Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

SLAS Technology 29 (2024) 100120

Contents lists available at ScienceDirect

SLAS Technology
journal homepage: www.elsevier.com/locate/slast

Employing synthetic biology to expand antibiotic discovery


Greta D. Cook , Nikolas M. Stasulli *
Department of Biology and Environmental Science, University of New Haven, 300 Boston Post Rd, Dodds Hall 316, West Haven 06516 USA

A R T I C L E I N F O A B S T R A C T

Keywords: Antimicrobial-resistant (AMR) bacterial pathogens are a continually growing threat as our methods for
Synthetic biology combating these infections continue to be overcome by the evolution of resistance mechanisms. Recent thera­
Biosynthetic gene cluster (BGC) peutic methods have not staved off the concern of AMR infections, so continued research focuses on new ways of
Cryptic BGCs
identifying small molecules to treat AMR pathogens. While chemical modification of existing antibiotics is
Natural products (NPs)
Mutagenesis
possible, there has been rapid development of resistance by pathogens that were initially susceptible to these
Endogenous gene expression compounds. Synthetic biology is becoming a key strategy in trying to predict and induce novel, natural anti­
Heterologous gene expression biotics. Advances in cloning and mutagenesis techniques applied through a synthetic biology lens can help
characterize the native regulation of antibiotic biosynthetic gene clusters (BGCs) to identify potential modifi­
cations leading to more potent antibiotic activity. Additionally, many cryptic antibiotic BGCs are derived from
non-ribosomal peptide synthase (NRPS) and polyketide synthase (PKS) biosynthetic pathways; complex, clus­
tered genetic sequences that give rise to amino acid-derived natural products. Synthetic biology can be applied to
modify and metabolically engineer these enzyme-based systems to promote rapid and sustainable production of
natural products and their variants. This review will focus on recent advances related to synthetic biology as
applied to genetic pathway characterization and identification of antibiotics from naturally occurring BGCs.
Specifically, we will summarize recent efforts to characterize BGCs via general genomic mutagenesis, endoge­
nous gene expression, and heterologous gene expression.

1. Introduction antibiotic resistant genes, bacteria possessing multiple ARGs become


labeled multidrug-resistant (MDR) or extremely drug resistant (XDR) [1,
The current antibiotic resistance crisis can largely be attributed to 6]. One problem with current broad-spectrum antibiotics is they lack
the overuse, misuse, and lack of new discoveries of antibiotic treat­ chemical diversity [8,9]; over 70 % of antibiotics developed between
ments. The identification of ‘modern’, biologically produced antibiotics 1981 and 2005 being modifications of four basic chemical scaffolds
began with Sir Alexander Fleming’s discovery of penicillin in 1928 (cephalosporins, macrolides, penicillins, and quinolones) [10]. More
[1–3]. After the first sulfonamide antibiotic was introduced for use in distinct chemical scaffolds such as glycopeptides, lipopeptides, amino­
humans in 1937, it didn’t take more than a hand full of years before glycosides, and tetracyclines are also produced in some more specific
resistance was reported [4]. Alongside the use of antibiotics to treat antibiotics, but they are applied narrowly in certain circumstances for
infection, antimicrobial-resistant (AMR) pathogens have evolved to selected bacterial infections [8].
overcome many of the antibiotics that are now commonly used [1,5,6]. Many antibiotics are naturally occurring, secreted compounds from
Antibiotic resistance can occur through genetic mutations within the bacteria and fungi and are therefore considered natural products (NPs).
genome or through horizontal gene transfer (HGT), acquiring foreign They are produced by microbes in response to interactions within their
DNA containing antibiotic resistance genes (ARGs) via transformation, communities and have vast chemical diversity and complexity, giving
transduction, or conjugation [6,7]. With the increase in use of antibi­ them more diverse bioactivity than we can currently generate on an
otics across human populations, animal populations, and within the industrial level [8]. Antibiotic NPs are considered secondary, or
environment (agriculturally) the likelihood of bacteria interacting via specialized, metabolites and are synthesized from metabolic in­
HGT to evolve, acquire, and accumulate new ARGs, making them termediates. They are considered secondary metabolites because they
resistant to multiple antibiotics, increases. Through the accumulation of are not required for the growth or survival of the bacteria, but rather

* Corresponding author.
E-mail address: nstasulli@newhaven.edu (N.M. Stasulli).

https://doi.org/10.1016/j.slast.2024.100120
Received 16 June 2023; Received in revised form 4 January 2024; Accepted 7 February 2024
Available online 8 February 2024
2472-6303/© 2024 The Authors. Published by Elsevier Inc. on behalf of Society for Laboratory Automation and Screening. This is an open access article under the
CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

play important roles in protection, molecular signaling, and environ­ expression focuses on the manipulation of antibiotic BGC transcription
mental interactions [11]. Natively produced antibiotics are small mol­ through cultural condition modifications, classical genetic techniques,
ecules containing complex chemical structures that act as a type of and chemical genetic techniques [12,24]. Heterologous gene expression
microbial warfare in the fight for resources. These small molecule NPs focuses on the reconstruction of BGCs into model microbial strains and
have evolved to interact with biological macromolecules, making novel involves cloning and reconstructing biosynthetic pathways [25,26].
NPs excellent candidates for new antibiotics [8]. As noted previously, Being able to activate, characterize, and manipulate antibiotic BGCs
the NPs we currently know of produce chemical diversity around basic from microbes is one of our best strategies for identifying novel, or more
antibiotic molecule scaffolds: peptides, polyketides, carbohydrates, al­ potent, antibiotics that can help address the growing concern of AMR
kaloids, or terpenes backbones [8]. These structural cores of secondary pathogens. Synthetic biology can offer new options to fight the rapid
metabolite antibiotics are produced by biosynthetic enzyme families: emergence of AMR by genetically engineering bacteria to identify new
nonribosomal peptide synthetases (NRPS), polyketide synthases (PKS), scaffolds, enzymatically modify existing scaffolds, and purify com­
terpene synthases (TPS), and dimethylallyl-tryptophan synthases pounds to generate chemical libraries for antibiotic screening. With the
(DMATS) [12–14]. This core is further modified by tailoring enzymes help of these technologies, the efforts around BGC manipulation and
and regulatory proteins to create chemical diversity. The genes encoding characterization are developing rapidly and can be the driver for the
the biosynthetic enzymes, tailoring enzymes, regulatory proteins, and next generation of antibiotics. Recent reviews on activating cryptic BGCs
transportation genes are all clustered around the same location in the [27] and genomic engineering through synthetic biology [28] have been
chromosome [12]. These clusters are collectively known as biosynthetic thorough, however, in this review we focus specifically on recent
gene clusters (BGCs). application of synthetic biology for unlocking BGCs to identify and
Biosynthetic gene clusters encode many small molecule NPs, or characterize novel antibiotics.
secondary metabolites, in bacteria [15]. These BGCs are being targeted
for their efficient production of complex natural products that would be 2. Flexible genomic mutagenesis strategies
much harder to generate under laboratory settings. One issue that arises
when studying BGC is that many are transcriptionally and translation­ Mutations in an organism’s DNA, via various mutagenesis mecha­
ally silent under typical laboratory growth conditions, making any po­ nisms, are responsible for generating both genotypic and phenotypic
tential secondary metabolite, itself, inaccessible and difficult to study variation in a population [29]. Through molecular editing techniques
[15,16]. Secondary metabolites are usually regulated by an external bacterial genomes can be artificially edited by purposefully inducing
stimulus; if the stimulus is absent the metabolite will not be synthesized mutations. Synthetic biologists can take advantage of mutagenesis
[12]. BGCs that produce secondary metabolites are detectable in mi­ mechanisms in the lab setting to search for novel antibiotic variants after
crobial genome sequences and can be identified using genomic inducing mutations into genomes [30]. While this is the optimal
sequencing, and automated genome mining tools [16]. Programs such as outcome in a lab setting, naturally occurring mutations can be either
Secondary Metabolite Unique Regions Finder (SMURF) [17] and Anti­ advantageous or deleterious to the bacteria’s survival depending on its
biotics and Secondary Metabolite Analysis Shell (antiSMASH) [18,19] environment. Mutagenesis strategies employed in synthetic biology,
use advanced computer bioinformatics algorithms to predict and iden­ whether random or site-directed, aim to create and identify advanta­
tify BGCs within genomic data [12]. BGCs that are identified, but not geous mutations that have affected the BGCs involved in antibiotic
associated with known secondary metabolites, are termed “cryptic” production. Techniques used for inducing directed mutations to genet­
because they have been previously undetectable or transcriptionally ically modify bacteria include CRISPR systems [31], recombineering
silent. Once these cryptic BGCs are located in the genome bioengi­ [32], MAGE [33], and Targetron vectors [34]. While some of the tech­
neering techniques can be applied to manipulate the genome, stimu­ niques discussed in this section have direct applications to endogenous
lating the expression of the encoded NP or potentially modifying it to and heterologous gene expression systems, we aim to give an overview
generate more chemical diversity. of common mutagenesis strategies synthetic biologists could take
Bioengineering and molecular cloning principles and techniques can advantage of when activating and studying cryptic BGCs (Table 1).
be applied to biological systems that produce protein or small molecule Recombineering, or recombination-mediated genetic engineering,
products that have defined and controllable properties. These tech­ describes the incorporation of mutations into a genome by surrounding
niques generally aim to transform genes, gene circuits, and metabolic the desired modifications with homologous regions of DNA. [32,35].
pathways to modify, reinvent, or build novel biological systems [20,21]. These homologous nucleotide sequences are identical to sequences
Synthetic biology can take advantage of these techniques to contribute present in the genome where the mutation or genetic element being
to the fight against AMR bacterial pathogens by expanding the chemical introduced will be located. Homologous recombination-based strategies
diversity of antibiotics through activating and manipulating cryptic introduce specific genetic elements (full genes, small mutations, new or
BGCs encoding potentially novel antibiotic secondary metabolites. modified promoters, etc.) into the genome and require various, some­
Techniques focused on genomic mutagenesis, endogenous gene times bacteria specific, recombination proteins along with the known
expression, and heterologous gene expression can be utilized to elicit homologous DNA sequences [35,36]. One common example of this type
and study potentially novel antibiotics from understudied and envi­ of system is the λ-Red recombinase system [37] that can be used for
ronmental microbes [22,23]. Outside of antibiotic discovery and pro­ introducing insertions, deletions, or point mutations. This mechanism
duction, synthetic biology also has potential applications in biofuel utilizes the introduction of the desired mutation amongst homologous
production, industrial chemical/natural product substitution synthesis, genomic regions of DNA for recombination [35]. The λ-Red recombinase
biomedical and bacterial infection applications, disease diagnosis, and system uses the lambda bacteriophage and Exo, Beta, and Gam proteins
treatment through the same methods that can be used to address the to introduce genomic mutations [36,37]. The Gam protein protects
AMR crisis [20]. linear DNA from endogenous RecBCD and SbcCD nucleases [38]. The
In this review, synthetic biology’s role in studying cryptic antibiotic Exo protein is a double stranded DNA-dependent exonuclease possessing
BGCs through genomic mutagenesis, endogenous gene expression, and 5′ to 3′ activity to degrade dsDNA to create ssDNA or partially degraded
heterologous gene expression will be explored. Within mutagenesis dsDNA with 3′ overhangs [38,39]. The Beta protein protects the ssDNA
strategies, genetically modifying bacteria, we review recombination- created by Exo protein and aids in annealing within the target cell [39].
mediated genetic engineering (recombineering), Multiplex Automated By introducing mutations, the λ-Red recombinase system aims to modify
Genome Engineering (MAGE), Targetrons, Global transcription ma­ genes within the biosynthetic gene cluster to exchange modules of BGCs
chinery engineering (gTME), and clustered regularly interspaced short to produce second-generation derivatives of existing antibiotics. As an
palindromic repeats (CRISPR-Cas9) systems. Endogenous gene example, the λ-Red recombinase method was used to inactivate clo-hal

2
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

Table 1 protein-based transcriptional interference system that can control pro­


Summary of flexible mutagenesis strategies. tein expression levels. This CRISPRi technology silences transcription
Category Strategy Examples of use using designed guide-RNA and a mutant Cas9 protein without replacing
or modifying the gene promoter region [30,50]. While this system is
Recombination-mediated Methods of DNA mutagenesis [32,35–42]
genetic engineering relying on homologous more in line with endogenous expression control, it will be covered here
(recombineering) sequences surrounding the for content consistency. CRISPRi systems have the potential to offer
region of interest specific gene knockouts with little to no off-target binding [51]. The
CRISPR/Cas9 systems Methods of DNA mutagenesis • Basic CRISPR/ mutant dCas9 protein lacks nuclease activity but retains the ability to
relying on specific targeting of Cas [43–46]
the exact DNA sequence that is to • with EvolvR
form a complex with the guide-RNA. When binding to protein-coding
be modified. Relies on designed [47,48] genes, the CRISPRi technique can block transcription, controlling gene
RNA molecules for targeting • CREATE [31, expression without altering the genome sequence. The CRISPRi tech­
DNA and a Cas endonuclease for 49] nique has been successful in knocking out gene expression within yeast
cleavage of DNA at the site of • CRISPRi
[52] and E. coli [50]. CRISPRi methods have the potential to be utilized
mutagenesis. [50–52]
Multiplex automated High-throughput methods, using • Basic MAGE in studying BGCs by selectively knocking down the expression-specific
genome engineering specifically designed pools of [55] genes within a gene cluster to study regulation or modifications of
(MAGE) synthetic oligonucleotides, that • Cos-MAGE antibiotic compounds. CRISPR/Cas9 systems have become a tool in
induce mutations by taking [33] synthetic biology for their precise editing, multigene editing, and precise
advantage of natural cellular • with CRISPR
processes. [56,58]
regulating as recently reviewed thoroughly by Li et al. [53]. They are
• Port-MAGE effective tools for generating targeted mutations with potentially
[57] high-throughput capacity [54].
• dReaMGE A more high-throughput technique (a feature that is always desirable
[60]
when bio-prospecting for new or modified molecules) the Multiplex
Targetrons Methods of using retrohoming • Basic
with a modified type-II intronic targetron [34, Automated Genome Engineering (MAGE) method, was developed at the
RNA, with ribozyme and reverse 61–63,67] Wyss Institute, Harvard Medical School, to mimic the natural principles
transcriptase activity, to target • ClosTron [64] of evolution in genome editing [55]. MAGE uses synthetic oligonucle­
mutation incorporation into the • with CRISPR otide pools and the intrinsic bacterial cell cycle, along with nucleotide
genome. [63,65,66]
introduction (cell induction, incorporation of ssDNA, and trans­
Global transcription Using error-prone polymerase [58,68–71]
machinery engineering chain reaction to generate a formation via electroporation) and cell recovery (outgrowth) methods,
(gTME) mutant library, usually targeting to genetically modify cells and grow them over generations. The MAGE
a transcription factor that may system, for the first time, allowed multiple, small mutational insertions
regulate BGCs.
to be introduced at different locations simultaneously [30,55]. Multiple
constructed, site-specific DNA segments are introduced to a bacterial
and cloz genes within the biosynthetic gene cluster responsible for population and the cells are manipulated, via transformation tech­
producing the antibiotic clorobiocin. The BGC was then engineered to niques, to take up the DNA, ultimately leading to homologous recom­
add a methyl group to create a clorobiocin analog. Unfortunately, in this bination and introduction of genomic diversity into the population.
case, this modification reduced potency compared to the original com­ After each recovery period, post mutation incorporation, more DNA can
pound [40]. This method has also been used for the biosynthesis of the be transformed to promote the accumulation of insertions. All of the
antibiotic daptomycin (cubicin), which has been approved in the US to oligonucleotides are specifically created, not randomly generated se­
treat skin infections caused by Staphylococcus aureus [41]. quences, so this method can be used by synthetic biologists to target
Second-generation derivatives of daptomycin have been made using the various parts of a BGC at once. The MAGE method has been modified
lambda-red recombinase system by modifying multiple modules of its and refined to tailor to new hypotheses and desired outcomes of ex­
NRPS gene cluster [42]. periments. The Co-selection MAGE (CoS-MAGE) [33],
The clustered regularly interspaced short palindromic repeats CRISPR-optimized MAGE-recombineering [56], and pORTMAGE [57]
(CRISPR)/Cas9 system is becoming more and more popular and is used systems are examples of refined MAGE techniques. CoS-MAGE selects
as a genome editing tool. CRISPR gene editing will be mentioned several subpopulations of cells that already have acquired mutations and are
times as part of the improvement of other techniques, but is also useful known to be more-highly competent. The selecting of subpopulations
on its own, allowing for site-specific gene editing within bacterial cells already containing a mutation increases the likelihood of adding more
[43]. The CRISPR system is relatively easy to use and constantly being mutated sites through further rounds of MAGE; the selected clones being
refined and modified to manipulate the genome [44,45]. This system more electrocompetent or more amenable to olgio-based editing. This
targets specific DNA sites through complementary base pairing of selection of known, recombination-competent cells allows for a higher
designed guide-RNA, inducing a double-strand DNA break, via the Cas9 recombination efficiency of larger insertions (<2 % efficiency for >20
nuclease, at a specific protospacer adjacent motif (PAM) sequence. bp insertion in the original MAGE protocol) during subsequent rounds of
Either non-homologous end-joining or homology-directed repair MAGE DNA incorporation. This increased efficiency of incorporation of
mechanisms are used to complete the editing procedure after the large base pair insertions can allow for directed insertion of entire
introduction of the desired mutation [43,46]. The CRISPR/Cas system promoter sequences at more than one site in the genome [30,33].
can be applied with the EvolvR system [47] for the introduction of However, the CoS-MAGE system was not sufficient to improve the
semi-random mutations at locations targetable by CRISPR/Cas9. A overall recombineering efficiency of the first round of MAGE. An addi­
mutant nicking Cas9 protein (nCas9) is coupled to an error-prone DNA tional adaptation using CRISPR/Cas system was applied to further
polymerase I to introduce mutations [48]. This EvolvR system could be optimize recombineering [56,58]. The CRISPR-optimized MAGE-r­
used to target and induce mutations to activate and/or modify BGCs. ecombineering system allows for the introduction of mutations within
CRISPR has also been used to generate and identify genetic variations multiple genomic locations at a much higher efficiency [58]. The MAGE
using the CRISPR-enabled trackable genome engineering (CREATE) system also developed into the portable MAGE (pORTMAGE) system
system [30,49]. The CREATE system can be used to produce genetic [57] to introduce multiple mutations to many bacterial genomes
variant libraries for evaluation [31]. A variation of the CRISPR system, simultaneously using a single plasmid for transformation with little to no
CRISPR interference (CRISPRi), uses a nuclease-deficient Cas9 (dCas9) off-target mutagenesis [57]. Antithetical to the goals of this review, the
pORTMAGE system has been used to introduce 10 antibiotic-resistance

3
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

mutations into the genomes of Salmonella enterica and Escherichia coli to has the potential to modulate BGC expression by employing random
study the conservation of antibiotic-resistant molecular mechanisms mutagenesis to activate BGC transcription and produce novel antibiotic
within these bacteria [57]. Overall, the MAGE system edits the bacterial diversity. Synthetic biology has been applied in and along with muta­
genome by inducing mutations at multiple target sites to generate genesis strategies to try and induce mutations at specific locations,
functional changes in the cell and can be applied in synthetic biology ideally, not randomly, to activate cryptic BGCs. Mutagenesis strategies
research to modify promoters, ribosomal-binding sites (RBSs), or protein optimally stimulate changes within the genome to alter gene expression
coding genes to optimize biosynthetic pathways for NP production [59]. and are being applied to cryptic BGCs to activate them directly or
Most recently, the double-stranded DNA Recombineering-assisted indirectly by knocking out or in genomic content in hopes of inducing
Multiplex Genome Engineering (dReaMGE) system allows for rapid the novel NP production.
mutagenesis through insertions and deletions of kilobases at multiple
sites without double-strand breaks or mismatch repair [60]. The 3. Endogenous gene expression
dReaMGE system has been applied to manipulate BGCs for inactivation,
promoter replacement, and combinatorial modification of transcrip­ Endogenous, or homologous, gene expression specifically focuses
tional regulators for the improved production of NPs and the discovery around altering the levels of a gene being transcribed within an organ­
of additional derivatives [60]. Specifically, In Schlegelella brevitalea DSM ism [72], and generally refers to overexpression or repression of genes
7029 dReaMGE was used to simultaneously replace native promoters of within the organism it originates. The advantage of studying a gene
the glbC, glbF, and glbB genes within the glb BGC to enhance the pro­ endogenously is that it is tested in the context in which it resides and
duction of the antibiotic glidobactin A [60]. physiological relevance is more likely to be observed. However,
Site-specific mutagenesis can also be achieved through Targetrons, endogenous studies are not always possible. This may be most relevant
modified mobile type-II introns that target specific DNA sites by RNA- when trying to identify novel antibiotics since some environmentally
DNA base pairing. Type-II introns are capable of inserting themselves identified NP producers are not culturable under lab conditions; pre­
into DNA sequences after they are spliced out of RNA during post- venting the use of endogenous expression studies [27,73–78]. Within
transcriptional processing. The RNA intron utilizes its ribozyme activ­ synthetic biology BGC studies, procedures around endogenous expres­
ity, breaking and forming bonds with nucleic acids, to reverse splice and sion focus mainly on the manipulation of promoters, the overexpression
insert directly into the target DNA site [34,61]. This ability of ribozymes of transcriptional activators, and the elimination of transcriptional re­
to insert into specific DNA target sequences is termed retrohoming and is pressors to control transcription [12]. In the context of searching for
the basis of this technique [62]. Targetrons are redesigned type-II novel antibiotic BGCs, these techniques are most effectively utilized in
intronic RNA, used for site-specific mutagenesis, consisting of an auto­ cases where a culturable and genetically manipulatable organism has
catalytic intronic RNA (ribozyme) and an intron-encoded reverse tran­ been found to contain cryptic gene clusters (Table 2).
scriptase [28,63]. Utilizing RNA-based introns is advantageous because As noted previously, cultivation conditions of an organism can
they are independent of the host’s recombination machinery and impact gene expression and the expression of BGCs. More times than
contain heterologous sequences that can be easily re-targeted for future not, what is ‘optimal’ growth as defined in a lab setting is not optimal for
gene delivery and insertions [63]. ClosTron technology is one the expression of BGCs. Alterations in temperature and nutrient avail­
Targetron-based system used to successfully edit bacterial genomes [28, ability have been shown to contribute to the activation of BGCs and their
64]. The ClosTron system incorporates a specialized natural product synthesis [74,79,80]. For example, a 2 ◦ C increase in the
retrotransposition-activated selectable marker (RAM) site within the culture temperature of Streptomyces hygroscopicus increased the tran­
type-II intron. The RAM is an inactivated marker that becomes activated scription of the validamycin BGC [79]. Typically, culture modification is
by the relocation and insertion of the type-II intron [64]. The ClosTron the first step in trying to activate cryptic BGCs by replicating environ­
system was initially based on the ermB gene encoding for erythromycin mental stressors. One method of altering culturing conditions to stim­
resistance [64]. The ermB gene is inactivated by the insertion of a DNA ulate NP production is called one strain-many compounds (OSMAC)
fragment including a type-I intron in a way that blocks splicing of the [81]. OSMAC has enhanced the production of terrosamycin, through test
mRNA transcript, resulting in no production of the ErmB protein growth Streptomyces sp. RKND004 in 14 different media, which has
(leaving the bacteria now susceptible to erythromycin) [64]. RAM ele­ antibacterial activity similar to other polyether ionophores and also
ments specific to other antibiotic resistance genes should theoretically demonstrated anticancer activates [82]. OSMAC methods can be effec­
be able to be created using specific exon sequences required for splicing tive at mimicking the environmental stress conditions to activate cryptic
within the targeted structural gene relating to antibiotic resistance [64]. BGCs but they lack the presence of other microbes and their interactions;
Targetron systems can also be paired with CRISPR to overcome an in­ another contributing factor to the modulation of gene expression. Mi­
tron’s limitation of splicing and insertion when containing foreign DNA crobial cocultures, which contain two or more bacterium, take advan­
[63,65,66]. The system has also recently been modified to include tage of inter-bacterial interactions to try and activate cryptic metabolites
temperature induction of targetron mutagenesis (Thermotargetron) [83–88]. The paradigms for interaction are most often nutrient
[67]. Synthetic biologists can take advantage of the ability of targetrons changes/starvation, signaling molecules, physical contact, and antibi­
to knock in and knock out sequences, or full genes, and may be able to osis via cytotoxic compounds [27,89]. For example, the biosynthesis of
manipulate gene function or regulation to optimize the expression of NPs is altered when colonies of Streptomyces coelicolor and Myxococcus
BGCs. xanthus are grown next to each other in low-iron conditions. In this
A final mutagenesis strategy useful to synthetic biologists may be the environment, M. xanthus upregulates a BGC expressing an iron-chelating
Global transcription machinery engineering (gTME) system, a muta­ siderophore, myxochelin, while S. coelicolor upregulates a BGC
genesis strategy using error-prone polymerase chain reaction (PCR) to expressing an antibiotic, actinorhodin, that inhibits M. xanthus growth
produce a random mutant library with genetic variation in a desired [90,91]. Environmental stress and the presence of different microbes
transcription factor(s) [30,58]. These mutated transcription factors will can obviously influence the NP synthesis, potentially unlocking cryptic
ideally generate transcriptomic expression changes during growth to antibiotics. However, when altering culture conditions fails to activate
unlock specific phenotypes [68,69]. The gTME strategy was first used in BGCs and their natural product expression we can turn to synthetic
yeast to improve its ethanol tolerance and the production efficiency of biology for other endogenous expression methods via both classical and
glucose to ethanol for applications in biofuel production [70]. It has also chemical genetics [27].
been successfully used in bacteria to engineer organic-solvent tolerant The alteration of bacterial genomes through classical genetics, to
E. coli strains for industrial applications [68,71]. While not yet used in manipulate endogenous gene expression, includes techniques such as
the search for naturally created antibiotic variations, the gTME method promoter manipulation, transcription factor manipulation, and

4
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

Table 2 stambomycins [101], and in Streptomyces venezuelae to discover a new


Summary of endogenous gene expression strategies. biaryl polyketide, venemycin [102]. The inactivation of repressors
Category Strategy Implementation through gene deletion or transcription factor decoys can also increase
the expression of BGCs and their natural product synthesis [103].
Cultural Condition Growth condition • Culture the same bacterial
Modifications alterations: one strain strain in media with Recently, hybrid regulatory proteins like LuxR- and LysR-type regulators
many compounds differing growth conditions with native DNA binding domains and ligand-binding domains have
(OSMAC) to elicit or modify the been used to control the expression of BGCs [104]. For example, a LuxR
production of metabolites response regulator, which regulates a cryptic BGC in Pseudomonas sp.
[74,79–82]
○ Temperature
SZ57, was replaced with a mutated LuxR containing its DNA-binding
○ Carbon source and other domain and ligand-binding domains from a LasR protein and a Lux
nutrients protein capable of binding to 3-oxo-decanoyl-homoserine lactone (HSL)
○ Inducing stress
[104]. In the presence of the HSL ligand, the LuxR protein upregulated
Coculture • Involves multiple organisms
the cryptic BGC in Pseudomonas sp. SZ57 to discover the NP pseudo­
to model bacterial
interactions [83–88] momol [104]. In addition to upregulated hybrid proteins, synthetic and
• May allow for both physical artificial transcription factors can also upregulate the expression of BGCs
and/or chemical [12]. Furthermore, genomic sequences can be specifically modified. For
communication [27,89–91] instance, upstream activation sequences (UASs) have been integrated
Classical Genetics Direct DNA • Reporter-guided mutant
using cloning modifications selection of BGCs [92–95]
into native promoters to reprogram the transcriptional regulation sys­
strategies • Regulatory gene deletion, tem of targeted genes to interact with specific transcription factors. This
(mutagenesis and duplication or modification method had been successful in Aspergilli [105], Ustilago maydis [106],
reporters) [96–104,108] and Trichoderma reesei [107]. However, the replacement of native pro­
• Promoter manipulation,
moters with inducible or constitutively active promoters, rather than
introduction, or
replacement [105,106,108] their modification, can be the most direct way to control BGC expression
Transcription factor • Creation of transcription [108]. The native promoter responsible for the production of 6‑epi-­
manipulation factor decoys to manipulate alteramides was replaced with a constitutive promoter, through
repressor binding [12,103] conjugation-mediated homologous recombination, using plasmid
• Manipulation of
pOJ313, to enhance its endogenous NP production [108]. An advantage
transcription factor levels
[105,107] to inserting new promoters is that the cryptic NP BGC being stimulated
• Usage of synthetic can be more directly controlled. However, because of the number of
transcription factor [105, genes present in some BGCs, not all genes are under control of a single
107]
promoter. Having multiple separate genes or operons can limit some
RNAP engineering • Mutations in ribosomal or
RNA polymerase genes that classical genetic methods since several promoter modifications may be
lead to transcriptional needed to address the different gene regions within a cluster; that is if all
changes [109–113] of the promoter regions can even be identified within the full BGC [27].
Chemical Genetics ‘Microbial hormone’ • The use of bacterial Along with promoter manipulation, other alterations to transcriptional
treatment signaling molecules to
and translational proteins are being explored for the activation and
manipulate gene expression
[114–118] production of NPs. Ribosome engineering or RNA polymerase (RNAP)
High throughput • Use reporter gene under engineering target and manipulate core biology dogma enzymes to
elicitor screening native promoter to detect create mutated transcription or translation machinery complexes that
(HiTES) transcription during
can be more stable under stressful environmental conditions, like amino
challenge with a screen of
natural product molecules
acid deprivation, to activate BGCs and encourage NP synthesis
[119–122] [109–111]. This method is based on the observation that development of
• Detection of transcriptional resistance to streptomycin, through an accrued mutation in ribosomal
changes from natural protein S12 (RpsL), not only made Streptomyces lividans
products without
streptomycin-resistant but upregulated the production of an unrelated
mutagenesis: Bioactivity
HiTES [123,124], antibiotic, actinorhodin [110]. This mutation, mapped to the Lysine-88
HiTES-imagine mass spec­ residue in RpsL, provides more structural stability and leads to increased
trometry [125] levels of off-target protein synthesis [110]. RNAP is responsible for
discovering the antibiotic NPs, piperidamycins, through the increased
promoter affinity of mutated RNAP for secondary metabolite BGCs
repressor deletion [27]. Strategies such as genome-wide and
biosynthesis [112,113].
transposon-based mutagenesis have been combined with reporter-based
Alternative methods of modifying endogenous gene expression are
assays and mass spectrometry to activate and discover new novel
through chemical genetics where small compounds are employed to
products possessing antimicrobial activity [73,92–95]. In one example,
activate silent BGCs. The aim is to test these small molecules in hopes
Guo et al. successfully applied reporter-guided mutant selection (RGMS)
that they elicit a modification in transcriptional and/or translational
to activate the pga silent BGC, encoding an unknown product, in Strep­
host machinery to activate silent BGCs. Mass spectrometry (MS), or
tomyces sp. PGA64. This technique employs random mutagenesis and a
some similar technique, can then be used for the detection of natural
promoter-reporter system to monitor transcription of targeted gene
products after the chemical challenge of the desired microbe [27].
clusters. This led to the discovery of two new anthraquinone amino­
Similarly to culture modifications, the addition of chemical signals can
glycoside antibiotics: gaudimycin D and E [94]. Silent BGC regulators
affect BGC activation. For example, ‘microbial hormone’ treatment has
can be induced to elevate natural product production through the
been used to inactivate transcriptional regulators, inhibiting the tran­
introduction of synthetic promoters or the insertion of multiple copies of
scription of BGCs to express natural products. In one example, the TetR
a regulatory gene [96–101]. For example, large ATP binding regulators
family of transcriptional regulators has been shown to regulate the
from the LuxR family (LAL) of proteins were constitutively overex­
expression of NP BGCs and keep them silent under laboratory growth
pressed to encourage binding to promoter regions upstream/within a
conditions [114,115]. ArpA, a TetR family transcriptional repressor
BGC in Streptomyces ambofaciens to identify glycosylated macrolides,
protein that regulates the production of streptomycin in Streptomyces

5
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

griseus [116], can be inactivated by ‘microbial hormone’ treatment using production of the reporter molecule. Any output from the reporter gene
the signaling molecule A-factor [117]. The A-factor molecule binds to is then initially assumed to be indicative of an activation of the targeted
ArpA causing it to release from the promoter of adpA, which encodes BGC [119]. The HiTES method was applied to Burkholderia thailandensis,
global activators of secondary metabolism. With ArpA inactivated, the using a library of NPs and clinical agents as elicitors, to express the
production of streptomycin increases [118]. Being able to artificially cryptic malleilactone (mal) BGC [119]. Using a malL-lacZ reporter strain
encourage the overproduction of potentially novel antibiotics would of B. thailandensis, which created a translational fusion of the LacZ re­
make it easier to study their structure, chemical properties, and the porter to the MalL protein that is essential for malleilactone synthesis,
factors contributing to their production. Chemical genetics can also low-dose antibiotics trimethoprim and piperacillin were discovered to
incorporate synthetic biology approaches and utilize more high be the best elicitors of the mal BGC. [119–121]. While the BGC in this
throughput methods that rely on mutagenesis. One example is the high example encodes a virulence factor involved in nematode infections
throughput elicitor screening (HiTES) method that specifically utilizes [119] this technique is adaptable to study antibiotic BGCs as well [122].
small, synthetic elicitor molecules to induce BGC expression [119]. In fact, this technique has been adapted to even more high-throughput
Through this technique a reporter gene is cloned inside the targeted BGC approaches that don’t require mutagenesis of the organism in ques­
where it could be transcribed under a native promoter. This reporter tion: Bioactivity-HiTES [123,124] and HiTES-IMS [125]. Overall,
strain, for the specific BGC, is then challenged with a predetermined set endogenous expression techniques aim to activate silent BGCs natively
of elicitor molecules and the bacteria cultures are monitored for to produce novel antibiotics within the original bacterial strain.

Fig. 1. A visual depiction of a heterologous gene expression scheme indicating strategies that could be utilized in synthetic biology to target BGCs for expression
and study.

6
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

Synthetic biology is applied to endogenous expression methods by linearized TAR cloning vectors contain homologous sequences of target
building promoters, repressing transcriptional regulators, modifying BGCs to be used as recognition sequences for homologous recombina­
receptor proteins, and employing small chemical molecules to elicit a tion [16,152]. These sequences are also called “homology arms” and
transcriptional or translational change, generally in tandem with culti­ sequentially match either end of the targeted BGC. The TAR vector is
vation conditions and coculturing, to activate cryptic BGCs. linearized and homologously combined with the target BGC of isolated
host DNA to form a BAC or YAC to be transformed into a heterologous
4. Heterologous gene expression host for NP production research [152].
After BGCs have been mobilized, they are genetically modified to fit
In opposition to endogenous expression, heterologous gene expres­ the needs of the heterologous bacterial host. Generally, native regula­
sion relates to incorporating and altering the levels of a non-native gene, tory regions, like promotors, are replaced with inducible or
or genes, that originate(s) from a distinct species or cell type. With the constitutively-active promoters. Typically, Red/ET recombination and λ
development of synthetic biology, heterologous gene expression has phage proteins (Redα and Redβ) or Rec phage proteins (RecE and RecT)
been able to better characterize NPs produced by BGCs, especially those are utilized to replace or inactivate sections of the target BGC [16].
from unculturable organisms [25,71,126–133]. Heterologous gene These promoters are reprogrammed with the goal of optimizing the
expression has been the main focus of synthetic biology to discover expression of BGCs within the heterologous host. The availability of
novel antibiotics. These heterologous expression systems generally use synthetic parts, such as promoters and ribosome binding sites (RBSs),
cloning to express the BGCs from unculturable microbes in a heterolo­ has allowed for the manipulation of BGCs for specific gene expression,
gous host [16,134]. In short, this process involves first identifying and can increase and improve NP production, or can activate silent BGCs
mobilizing target BGCs onto a cloning vector for genetic exchange with a [25]. A suitable heterologous host is necessary for successful, high-level
more easily manipulated and cultured host. The regulatory elements of NP production [16,154].
the BGC must then be modified on the cloning vector to match the new After editing the vectors, they need to be delivered into the heter­
host bacteria before genetic exchange can occur. After horizontal gene ologous host for cloning and induction of potential NP expression.
transfer of the final modified cloning vector into the heterologous host, Transformation via electroporation and conjugation are the typical
the BGC can be expressed so the produced NP can be studied further [16, horizontal gene transfer methods used to insert the BGC-containing
24–26] (Fig. 1). For the best outcomes of heterologous expression, the plasmid into the host [27]. The first case of successful heterologous
BGC’s boundaries need to be accurately defined to include all genes expression of a natural product in Streptomyces salinispora used the
required for NP biosynthesis. The heterologous host also needs to have TAR-mediate pCAP01 vector to capture the whole enterocin BGC and
all necessary machinery for expression of the BGC products. Bio­ conjugate the plasmid into S. lividans TK23 and S. coelicolor M1146[16,
informatic programs previously mentioned, antiSMASH [18,19], and 132]. Synthetic biology is used in heterologous gene expression methods
SMURF [17] can be used to identify the BGCs and their surrounding to help activate and better characterize the production of NPs through
genomic content. Typically, heterologous gene expression has been used BGC activation. These heterologous methods, while not always
to link the natural product chemotype with its genotype [25]. high-throughput, may represent the best possibility for discovering
The first step in heterologous gene expression is identifying target novel antibiotic BGCs because of the ability to study gene clusters from
BGCs and cloning them into DNA vectors, which can be done through organisms without the need to culture them. Most of the bacterial world
both targeted and untargeted methods. Targeted methods for identifying is still unculturable to us and may be hiding many genomic secrets in
BGCs include genomic sequencing followed by DNA synthesis or direct plain sight.
cloning [135–138]. Untargeted methods for identifying BGCs include
constructing random genomic libraries via mutagenesis followed by 5. Final thoughts
select clone recovery [138,139]. CONKAT-seq, a targeted sequence
workflow using library-wide co-occurrence analysis, has been used to Synthetic biology can be applied to existing genome editing tools by
identify, locate, and assess multiple BGCs concurrently [138]. This using created and engineered components for controlling and manipu­
software uses amplicon barcodes to identify the domain sequence (i.e., lating gene expression. Synthetic biology can also be used to manipulate
NRPS, PKS, TPS, and DMATS) and uses this information to locate whole gene circuits or construct metabolic pathways in non-native organisms.
BGCs within the original genome library [138]. There are abundant techniques that can be modified and tailored for
After the identification of BGCs, and their boundaries, they need to cryptic BGC activation. These methods are being developed and applied
be mobilized to prepare them for integration into the host. Techniques within the ground-breaking, rapidly expanding field of synthetic
involved in mobilization of BGCs commonly include Gibson assembly biology. Genomic mutagenesis strategies are being applied in synthetic
[140] or insertion of BGCs into a cloning vector (cosmid [62,141], fos­ biology to stimulate genetic variations within a bacterium’s genome to
mid [16,75,142], bacterial artificial chromosome (BAC) [143,144], activate BGCs and produce their encoded natural product. Endogenous
P1-derived artificial chromosome (PAC) libraries [145,146], yeast arti­ and heterologous expression systems are being used in synthetic biology
ficial chromosome (YAC) [147,148]), or CRISPR/cas9 associated tar­ to activate cryptic BGCs using cloning and genetic engineering tech­
geting of chromosome segments (CATCH) [149]). Gibson assembly joins niques. While each technique has its advantages and disadvantages for
PCR-amplified genes directly from genomic DNA [150]. Cosmid vectors activating cryptic BGCs, the drawbacks of mutagenesis strategies,
contain specific Lambda phage cos regions for packaging DNA into endogenous expression, and heterologous expression systems may prove
phages for DNA transduction [141]. Fosmid vectors are similar to cos­ to be challenging for synthetic biologists to overcome. Mutagenesis
mid vectors, but instead contain the F-factor genes necessary for strategies may be successful in producing novel products if the BGCs
conjugation as a DNA transfer method instead of transduction [142]. have been located and defined. The biggest obstacle involves identifying
CRISPR/Cas9-assisted targeting of chromosome segments (CATCH) can the BGC and being certain it’s responsible for producing a novel product
excise ~150 kb sized BGCs and ligate them into cloning vectors [25]. [138]. Endogenous gene expression systems may prove challenging for
Mobilizing pieces of DNA that contain full BGCs can require even larger synthetic biologists to produce enough natural products to be studied
cloning vectors to be constructed and may have to rely on BACs or YACs efficiently [119]. With Heterologous expression techniques, synthetic
depending on the size or application [148,151]. One method of large biologists may find producing novel products time-consuming and
DNA segment mobilization, transformation-associated recombination difficult due to a large amount of manipulation in different locations of
(TAR), utilizes homologous recombination in yeast to combine small the BGC to ensure the mobilization vector is complementary to the
TAR vectors with target BGCs [152]. The TAR approach is useful in heterologous host [27]. Efforts should be made to improve the ability to
mobilizing silent BGCs in unculturable bacteria [16,153]. The ends of locate and identify BGCs and make techniques more high throughput.

7
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

The goal of activating these cryptic BGCs is to discover, characterize, [17] Khaldi N, et al. SMURF: genomic mapping of fungal secondary metabolite
clusters. Fungal Genet Biol 2010;47(9):736–41. https://doi.org/10.1016/j.
and use novel natural products as antibiotics. If these techniques can be
fgb.2010.06.003. Sep.
effectively adapted and harnessed to focus on antibiotic BGC discovery [18] Blin K, et al. antiSMASH 7.0: new and improved predictions for detection,
and characterization, we may be able to enter a new generation of regulation, chemical structures and visualisation. Nucleic Acids Res 2023.
antibiotic production that can turn back the clock and prevent the https://doi.org/10.1093/nar/gkad344. May.
[19] Medema MH, et al. antiSMASH: rapid identification, annotation and analysis of
looming danger of the ever-expanding list of MDR and XDR bacterial secondary metabolite biosynthesis gene clusters in bacterial and fungal genome
pathogens. sequences. Nucleic Acids Res 2011;39(suppl_2):W339–46. https://doi.org/
10.1093/nar/gkr466. Jul.
[20] Zhao N, et al. Synthetic biology-inspired cell engineering in diagnosis, treatment,
Funding and drug development. Signal Transd Target Therapy 2023;8(1). https://doi.org/
10.1038/s41392-023-01375-x. Springer Nature, Dec. 01.
[21] McCarty NS, Ledesma-Amaro R. Synthetic biology tools to engineer microbial
GDC is supported by a Provost Office Research Assistantship within communities for biotechnology. Trends Biotechnol 2019;37(2):181–97. https://
the Department of Biology & Environmental Science at the University of doi.org/10.1016/j.tibtech.2018.11.002. Feb.
New Haven, West Haven, CT, USA. This funding source had no input in [22] Kim E, Moore BS, Yoon YJ. Reinvigorating natural product combinatorial
biosynthesis with synthetic biology. Nat Chem Biol 2015;11(9):649–59. https://
this manuscript.
doi.org/10.1038/nchembio.1893. Sep.
[23] Keasling JD. Synthetic biology and the development of tools for metabolic
CRediT authorship contribution statement engineering. Metab Eng 2012;14(3):189–95. https://doi.org/10.1016/j.
ymben.2012.01.004. May.
[24] Alam K, Hao J, Zhang Y, Li A. Synthetic biology-inspired strategies and tools for
Greta D. Cook: Writing – review & editing. Nikolas M. Stasulli: engineering of microbial natural product biosynthetic pathways. Biotechnol Adv
Writing – review & editing. 2021;49:107759. https://doi.org/10.1016/j.biotechadv.2021.107759. Jul.
[25] Kang HS, Kim ES. Recent advances in heterologous expression of natural product
biosynthetic gene clusters in Streptomyces hosts. Curr Opin Biotechnol 2021;69:
118–27. https://doi.org/10.1016/j.copbio.2020.12.016. Elsevier LtdJun. 01.
Declaration of competing interest [26] Lee N, Hwang S, Lee Y, Cho S, Palsson B, Cho BK. Synthetic biology tools for novel
secondary metabolite discovery in streptomyces. J Microbiol Biotechnol 2019;29
(5):667–86. https://doi.org/10.4014/jmb.1904.04015. Korean Society for
The authors declare that they have no known competing financial Microbiology and BiotechnologyMay 28.
interests or personal relationships that could have appeared to influence [27] Covington BC, Xu F, Seyedsayamdost MR. A natural product chemist’s guide to
the work reported in this paper. unlocking silent biosynthetic gene clusters. Annu Rev Biochem 2021;90:763–88.
https://doi.org/10.1146/annurev-biochem-081420-102432. Annual Reviews
IncJun. 20.
References [28] Krishnamurthy M, Moore RT, Rajamani S, Panchal RG. Bacterial genome
engineering and synthetic biology: combating pathogens. BMC Microbiol 2016;16
(1):1–11. https://doi.org/10.1186/s12866-016-0876-3. BioMed CentralNov. 04.
[1] Huemer M, Mairpady Shambat S, Brugger SD, Zinkernagel AS. Antibiotic
[29] Durland J, Ahmadian-Moghadam H. Genetics, mutagenesis. StatPearls Publishing
resistance and persistence—implications for human health and treatment
LLC; 2023.
perspectives. EMBO Rep 2020;21(12). https://doi.org/10.15252/
[30] Yang J, Kim B, Kim GY, Jung GY, Seo SW. Synthetic biology for evolutionary
embr.202051034. Dec.
engineering: from perturbation of genotype to acquisition of desired phenotype.
[2] Fleming A. Classics in infectious diseases: on the antibacterial action of cultures
Biotechnol Biofuels 2019;12(1). https://doi.org/10.1186/s13068-019-1460-5.
of a penicillium, with special reference to their use in the isolation of B.
BioMed Central Ltd., May 09.
influenzae by Alexander Fleming. Reprinted from the Br J Exp Pathol 1980;2(1):
[31] Liang L, et al. CRISPR EnAbled trackable genome engineering for isopropanol
129–39. 10:226-236, 1929.’, Rev Infect Dis.
production in Escherichia coli. Metab Eng 2017;41:1–10. https://doi.org/
[3] Tan S, Tatsumura Y. Alexander fleming (1881–1955): discoverer of penicillin.
10.1016/j.ymben.2017.02.009. May.
Singapore Med J 2015;56(07):366–7. https://doi.org/10.11622/smedj.2015105.
[32] Yu D, Ellis HM, Lee EC, Jenkins NA, Copeland NG, Court DL. An efficient
Jul.
recombination system for chromosome engineering in Escherichia coli. Proc Natl
[4] Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol
Acad Sci 2000;97(11):5978–83. https://doi.org/10.1073/pnas.100127597. May.
Biol Rev 2010;74(3):417–33. https://doi.org/10.1128/MMBR.00016-10. Sep.
[33] Wang HH, Kim H, Cong L, Jeong J, Bang D, Church GM. Genome-scale promoter
[5] C.L. Ventola, ‘The antibiotic resistance crisis: part 1: causes and threats.’, P T, vol.
engineering by coselection MAGE. Nat Methods 2012;9(6):591–3. https://doi.
40, no. 4, pp. 277–83, Apr 2015.
org/10.1038/nmeth.1971. Jun.
[6] Larsson DGJ, Flach CF. Antibiotic resistance in the environment. Nat Rev
[34] Yao J, Zhong J, Fang Y, Geisinger E, Novick RP, Lambowitz AM. Use of targetrons
Microbiol 2022;20(5):257–69. https://doi.org/10.1038/s41579-021-00649-x.
to disrupt essential and nonessential genes in Staphylococcus aureus reveals
Nature ResearchMay 01.
temperature sensitivity of Ll.LtrB group II intron splicing. RNA 2006;12(7):
[7] Reygaert WC. An overview of the antimicrobial resistance mechanisms of
1271–81. https://doi.org/10.1261/rna.68706. Jul.
bacteria. AIMS Microbiol 2018;4(3):482–501. https://doi.org/10.3934/
[35] Wannier TM, et al. Recombineering and MAGE. Nat Rev Methods Primers 2021;1
microbiol.2018.3.482.
(1):7. https://doi.org/10.1038/s43586-020-00006-x. Jan.
[8] Thaker MN, Wright GD. Opportunities for synthetic biology in antibiotics:
[36] Ellis HM, Yu D, DiTizio T, Court DL. High efficiency mutagenesis, repair, and
expanding glycopeptide chemical diversity. ACS Synth Biol 2015;4(3):195–206.
engineering of chromosomal DNA using single-stranded oligonucleotides. Proc
https://doi.org/10.1021/sb300092n. American Chemical SocietyMar. 20.
Natl Acad Sci 2001;98(12):6742–6. https://doi.org/10.1073/pnas.121164898.
[9] Fischbach MA, Walsh CT. Antibiotics for emerging pathogens. Science 2009;325
Jun.
(5944):1089–93. https://doi.org/10.1126/science.1176667.
[37] Costantino N, Court DL. Enhanced levels of λ Red-mediated recombinants in
[10] Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25
mismatch repair mutants. Proc Natl Acad Sci 2003;100(26):15748–53. https://
years. J Nat Prod 2007;70(3):461–77. https://doi.org/10.1021/np068054v. Mar.
doi.org/10.1073/pnas.2434959100. Dec.
[11] Oladipo A, Enwemiwe V, Ejeromedoghene O, Adebayo A, Ogunyemi O, Fu F.
[38] Thomason LC, Sawitzke JA, Li X, Costantino N, Court DL. Recombineering:
Production and functionalities of specialized metabolites from different organic
genetic engineering in bacteria using homologous recombination. Curr Protoc
sources. Metabolites 2022;12(6):534. https://doi.org/10.3390/
Mol Biol 2014;106:1.16.1–1.16.39. https://doi.org/10.1002/0471142727.
metabo12060534. Jun.
mb0116s106. Apr.
[12] Mózsik L, Iacovelli R, Bovenberg RAL, Driessen AJM. Transcriptional activation
[39] Sharan SK, Thomason LC, Kuznetsov SG, Court DL. Recombineering: a
of biosynthetic gene clusters in filamentous fungi. Front Bioeng Biotechnol 2022;
homologous recombination-based method of genetic engineering. Nat Protoc
10. https://doi.org/10.3389/fbioe.2022.901037. Jul.
2009;4(2):206–23. https://doi.org/10.1038/nprot.2008.227. Feb.
[13] Brown AS, Calcott MJ, Owen JG, Ackerley DF. Structural, functional and
[40] Eustáquio AS, Gust B, Luft T, Li SM, Chater KF, Heide L. Clorobiocin biosynthesis
evolutionary perspectives on effective re-engineering of non-ribosomal peptide
in streptomyces. Chem Biol 2003;10(3):279–88. https://doi.org/10.1016/S1074-
synthetase assembly lines. Nat Prod Rep 2018;35(11):1210–28. https://doi.org/
5521(03)00051-6. Mar.
10.1039/C8NP00036K.
[41] Fowler VG, et al. Daptomycin versus standard therapy for bacteremia and
[14] Nivina A, Yuet KP, Hsu J, Khosla C. Evolution and diversity of assembly-line
endocarditis caused by staphylococcus aureus. N Engl J Med 2006;355(7):
polyketide synthases. Chem Rev 2019;119(24):12524–47. https://doi.org/
653–65. https://doi.org/10.1056/NEJMoa053783. Aug.
10.1021/acs.chemrev.9b00525. Dec.
[42] Nguyen KT, et al. Combinatorial biosynthesis of novel antibiotics related to
[15] Rutledge PJ, Challis GL. Discovery of microbial natural products by activation of
daptomycin. Proc Natl Acad Sci 2006;103(46):17462–7. https://doi.org/
silent biosynthetic gene clusters. Nat Rev Microbiol 2015;13(8):509–23. https://
10.1073/pnas.0608589103. Nov.
doi.org/10.1038/nrmicro3496. Aug.
[43] Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E.
[16] Liu Z, Zhao Y, Huang C, Luo Y. Recent advances in silent gene cluster activation in
A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial
streptomyces. Front Bioeng Biotechnol 2021;9. https://doi.org/10.3389/
fbioe.2021.632230. Feb.

8
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

immunity. Science 1979;337(6096):816–21. https://doi.org/10.1126/ [71] Yang C, Huang C, Zhang W, Zhu Y, Zhang C. Heterologous expression of fluostatin
science.1225829. Aug. 2012. gene cluster leads to a bioactive heterodimer. Org Lett 2015;17(21):5324–7.
[44] Nidhi S, et al. Novel CRISPR–cas systems: an updated review of the current https://doi.org/10.1021/acs.orglett.5b02683. Nov.
achievements, applications, and future research perspectives’. Int J Mol Sci 2021; [72] Martínez-Espinosa RM. Heterologous and Homologous Expression of Proteins
22(7):3327. https://doi.org/10.3390/ijms22073327. Mar. from Haloarchaea: denitrification as case of study. Int J Mol Sci 2019;21(1):82.
[45] Zhu Y. Advances in CRISPR/Cas9. Biomed Res Int 2022;2022:1–13. https://doi. https://doi.org/10.3390/ijms21010082. Dec.
org/10.1155/2022/9978571. Sep. [73] B.C. Covington and M.R. Seyedsayamdost, ‘Guidelines for metabolomics-guided
[46] Gardiner J, Ghoshal B, Wang M, Jacobsen SE. CRISPR–Cas-mediated transposon mutagenesis for microbial natural product discovery’, 2022, pp.
transcriptional control and epi-mutagenesis. Plant Physiol 2022;188(4):1811–24. 305–23. 10.1016/bs.mie.2021.11.020.
https://doi.org/10.1093/plphys/kiac033. Mar. [74] Romano S, Jackson S, Patry S, Dobson A. Extending the “one strain many
[47] Halperin SO, Tou CJ, Wong EB, Modavi C, Schaffer DV, Dueber JE. CRISPR- compounds” (OSMAC) principle to marine microorganisms. Mar Drugs 2018;16
guided DNA polymerases enable diversification of all nucleotides in a tunable (7):244. https://doi.org/10.3390/md16070244. Jul.
window. Nature 2018;560(7717):248–52. https://doi.org/10.1038/s41586-018- [75] Nah HJ, Pyeon HR, Kang SH, Choi SS, Kim ES. Cloning and heterologous
0384-8. Aug. expression of a large-sized natural product biosynthetic gene cluster in
[48] Sadanand S. EvolvR-ing to targeted mutagenesis. Nat Biotechnol 2018;36(9). streptomyces species. Front Microbiol 2017;8. https://doi.org/10.3389/
https://doi.org/10.1038/nbt.4247. 819–819Oct. fmicb.2017.00394. Mar.
[49] Garst AD, et al. Genome-wide mapping of mutations at single-nucleotide [76] Mao D, Okada BK, Wu Y, Xu F, Seyedsayamdost MR. Recent advances in
resolution for protein, metabolic and genome engineering. Nat Biotechnol 2017; activating silent biosynthetic gene clusters in bacteria. Curr Opin Microbiol 2018;
35(1):48–55. https://doi.org/10.1038/nbt.3718. Jan. 45:156–63. https://doi.org/10.1016/j.mib.2018.05.001. Oct.
[50] Qi LS, et al. Repurposing CRISPR as an RNA-guided platform for sequence- [77] Ju KS, Zhang X, Elliot MA. New kid on the block: LmBu expands the repertoire of
specific control of gene expression. Cell 2013;152(5):1173–83. https://doi.org/ specialized metabolic regulators in streptomyces. J Bacteriol 2018;200(2).
10.1016/j.cell.2013.02.022. Feb. https://doi.org/10.1128/JB.00559-17. Jan.
[51] McGlincy NJ, Meacham ZA, Reynaud KK, Muller R, Baum R, Ingolia NT. [78] Du C, van Wezel GP. Mining for microbial gems: integrating proteomics in the
A genome-scale CRISPR interference guide library enables comprehensive postgenomic natural product discovery pipeline. Proteomics 2018;18(18):
phenotypic profiling in yeast. BMC Genom 2021;22(1):205. https://doi.org/ 1700332. https://doi.org/10.1002/pmic.201700332. Sep.
10.1186/s12864-021-07518-0. Dec. [79] Liao Y, Wei ZH, Bai L, Deng Z, Zhong JJ. Effect of fermentation temperature on
[52] Deaner M, Alper HS. Systematic testing of enzyme perturbation sensitivities via validamycin A production by Streptomyces hygroscopicus 5008. J Biotechnol
graded dCas9 modulation in Saccharomyces cerevisiae. Metab Eng 2017;40: 2009;142(3–4):271–4. https://doi.org/10.1016/j.jbiotec.2009.04.015. Jul.
14–22. https://doi.org/10.1016/j.ymben.2017.01.012. Mar. [80] Schwarz J, Hubmann G, Rosenthal K, Lütz S. Triaging of culture conditions for
[53] Li W, Huang C, Chen J. The application of CRISPR /Cas mediated gene editing in enhanced secondary metabolite diversity from different bacteria. Biomolecules
synthetic biology: challenges and optimizations. Front Bioeng Biotechnol 2022; 2021;11(2):193. https://doi.org/10.3390/biom11020193. Jan.
10. https://doi.org/10.3389/fbioe.2022.890155. Aug. [81] Wei H, Lin Z, Li D, Gu Q, Zhu T. ‘[OSMAC (one strain many compounds) approach
[54] Liu HJ, et al. High-throughput CRISPR/Cas9 mutagenesis streamlines trait gene in the research of microbial metabolites-a review. Wei Sheng Wu Xue Bao 2010;
identification in maize. Plant Cell 2020;32(5):1397–413. https://doi.org/ 50(6):701–9. Jun.
10.1105/tpc.19.00934. May. [82] Sproule A, et al. Terrosamycins A and B, Bioactive Polyether Ionophores from
[55] Wang HH, et al. Programming cells by multiplex genome engineering and Streptomyces sp. RKND004 from prince edward island sediment. Mar Drugs
accelerated evolution. Nature 2009;460(7257):894–8. https://doi.org/10.1038/ 2019;17(6):347. https://doi.org/10.3390/md17060347. Jun.
nature08187. Aug. [83] Pettit RK. Mixed fermentation for natural product drug discovery. Appl Microbiol
[56] Ronda C, Pedersen LE, Sommer MOA, Nielsen AT. CRMAGE: CRISPR optimized Biotechnol 2009;83(1):19–25. https://doi.org/10.1007/s00253-009-1916-9.
MAGE recombineering. Sci Rep 2016;6(1):19452. https://doi.org/10.1038/ May.
srep19452. Jan. [84] Okada BK, Seyedsayamdost MR. Antibiotic dialogues: induction of silent
[57] Nyerges Á, et al. A highly precise and portable genome engineering method biosynthetic gene clusters by exogenous small molecules. FEMS Microbiol Rev
allows comparison of mutational effects across bacterial species. Proc Natl Acad 2017;41(1):19–33. https://doi.org/10.1093/femsre/fuw035. Jan.
Sci 2016;113(9):2502–7. https://doi.org/10.1073/pnas.1520040113. Mar. [85] M.R. Seyedsayamdost, M.F. Traxler, J. Clardy, and R. Kolter, ‘Old meets new:
[58] Tan F, et al. Using global transcription machinery engineering (gTME) to improve using interspecies interactions to detect secondary metabolite production in
ethanol tolerance of Zymomonas mobilis. Microb Cell Fact 2016;15(1):4. https:// actinomycetes’, 2012, pp. 89–109. 10.1016/B978-0-12-404634-4.00005-X.
doi.org/10.1186/s12934-015-0398-y. Dec. [86] Shank EA, Kolter R. New developments in microbial interspecies signaling. Curr
[59] Singh V, Braddick D. Recent advances and versatility of MAGE towards industrial Opin Microbiol 2009;12(2):205–14. https://doi.org/10.1016/j.mib.2009.01.003.
applications. Syst Synth Biol 2015;9(S1):1–9. https://doi.org/10.1007/s11693- Apr.
015-9184-8. Dec. [87] Lyng M, Kovács ÁT. Frenemies of the soil: bacillus and Pseudomonas interspecies
[60] Wang X, et al. Improved dsDNA recombineering enables versatile multiplex interactions. Trends Microbiol 2023. https://doi.org/10.1016/j.tim.2023.02.003.
genome engineering of kilobase-scale sequences in diverse bacteria. Nucleic Acids Mar.
Res 2022;50(3). https://doi.org/10.1093/nar/gkab1076. e15–e15Feb. [88] Straight PD, Willey JM, Kolter R. Interactions between streptomyces coelicolor
[61] Grassi G, Marini JC. Ribozymes: structure, function and potential therapy for and bacillus subtilis : role of surfactants in raising aerial structures. J Bacteriol
dominant genetic disorders. Ann Med 1996;28(6):499–510. https://doi.org/ 2006;188(13):4918–25. https://doi.org/10.1128/JB.00162-06. Jul.
10.3109/07853899608999114. Jan. [89] UEDA K, et al. Wide distribution of interspecific stimulatory events on antibiotic
[62] Gorniaková D, et al. Activation of a cryptic manumycin-type biosynthetic gene production and sporulation among streptomyces species. J Antibiot 2000;53(9):
cluster of saccharothrix espanaensis DSM44229 by series of genetic 979–82. https://doi.org/10.7164/antibiotics.53.979 (Tokyo).
manipulations. Microorganisms 2021;9(3):559. https://doi.org/10.3390/ [90] Lee N, et al. Iron competition triggers antibiotic biosynthesis in Streptomyces
microorganisms9030559. Mar. coelicolor during coculture with Myxococcus xanthus. ISME J 2020;14(5):
[63] Velázquez E, de Lorenzo V, Al-Ramahi Y. Recombination-independent genome 1111–24. https://doi.org/10.1038/s41396-020-0594-6. May.
editing through CRISPR/Cas9-enhanced targetron delivery. ACS Synth Biol 2019; [91] Coisne S, Béchet M, Blondeau R. Actinorhodin production by Streptomyces
8(9):2186–93. https://doi.org/10.1021/acssynbio.9b00293. Sep. coelicolor A3(2) in iron-restricted media. Lett Appl Microbiol 1999;28(3):
[64] Kuehne SA, Minton NP. ClosTron-mediated engineering of clostridium. 199–202. https://doi.org/10.1046/j.1365-2672.1999.00509.x. Mar.
Bioengineered 2012;3(4):247–54. https://doi.org/10.4161/bioe.21004. [92] Ahmed Y, Rebets Y, Tokovenko B, Brötz E, Luzhetskyy A. Identification of
[65] Velázquez E, Al-Ramahi Y, de Lorenzo V. CRISPR/Cas9-enhanced targetron butenolide regulatory system controlling secondary metabolism in Streptomyces
insertion for delivery of heterologous sequences into the genome of gram- albus J1074. Sci Rep 2017;7(1):9784. https://doi.org/10.1038/s41598-017-
negative bacteria. Curr Protoc 2022;2(9). https://doi.org/10.1002/cpz1.532. 10316-y. Aug.
Sep. [93] Xu Z, et al. Large-scale transposition mutagenesis of streptomyces coelicolor
[66] Velázquez E, Al-Ramahi Y, Tellechea-Luzardo J, Krasnogor N, de Lorenzo V. identifies hundreds of genes influencing antibiotic biosynthesis. Appl Environ
Targetron-assisted delivery of exogenous DNA sequences into pseudomonas Microbiol 2017;83(6). https://doi.org/10.1128/AEM.02889-16. Mar.
putida through CRISPR-aided counterselection. ACS Synth Biol 2021;10(10): [94] Guo F, et al. Targeted activation of silent natural product biosynthesis pathways
2552–65. https://doi.org/10.1021/acssynbio.1c00199. Oct. by reporter-guided mutant selection. Metab Eng 2015;28:134–42. https://doi.
[67] Zhao X, et al. ‘[A temperature-inducible Targetron system for efficient gene org/10.1016/j.ymben.2014.12.006. Mar.
inactivation in Escherichia coli]. Sheng Wu Gong Cheng Xue Bao 2020;36(8): [95] Mao D, Yoshimura A, Wang R, Seyedsayamdost MR. Reporter-guided transposon
1659–71. https://doi.org/10.13345/j.cjb.190547. Aug. mutant selection for activation of silent gene clusters in burkholderia
[68] Alper H, Stephanopoulos G. Global transcription machinery engineering: a new thailandensis. ChemBioChem 2020;21(13):1826–31. https://doi.org/10.1002/
approach for improving cellular phenotype. Metab Eng 2007;9(3):258–67. cbic.201900748. Jul.
https://doi.org/10.1016/j.ymben.2006.12.002. May. [96] Xie C, Deng JJ, Wang HX. Identification of AstG1, A LAL family regulator that
[69] A.M. Lanza and H.S. Alper, ‘Global strain engineering by mutant transcription positively controls ansatrienins production in streptomyces sp. XZQH13’. Curr
factors’, 2011, pp. 253–74. 10.1007/978-1-61779-197-0_15. Microbiol 2015;70(6):859–64. https://doi.org/10.1007/s00284-015-0798-6.
[70] Alper H, Moxley J, Nevoigt E, Fink GR, Stephanopoulos G. Engineering yeast Jun.
transcription machinery for improved ethanol tolerance and production. Science [97] Cai X, et al. Manipulation of regulatory genes reveals complexity and fidelity in
1979;314(5805):1565–8. https://doi.org/10.1126/science.1131969. Dec. 2006. hormaomycin biosynthesis. Chem Biol 2013;20(6):839–46. https://doi.org/
10.1016/j.chembiol.2013.04.018. Jun.

9
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

[98] Daniel-Ivad M, et al. An engineered allele of afsQ1 facilitates the discovery and [124] Moon K, Xu F, Zhang C, Seyedsayamdost MR. Bioactivity-HiTES unveils cryptic
investigation of cryptic natural products. ACS Chem Biol 2017;12(3):628–34. antibiotics encoded in actinomycete bacteria. ACS Chem Biol 2019;14(4):767–74.
https://doi.org/10.1021/acschembio.6b01002. Mar. https://doi.org/10.1021/acschembio.9b00049. Apr.
[99] Krause J, Handayani I, Blin K, Kulik A, Mast Y. Disclosing the potential of the [125] Xu F, et al. A genetics-free method for high-throughput discovery of cryptic
SARP-type regulator PapR2 for the activation of antibiotic gene clusters in microbial metabolites. Nat Chem Biol 2019;15(2):161–8. https://doi.org/
streptomycetes. Front Microbiol 2020;11. https://doi.org/10.3389/ 10.1038/s41589-018-0193-2. Feb.
fmicb.2020.00225. Feb. [126] Reynolds KA, et al. Isolation and structure elucidation of lipopeptide antibiotic
[100] Pan Y, Wang L, He X, Tian Y, Liu G, Tan H. SabR enhances nikkomycin production taromycin B from the activated taromycin biosynthetic gene cluster. J Antibiot
via regulating the transcriptional level of sanG, a pathway-specific regulatory 2018;71(2):333–8. https://doi.org/10.1038/ja.2017.146 (Tokyo)Feb.
gene in Streptomyces ansochromogenes. BMC Microbiol 2011;11(1):164. https:// [127] Kepplinger B, et al. Mode of action and heterologous expression of the natural
doi.org/10.1186/1471-2180-11-164. Dec. product antibiotic vancoresmycin. ACS Chem Biol 2018;13(1):207–14. https://
[101] Laureti L, et al. Identification of a bioactive 51-membered macrolide complex by doi.org/10.1021/acschembio.7b00733. Jan.
activation of a silent polyketide synthase in Streptomyces ambofaciens. Proc Natl [128] Inahashi Y, et al. Identification and heterologous expression of the actinoallolide
Acad Sci 2011;108(15):6258–63. https://doi.org/10.1073/pnas.1019077108. biosynthetic gene cluster. J Antibiot 2018;71(8):749–52. https://doi.org/
Apr. 10.1038/s41429-018-0057-8 (Tokyo)Aug.
[102] Thanapipatsiri A, et al. Discovery of Unusual Biaryl Polyketides by Activation of a [129] Huang F, Lei J, Dou X, Luan X, Zhong J. Nighttime medium-scale traveling
Silent Streptomyces venezuelae Biosynthetic Gene Cluster. ChemBioChem 2016; ionospheric disturbances from airglow imager and global navigation satellite
17(22):2189–98. https://doi.org/10.1002/cbic.201600396. Nov. systems observations. Geophys Res Lett 2018;45(1):31–8. https://doi.org/
[103] Wang B, Guo F, Dong SH, Zhao H. Activation of silent biosynthetic gene clusters 10.1002/2017GL076408. Jan.
using transcription factor decoys. Nat Chem Biol 2019;15(2):111–4. https://doi. [130] Saugar I, Molloy B, Sanz E, Sánchez MB, Fernández-Lobato M, Jiménez A.
org/10.1038/s41589-018-0187-0. Feb. Characterization of the biosynthetic gene cluster (ata) for the A201A
[104] Mukherji R, Zhang S, Chowdhury S, Stallforth P. Chimeric LuxR transcription aminonucleoside antibiotic from Saccharothrix mutabilis subsp. capreolus.
factors rewire natural product regulation. Angew Chem Int Ed 2020;59(15): J Antibiot 2017;70(4):404–13. https://doi.org/10.1038/ja.2016.123 (Tokyo)Apr.
6192–5. https://doi.org/10.1002/anie.201914449. Apr. [131] Leipoldt F, et al. Warhead biosynthesis and the origin of structural diversity in
[105] Vogt K, Bhabhra R, Rhodes JC, Askew DS. Doxycycline-regulated gene expression hydroxamate metalloproteinase inhibitors. Nat Commun 2017;8(1):1965.
in the opportunistic fungal pathogen Aspergillus fumigatus. BMC Microbiol 2005; https://doi.org/10.1038/s41467-017-01975-6. Dec.
5(1):1. https://doi.org/10.1186/1471-2180-5-1. [132] Bonet B, Teufel R, Crüsemann M, Ziemert N, Moore BS. Direct capture and
[106] Zarnack K, et al. Tetracycline-regulated gene expression in the pathogen Ustilago heterologous expression of salinispora natural product genes for the biosynthesis
maydis. Fungal Genet Biol 2006;43(11):727–38. https://doi.org/10.1016/j. of enterocin. J Nat Prod 2015;78(3):539–42. https://doi.org/10.1021/
fgb.2006.05.006. Nov. np500664q. Mar.
[107] Derntl C, Mach RL, Mach-Aigner AR. Fusion transcription factors for strong, [133] Gomez-Escribano JP, Bibb MJ. Engineering Streptomyces coelicolor for
constitutive expression of cellulases and xylanases in Trichoderma reesei. heterologous expression of secondary metabolite gene clusters. Microb Biotechnol
Biotechnol Biofuels 2019;12(1):231. https://doi.org/10.1186/s13068-019-1575- 2011;4(2):207–15. https://doi.org/10.1111/j.1751-7915.2010.00219.x. Mar.
8. Dec. [134] Schloss PD, Handelsman J. Metagenomics for studying unculturable
[108] Olano C, et al. Activation and identification of five clusters for secondary microorganisms: cutting the Gordian knot. Genome Biol 2005;6(8):229. https://
metabolites in Streptomyces albus J1074. Microb Biotechnol 2014;7(3):242–56. doi.org/10.1186/gb-2005-6-8-229.
https://doi.org/10.1111/1751-7915.12116. May. [135] Enghiad B, et al. Cas12a-assisted precise targeted cloning using in vivo Cre-lox
[109] Wang YH, Wei KY, Smolke CD. Synthetic biology: advancing the design of diverse recombination. Nat Commun 2021;12(1):1171. https://doi.org/10.1038/s41467-
genetic systems. Annu Rev Chem Biomol Eng 2013;4(1):69–102. https://doi.org/ 021-21275-4. Feb.
10.1146/annurev-chembioeng-061312-103351. Jun. [136] Yamanaka K, et al. Direct cloning and refactoring of a silent lipopeptide
[110] Shima J, Hesketh A, Okamoto S, Kawamoto S, Ochi K. Induction of actinorhodin biosynthetic gene cluster yields the antibiotic taromycin A. Proc Natl Acad Sci
production by rpsL (encoding ribosomal protein S12) mutations that confer 2014;111(5):1957–62. https://doi.org/10.1073/pnas.1319584111. Feb.
streptomycin resistance in Streptomyces lividans and Streptomyces coelicolor A3 [137] Harvey CJB, et al. HEx: a heterologous expression platform for the discovery of
(2). J Bacteriol 1996;178(24):7276–84. https://doi.org/10.1128/jb.178.24.7276- fungal natural products. Sci Adv 2018;4(4). https://doi.org/10.1126/sciadv.
7284.1996. Dec. aar5459. Apr.
[111] Imai Y, Sato S, Tanaka Y, Ochi K, Hosaka T. Lincomycin at subinhibitory [138] Libis V, et al. Multiplexed mobilization and expression of biosynthetic gene
concentrations potentiates secondary metabolite production by streptomyces spp. clusters. Nat Commun 2022;13(1):5256. https://doi.org/10.1038/s41467-022-
Appl Environ Microbiol 2015;81(11):3869–79. https://doi.org/10.1128/ 32858-0. Sep.
AEM.04214-14. Jun. [139] Hover BM, et al. Culture-independent discovery of the malacidins as calcium-
[112] Wang G, Hosaka T, Ochi K. Dramatic activation of antibiotic production in dependent antibiotics with activity against multidrug-resistant Gram-positive
streptomyces coelicolor by cumulative drug resistance mutations. Appl Environ pathogens. Nat Microbiol 2018;3(4):415–22. https://doi.org/10.1038/s41564-
Microbiol 2008;74(9):2834–40. https://doi.org/10.1128/AEM.02800-07. May. 018-0110-1. Feb.
[113] Hosaka T, et al. Antibacterial discovery in actinomycetes strains with mutations [140] Wan J, Ma N, Yuan H. Recent advances in the direct cloning of large natural
in RNA polymerase or ribosomal protein S12. Nat Biotechnol 2009;27(5):462–4. product biosynthetic gene clusters. Eng Microbiol 2023;3(3):100085. https://doi.
https://doi.org/10.1038/nbt.1538. May. org/10.1016/j.engmic.2023.100085. Sep.
[114] Ahn SK, Cuthbertson L, Nodwell JR. Genome context as a predictive tool for [141] Collins J, Hohn B. Cosmids: a type of plasmid gene-cloning vector that is
identifying regulatory targets of the TetR family transcriptional regulators. PLoS packageable in vitro in bacteriophage lamda heads. 1978. Biotechnology 1992;
One 2012;7(11):e50562. https://doi.org/10.1371/journal.pone.0050562. Nov. 24:193–7.
[115] Cuthbertson L, Nodwell JR. The TetR family of regulators. Microbiol Mol Biol Rev [142] Vergin KL, Urbach E, Stein JL, DeLong EF, Lanoil BD, Giovannoni SJ. Screening of
2013;77(3):440–75. https://doi.org/10.1128/MMBR.00018-13. Sep. a fosmid library of marine environmental genomic DNA fragments reveals four
[116] Ohnishi Y, Kameyama S, Onaka H, Horinouchi S. The A-factor regulatory cascade clones related to members of the order planctomycetales. Appl Environ Microbiol
leading to streptomycin biosynthesis in Streptomyces griseus : identification of a 1998;64(8):3075–8. https://doi.org/10.1128/AEM.64.8.3075-3078.1998. Aug.
target gene of the A-factor receptor. Mol Microbiol 1999;34(1):102–11. https:// [143] Xu M, et al. Functional genome mining for metabolites encoded by large gene
doi.org/10.1046/j.1365-2958.1999.01579.x. Oct. clusters through heterologous expression of a whole-genome bacterial artificial
[117] Horinouchi S. A microbial hormone A-factor as a master switch for morphological chromosome library in streptomyces spp. Appl Environ Microbiol 2016;82(19):
differentiation and secondary metabolism in streptomyces griseus. Front Biosci 5795–805. https://doi.org/10.1128/AEM.01383-16. Oct.
2002;7(4):A897. https://doi.org/10.2741/A897. [144] Tao Q, Zhao H, Qiu L, Hong G. Construction of a full bacterial artificial
[118] Sidda JD, et al. Discovery of a family of γ-aminobutyrate ureas via rational chromosome (BAC) library of Oryza sativa genome. Cell Res 1994;4(2):127–33.
derepression of a silent bacterial gene cluster. Chem Sci 2014;5(1):86–9. https:// https://doi.org/10.1038/cr.1994.13. Dec.
doi.org/10.1039/C3SC52536H. [145] Jones AC, Gust B, Kulik A, Heide L, Buttner MJ, Bibb MJ. Phage P1-derived
[119] Seyedsayamdost MR. High-throughput platform for the discovery of elicitors of artificial chromosomes facilitate heterologous expression of the FK506 gene
silent bacterial gene clusters. Proc Natl Acad Sci 2014;111(20):7266–71. https:// cluster. PLoS One 2013;8(7):e69319. https://doi.org/10.1371/journal.
doi.org/10.1073/pnas.1400019111. May. pone.0069319. Jul.
[120] Okada BK, Wu Y, Mao D, Bushin LB, Seyedsayamdost MR. Mapping the [146] Coren JS, Sternberg N. Construction of a PAC vector system for the propagation of
Trimethoprim-Induced Secondary Metabolome of Burkholderia thailandensis. genomic DNA in bacterial and mammalian cells and subsequent generation of
ACS Chem Biol 2016;11(8):2124–30. https://doi.org/10.1021/ nested deletions in individual library members. Gene 2001;264(1):11–8. https://
acschembio.6b00447. Aug. doi.org/10.1016/S0378-1119(01)00330-4. Feb.
[121] Li A, et al. Multi-omic analyses provide links between low-dose antibiotic [147] Murray AW, Szostak JW. Construction of artificial chromosomes in yeast. Nature
treatment and induction of secondary metabolism in burkholderia thailandensis. 1983;305(5931):189–93. https://doi.org/10.1038/305189a0.
MBio 2020;11(1). https://doi.org/10.1128/mBio.03210-19. Feb. [148] Wang W, Zheng G, Lu Y. Recent advances in strategies for the cloning of natural
[122] Han EJ, Lee SR, Hoshino S, Seyedsayamdost MR. Targeted discovery of cryptic product biosynthetic gene clusters. Front Bioeng Biotechnol 2021;9. https://doi.
metabolites with antiproliferative activity. ACS Chem Biol 2022;17(11):3121–30. org/10.3389/fbioe.2021.692797. Jul.
https://doi.org/10.1021/acschembio.2c00588. Nov. [149] Jiang W, Zhao X, Gabrieli T, Lou C, Ebenstein Y, Zhu TF. Cas9-assisted targeting
[123] Moon K, Xu F, Seyedsayamdost MR. Cebulantin, a cryptic lanthipeptide antibiotic of chromosome segments CATCH enables one-step targeted cloning of large gene
uncovered using bioactivity-coupled HiTES. Angew Chem Int Ed 2019;58(18): clusters. Nat Commun 2015;6(1):8101. https://doi.org/10.1038/ncomms9101.
5973–7. https://doi.org/10.1002/anie.201901342. Apr. Sep.

10
G.D. Cook and N.M. Stasulli SLAS Technology 29 (2024) 100120

[150] Gibson DG, Young L, Chuang RY, Venter JC, Hutchison CA, Smith HO. Enzymatic [152] Kouprina N, Larionov V. TAR Cloning: perspectives for Functional Genomics,
assembly of DNA molecules up to several hundred kilobases. Nat Methods 2009;6 Biomedicine, and Biotechnology’. Mol Ther Methods Clin Dev 2019;14:16–26.
(5):343–5. https://doi.org/10.1038/nmeth.1318. May. https://doi.org/10.1016/j.omtm.2019.05.006. Sep.
[151] Park HS, Park JH, Kim HJ, Kang SH, Choi SS, Kim ES. BAC cloning and [153] J.J. Zhang, K. Yamanaka, X. Tang, and B.S. Moore, ‘Direct cloning and
heterologous expression of a giant biosynthetic gene cluster encoding antifungal heterologous expression of natural product biosynthetic gene clusters by
neotetrafibricin in streptomyces rubrisoli. Front Bioeng Biotechnol 2022;10. transformation-associated recombination’, 2019, pp. 87–110. 10.1016/bs.
https://doi.org/10.3389/fbioe.2022.964765. Aug. mie.2019.02.026.
[154] Lale R, et al. A universal approach to gene expression engineering. Synth Biol
2022;7(1). https://doi.org/10.1093/synbio/ysac017. Oct.

11

You might also like