dodson RJ, Durkin S, Brinkac LM, Daugherty SC, Sullivan SA, Rosovitz M, Gwinn ML, Zhou L, Nelson WC, Weidman J, Watkins K, Tran K, Khouri HM, Pierson E, Pierson L, Thomashow L, Loper J (2005) Complete genome sequence of the plant commensal Pseudomonas fluorescens pf-5. Nat Biotechnol 23:873–878

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  • Peterson LR (2009) Bad bugs, no drugs: no ESCAPE revisited. Clin Infect Dis 49:992–993

    Article  PubMed  Google Scholar 

  • Piddock LJV (2015) Teixobactin, the first of a new class of antibiotics discovered by iChip technology. J Antimicrob Chemother 70:2679–2680

    Article  PubMed  CAS  Google Scholar 

  • Pimentel-Elardo SM, Sorensen D, Ho L, Ziko M, Bueler SA, Lu S (2015) Activity-independent discovery of secondary metabolites using chemical elicitation and cheminformatic inference. ACS Chem Biol 10:2616–2623

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Prieto C, Garcia Estrada C, Lorenzana D, Martin JF (2012) NRPS sp: non-ribosomal peptide synthase substrate predictor. Bioinformatics 28:426–427

    Article  PubMed  CAS  Google Scholar 

  • Raaijmakers JM, Mazzola M (2012) Diversity and natural functions of antibiotics produced by beneficial and plant pathogenic bacteria. Annu Rev Phytopathol 50:403–424

    Article  PubMed  CAS  Google Scholar 

  • Rappe MS, Giovannoni SJ (2003) The uncultured microbial majority. Annu Rev Microbiol 57:369–394

    Article  PubMed  CAS  Google Scholar 

  • Ratcliff WC, Denison RF (2011) Alternative actions for antibiotics. Science 332:547–548

    Article  PubMed  CAS  Google Scholar 

  • Rausch C, Weber T, Kohlbacher O, Wohlleben W, Huson DH (2005) Specificity prediction of adenylation domains in nonribosomal peptide synthetases (NRPS) using transductive support vector machines (TSVMs). Nucleic Acids Res 33:5799–5808

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Rice LB (2006) Challenges in identifying new antimicrobial agents effective for treating infections with Acinetobacter baumannii and Pseudomonas aeruginosa. Clin Infect Dis 43:S100–S105

    Article  PubMed  CAS  Google Scholar 

  • Rice LB (2008) Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J Infect Dis 197:1079–1081

    Article  PubMed  Google Scholar 

  • Rigali S, Nothaft H, Noens EE, Schlicht M, Colson S, Muller M, Joris B, Koerten HK, Hopwood DA (2006) The sugar phosphotransferase system of Streptomyces coelicolor is regulated by the GntR-family regulator DasR and links N-acetylglucosamine metabolism to the control of development. Mol Microbiol 61:1237–1251

    Article  PubMed  CAS  Google Scholar 

  • Rigali S, Titgemeyer F, Barends S, Mulder S, Thomae AW, Hopwood DA, van Wezel GP (2008) Feast or famine: the global regulator DasR links nutrient stress to antibiotic production by Streptomyces. EMBO Rep 9:670–675

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ro DK, Paradise EM, Ouellet M, Fisher KJ, Newman KL, Ndungu JM, Ho KA, Eachus RA, Ham TS, Kirby J (2006) Production of the antimalarial drug precursor artemisinic acid in engineered yeast. Nature 440:940–943

    Article  PubMed  CAS  Google Scholar 

  • Rodriguez M, Nunez LE, Brana AF, Mendez C, Salas JA, Blanco G (2008) Identification of transcriptional activators for thienamycin and cephamycin c biosynthetic genes within the thienamycin gene cluster from Streptomyces cattleya. Mol Microbiol 69:633–645

    Article  PubMed  CAS  Google Scholar 

  • Romano S, Dittmar T, Bondarev V, Weber RJ, Viant MR, Schulz-Vogt HN (2014) Exo-metabolome of Pseudovibrio sp. FO-BEG1 analyzed by ultra-high resolution mass spectrometry and the effect of phosphate limitation. PLoS One 9:e96038

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Romero D, Traxler MF, Lopez D, Kolter R (2011) Antibiotics as signal molecules. Chem Rev 111:5492–5505

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Ross AC, Gulland LE, Dorrestein PC, Moore BS (2015) Targeted capture and heterologous expression of the Pseudoalteromonas alterochromide gene cluster in Escherichia coli represents a promising natural product exploratory platform. ACS Synth Biol 4:414–420

    Article  PubMed  CAS  Google Scholar 

  • Roszak DB, Colwell RR (1987) Survival strategies of bacteria in the natural environment. Microbiol Rev 51:365–379

    PubMed  PubMed Central  CAS  Google Scholar 

  • Röttig M, Medema MH, Blin K, Weber T, Rausch C, Kohlbacher O (2011) NRPSpredictor2—a web server for predicting NRPS adenylation domain specificity. Nucleic Acids Res 39:362–336

    Article  CAS  Google Scholar 

  • Sakai K, Kinoshita H, Shimizu T, Nihira T (2008) Construction of a citrinin gene cluster expression system in heterologous Aspergillus oryzae. J Biosci Bioeng 106:466–472

    Article  PubMed  CAS  Google Scholar 

  • Sarkar A, Funk AN, Scherlach K, Horn F, Schroeckh V, Chankhamjon P, Westermann M, Roth M, Brakhage AA, Hertweck C (2012) Differential expression of silent polyketide biosynthesis gene clusters in chemostat cultures of Aspergillus nidulans. J Biotechnol 160:64–71

    Article  PubMed  CAS  Google Scholar 

  • Schloss PD, Handelsman J (2004) Status of the microbial census. Microbiol Mol Biol Rev 68:686–691

    Article  PubMed  PubMed Central  Google Scholar 

  • Schmaler-Ripcke J, Sugareva V, Gebhardt P, Winkler R, Kniemeyer O, Heinekamp T, Brakhage AA (2009) Production of pyomelanin, a second type of melanin, via the tyrosine degradation pathway in Aspergillus fumigatus. Appl Environ Microbiol 75:493–503

    Article  PubMed  CAS  Google Scholar 

  • Schroeckh V, Nützmann HW, Brakhage AA (2014) Fungal-actinomycete interactions-wakening of silent fungal secondary metabolism gene clusters via interorganismic interactions. In: Osbourn A, Goss RJ, Carter GT (eds) Natural Products: Discourse, diversity and design. Hoboken, Wiley, pp 147–158

    Chapter  Google Scholar 

  • Seyedsayamdost MR (2014) High-throughput platform for the discovery of elicitors of silent bacterial gene clusters. Proc Natl Acad Sci U S A 111:7266–7271

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Shao Z, Rao G, Li C, Abil Z, Luo Y, Zhao H (2013) Refactoring the silent spectinabilin gene cluster using a plug-and-play scaffold. ACS Synth Biol 2:662–669

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Shwab EK, Bok JW, Tribus M, Galehr J, Graessle S, Keller NP (2007) Histone deacetylase activity regulates chemical diversity in Aspergillus. Eukaryot Cell 6:1656–1664

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Skinnider MA, Dejong CA, Rees PN, Johnston CW, Li H, Webster AL, Wyatt MA, Magarvey NA (2015) Genomes to natural products PRediction informatics for secondary metabolomes (PRISM). Nucleic Acids Res 43:9645–9662

    PubMed  PubMed Central  CAS  Google Scholar 

  • Spieckermann A (1912) Trink- und Gebrauchswasser. Zeitschrift für Lebensmitteluntersuchung und -Forschung A 24:710

    Google Scholar 

  • Spohn M, Kirchner N, Kulik A, Jochim A, Wolf F, Muenzer P, Borst O, Gross H, Wohlleben W, Stegmann E (2014) Overproduction of ristomycin A by activation of a silent gene cluster in Amycolatopsis japonicum MG417-CF17. Antimicrob Agents Chemother 58:6185–6196

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Starcevic A, Zucko J, Simunkovic J, Long PF, Cullum J, Hranueli D (2008) Clust scan: an integrated program package for the semi-automatic annotation of modular biosynthetic gene clusters and in silico prediction of novel chemical structures. Nucleic Acids Res 36:6882–6892

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Stevens DL, Ma Y, Salmi DB, McIndoo E, Wallace RJ, Bryant AE (2007) Impact of antibiotics on expression of virulence-associated exotoxin genes in methicillin-sensitive and methicillin-resistant Staphylococcus aureus. J Infect Dis 195(2):202–211

    Article  PubMed  CAS  Google Scholar 

  • Stevenson BS, Eichorst SA, Wertz JT, Schmidt TM, Breznak JA (2004) New strategies for cultivation and detection of previously uncultured microbes. Appl Environ Microbiol 70:4748–4755

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Takano E (2006) Gamma-butyrolactones: Streptomyces signaling molecules regulating antibiotic production and differentiation. Curr Opin Microbiol 9:287–294

    Article  PubMed  CAS  Google Scholar 

  • Tiedje JM (1994) Microbial diversity: of value to whom? ASM News 60:524–525

    Google Scholar 

  • Torsvik VL, Ovreas TF (2002) Prokaryotic diversity magnitude, dynamics, and controlling factors. Science 296:1064–1066

    Article  PubMed  CAS  Google Scholar 

  • Ugai T, Minami A, Gom K, Oikawa H (2016) Genome mining approach for harnessing the cryptic gene cluster in Alternaria solani: production of PKS–NRPS hybrid metabolite, didymellamide B. Tetrahedron Lett 57(25):2793–2796

    Article  CAS  Google Scholar 

  • Van Lanen SG, Shen B (2006) Microbial genomics for the improvement of natural product discovery. Curr Opin Microbiol 9:252–260

    Article  PubMed  CAS  Google Scholar 

  • van Wezel GP, McKenzie NL, Nodwell JR (2009) Applying the genetics of secondary metabolism in model actinomycetes to the discovery of new antibiotics. Methods Enzymol 458:117–141

    Article  PubMed  CAS  Google Scholar 

  • VanderMolen KM, Darveaux BA, Chen WL, Swanson SM, Pearce CJ, Oberlies NH (2014) Epigenetic manipulation of a filamentous fungus by the proteasome-inhibitor bortezomib induces the production of an additional secondary metabolite. RSC Adv 4:18329–18335

    Article  PubMed  CAS  Google Scholar 

  • Vögtli M, Chang PC, Cohen SN (1994) afsR2: a previously undetected gene encoding a 63-amino-acid protein that stimulates antibiotic production in Streptomyces lividans. Mol Microbiol 14:643–653

    Article  PubMed  Google Scholar 

  • Wald P, Pitkanen S, Boddy L (2004) Interspecific interactions between the rare tooth fungi Creolophus cirrhatus, Hericium erinaceus and H. coralloides and other wood decay species in agar and wood. Mycol Res 108:1447–1457

    Article  PubMed  Google Scholar 

  • Waldron C, Matsushima P, Rosteck PR Jr, Broughton MC, Turner J, Madduri K, Crawford KP, Merlo DJ, Baltz RH (2001) Cloning and analysis of the spinosad biosynthetic gene cluster of Saccharopolyspora spinosa. Chem Biol 8:487–499

    Article  PubMed  CAS  Google Scholar 

  • Weber T (2015) antiSMASH 3.0-a comprehensive resource for the genome mining of biosynthetic gene clusters. Nucleic Acids Res 39:W339–W346

    Google Scholar 

  • Weber T, Rausch C, Lopez P, Hoof I, Gaykova V, Huson DH, Wohlleben W (2009) CLUSEAN: a computer-based framework for the automated analysis of bacterial secondary metabolite biosynthetic gene clusters. J Biotechnol 140:13–17

    Article  PubMed  CAS  Google Scholar 

  • Westhoff S, van Leeuwe TM, Qachach O, Zhang Z, van Wezel GP, Rozen DE (2017) The evolution of no-cost resistance at sub-MIC concentrations of streptomycin in Streptomyces coelicolor. ISME J 11(5):1168–1178

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Whitman WB, Coleman DC, Wiebe WJ (1998) Prokaryotes: the unseen majority. Proc Natl Acad Sci U S A 95:6578–6583

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Wilkinson B, Micklefield J (2007) Mining and engineering natural-product biosynthetic pathways. Nat Chem Biol 3:379–386

    Article  PubMed  CAS  Google Scholar 

  • Wright G (2014) Perspective: synthetic biology revives antibiotics. Nature 509:S13

    Article  PubMed  CAS  Google Scholar 

  • Wright G (2015) Antibiotics: an irresistible newcomer. Nature 517:442–444

    Article  PubMed  CAS  Google Scholar 

  • Xu HS, Roberts N, Singleton FL, Attwell RW, Grimes DJ, Colwell RR (1982) Survival and viability of nonculturable Escherichia coli and Vibrio cholerae in the estuarine and marine environment. Microb Ecol 8:313–323

    Article  PubMed  CAS  Google Scholar 

  • Xu M, Wang Y, Zhao Z, Gao G, Huang SX, Kang Q, He X, Lin S, Pang X, Deng Z, Tao M (2016) Functional genome mining for metabolites encoded by large gene clusters through heterologous expression of a whole-genome bacterial artificial chromosome library in Streptomyces spp. Appl Environ Microbiol 82:5795–5805

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Yamanaka K, Reynolds KA, Kersten RD, Ryan KS, Gonzalez DJ, Nizet V, Dorrestein PC, Moore BS (2014) Direct cloning and refactoring of a silent lipopeptide biosynthetic gene cluster yields the antibiotic Taromycin A. Proc Natl Acad Sci USA 111:1957–1962

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Yim G, Wang HH, Davies J (2006) The truth about antibiotics. Int J Med Microbiol 296(2–3):163–170

    Article  PubMed  CAS  Google Scholar 

  • Zengler K, Toledo G, Rappe M, Elkins J, Mathur EJ, Short JM, Keller M (2002) Cultivating the uncultured. Proc Natl Acad Sci U S A 99:15681–15686

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  • Zhu H, Sandiford SK, van Wezel GP (2014) Triggers and cues that activate antibiotic production by actinomycetes. J Ind Microbiol Biotechnol 41(2):371–386

    Article  PubMed  CAS  Google Scholar 

  • Ziemert N, Podell S, Penn K, Badger JH, Allen E, Jensen PR (2012) The natural product domain seeker NaPDoS: a phylogeny based bioinformatic tool to classify secondary metabolite gene diversity. PLoS One 7:e34064

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to Prof. Bhagirath Singh, Vice Chancellor, MGS University, Bikaner, Rajasthan, India for providing the departmental research grant.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dharmesh Harwani.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Begani, J., Lakhani, J. & Harwani, D. Current strategies to induce secondary metabolites from microbial biosynthetic cryptic gene clusters. Ann Microbiol 68, 419–432 (2018). https://doi.org/10.1007/s13213-018-1351-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13213-018-1351-1

Keywords