New antimicrobials need to be discovered to fight the advance of multidrug-resistant pathogens. A promising approach is the screening for antimicrobial agents naturally produced by living organisms. As an alternative to studying the native producer, it is possible to use genetically tractable microbes as heterologous hosts to aid the discovery process, facilitate product diversification through genetic engineering, and ultimately enable environmentally friendly production. In this mini-review, we summarize the literature from 2017 to 2022 on the application of Escherichia coli and E. coli-based platforms as versatile and powerful systems for the discovery, characterization, and sustainable production of antimicrobials. We highlight recent developments in high-throughput screening methods and genetic engineering approaches that build on the strengths of E. coli as an expression host and that led to the production of antimicrobial compounds. In the last section, we briefly discuss new techniques that have not been applied to discover or engineer antimicrobials yet, but that may be useful for this application in the future.

Antimicrobial-resistant (AMR) infections pose a serious threat to society [1], driving the need to discover and develop new antimicrobial molecules. Promising sources of such antimicrobials are naturally occurring chemicals produced by microbes, plants, and animals—the so-called natural products (NPs) [2]. Traditionally, the utilization of NPs has relied on their isolation from the native producers [3], but this carries several limitations. In brief, the major challenges of working with native producers are: (1) the uncultivability of many organisms under controlled conditions; (2) their slow growth compared with model organisms, and thus the limited availability of biomass for NP extraction; (3) the dependence of NP production on specific environmental stimuli; and (4) the lack of genetic tools to optimize NP production and generate derivatives.

To bypass these limitations, model organisms can be used as heterologous production hosts, often streamlining the discovery and characterization of NPs. Heterologous expression is achieved through cloning and overexpression of the genes encoding the underlying biosynthetic enzymes in a genetically tractable plant, fungus or bacterium. The use of heterologous hosts even allows the exploration of mutant variants or combinations of genes not found in nature [3]. Microbial hosts are often preferred over plants because they have shorter reproductive cycles and can be manipulated at a genetic level more easily. The strengths and weaknesses of each microbial host are extensively covered in other reviews [4,5].

Escherichia coli is one of the earliest and simplest microbial hosts to be used for heterologous expression of single genes and entire biosynthetic pathways (Figure 1a), particularly of bacterial origin [6]. For the expression of eukaryotic genes, E. coli is not as well suited, because it cannot splice introns and introduce post-translational modifications, and often does not support the proper folding of eukaryotic enzymes [7]. Furthermore, it may not provide the required cofactors or partner enzymes (e. g, cytochrome P450 reductase) for enzymatic activity [8]. Many of these challenges can be overcome by carefully choosing the target enzymes, designing the expression constructs, and co-expressing important accessory proteins or pathways [8–10]. On the other hand, E. coli hosts have a high growth rate and the ability to yield large amounts of heterologous proteins. Furthermore, protocols for transformation with exogenous DNA and genetic manipulation in E. coli are well established and fast [11]. These features enable rapid clone–express–test cycles and high-throughput screening—fundamental advantages in the field of NP discovery and production. Indeed, E. coli has been successfully used to produce a wide range of bioactive NPs, including peptides, polyketides (PK), β-lactams, and more (Figure 1b, Table 1).

In the workflow for NP discovery and production in E. coli, candidate biosynthetic genes or pathways are first selected from the genome of antimicrobial producer organisms (Figure 1a, step 1). Often, enzymes within a pathway are encoded by colocalized genes in so-called biosynthetic gene clusters (BGCs), which makes it possible to identify them with dedicated bioinformatic tools [12–15]. Once a target is selected, dedicated E. coli strains or cell-free extracts—optimized for NP production [9,10,16–18]—are used for overexpression (step 2) and characterization of the pathway products (step 3). E. coli is also an ideal host to optimize product yields through modulation of gene expression and enzyme performance by metabolic engineering, directed evolution, and combinatorial biosynthesis (optional step 4). In the following sections, we provide a summary of recent advances in the discovery and heterologous production of antimicrobials based on this workflow.

Reconstitution of biosynthetic machineries in vivo

One of the simplest approaches for NP production in E. coli (Figure 1a, step 2) is to clone and overexpress a minimal set of genes of the target BGC. For example, the production of sublancin—an antimicrobial glycosylated RiPP (ribosomally synthesized and post-translationally modified peptide), or glycocin—was achieved by co-expressing the precursor peptide SunA and its cognate glycosyltransferase SunS [19]. As often done for this class of molecules [20], SunA was engineered with a fused His6 tag for purification and a recognition site for the protease Xa. The latter allows the in vitro release of the bioactive form from the leader peptide after purification, thereby preventing toxicity in the host. This system was efficiently used for structure–activity relationship studies of sublancin to identify amino acids crucial for its antimicrobial bioactivity. Similarly, Choudhary and Rao expressed a mutant library of enterocin 96, a class II bacteriocin, as inactive fusion peptides with a pH-cleavable tag, enabling the rapid identification of a variant with broadened antimicrobial spectrum [21]. Another type of RiPPs, the lanthipeptides, require dedicated enzymes called lanthipeptide synthetases that generate the typical lanthionine bridge through dehydration and cyclization reactions [22]. To obtain the mature NP, the precursor peptide can be either processed in vitro with purified enzymes or co-expressed with the synthetase in vivo. The latter approach was used to discover roseocin, a two-component lanthipeptide with antimicrobial activity against a panel of Gram-positive bacteria (including important pathogens such as MRSA and VRE) [23], and amylopeptins, lanthipeptides with narrow-spectrum antibacterial activity [24].

A powerful approach that can be readily employed with E. coli is the use of fusion proteins to express antimicrobial peptides (AMPs) that would otherwise be toxic to the host. With this strategy, antibacterial cryptopeptides derived from human pepsinogen A3 [25] and apolipoprotein B were produced [26,27]. Cryptopeptides are small peptides that derive from the cleavage of precursor proteins, and often exhibit functions unrelated to that of their parent protein [28,29]. For both AMPs, the peptides were fused to a carrier through an acid-labile Asp-Pro linker, which allowed expression in E. coli as inclusion bodies at high yields. Once purified, the linker allows the chemically controlled release of the bioactive peptides. The peptides produced by Cesaro et al. showed potent antimicrobial and anti-biofilm activity against several pathogens belonging to the concerning ESKAPE group [27,30].

Complementary to the expression of the minimal set of genes, it is also possible to reconstitute entire BGCs. This approach was used to characterize the antimicrobial glycocin pallidocin from a thermophilic bacterium [31]. The entire pal BGC—including precursor peptide, tailoring enzymes, and transporter—was expressed under the control of an inducible promoter in E. coli, which successfully produced the glycosylated peptide in its bioactive form. Bioassays with purified pallidocin showed activity against several thermophilic bacteria and Bacillus sp. [31]. Analogously, the novel lasso peptide microcin Y from Salmonella enterica was discovered by overexpressing the entire MccY BGC in E. coli [32]. This yielded the fully bioactive form of microcin Y, which showed antibacterial activity against several Gram-positive and Gram-negative AMR bacteria, including pathogenic isolates of Shigella and Salmonella [32]. In addition to these numerous examples of AMPs, BGCs of other compound classes have been successfully reconstituted in E. coli, such as nonribosomal peptides [33], diketopiperazines [34] and type I PK [35]. Most recently three type II PK systems were functionally reconstituted and expressed in E. coli [36,37], which may lead to the development of antimicrobial PKs in the future.

Cloning entire BGCs via PCR amplification of the target genes and assembly into suitable vectors is time-consuming and can prove altogether unfeasible, especially with larger BGCs (>10 kb). To streamline the process, alternative methods have been developed. Direct pathway cloning (DiPaC) [38] relies on long-amplicon PCR and HiFi DNA assembly, where small to mid-sized clusters (up to ∼25 kb) are amplified in one or more fragments and assembled in vitro into E. coli expression vectors. Among others, DiPaC was successfully used to reconstitute the BGC of phenazines from S. fonticola, and the BGCs of cyanobacterial nonribosomal peptides anabaenopeptins [38] and hapalosin [39], all of which possess valuable pharmacological properties. For larger BGCs, there are several methods based on in vivo homologous recombination that can be used to capture and mobilize the entire pathway directly from isolated genomic DNA [40–42]. Although they require complex workflows, it is advantageous that these methods do not require amplification steps, since BGCs are captured from genomic libraries. Recombination-based BGC capture has been successfully used to express BGCs up to 106 kb [41,43].

Despite the many successful examples discussed above, capturing and cloning entire BGCs might lead to unsuccessful transcription of the genes because their native regulatory systems may not function in the heterologous host. One possibility to overcome this is pathway refactoring [44], which typically involves the engineering of promoter and other regulatory regions to bypass the native regulatory system and achieve defined expression levels. Tools such as Golden Gate Cloning—a method for the simultaneous one-pot assembly of multiple DNA fragments into one single piece [45]—enable rapid refactoring of large pathways in a few steps [46]. This strategy is widely employed for the assembly of heterologous pathways in E. coli and was successfully used to rapidly discover four novel glycocins through overexpression of their refactored pathways in E. coli, two of which exhibited strong antibacterial activity against Bacillus cereus, a widespread foodborne pathogen [47].

Overall, the current genetic toolbox for E. coli is versatile, however, there remain challenges for its application for antimicrobial discovery. First, heterologous genes and NPs may be toxic to E. coli. Second, expression levels and solubility of the target enzymes might be suboptimal, preventing further characterization efforts. Both problems might be overcome with the use of cell-free expression systems.

E. coli-based cell-free platforms

E. coli-based cell-free protein synthesis (CFPS) was pioneered >60 years ago [48], yet only recently has it truly matured in terms of user-friendliness and reproducibility [49,50]. Apart from its applicability for the synthesis of toxic or unnatural proteins, CFPS offers possibilities for rapid in vitro BGC prototyping and modular pathway construction. For instance, candidate enzymes—even from different organisms—can be expressed using CFPS, and then combined in a mix-and-match fashion [51]. This approach was used to produce the industrially relevant terpenes limonene, pinene and bisabolene [52], as well as phytocannabinoid acids with therapeutic potential [53]. CFPS platforms can also be tailored to produce and characterize specific classes of NPs. For example, a customized cell-free platform expressing the biosynthetic machinery of nisin—one of the most studied lanthibiotics—was employed to rapidly prototype a series of putative lanthipeptides [18]. For that, the sequence of each candidate core peptide was fused to the nisin leader, so that the CFPS system would be able to directly process their bioactive form. In total, four novel lanthibiotics were identified, all of which showed activity against the indicator species Micrococcus luteus, and Enterococcus faecalis. Several other studies produced mature RiPPs with cell-free expression systems [54–58], yet bioactivity assays were not reported.

Since CFPS offers an open reaction system without the limitations of transport across membranes, it can be fully tailored to the specific application by providing cofactors, coenzymes, precursors, and other reaction components of choice.

Metabolic engineering of the E. coli host

Metabolic engineering is a powerful tool to increase product titers and enable further modifications of the core scaffold in a heterologous host. It can be used to optimize the metabolic flux towards the desired product by enzyme engineering, removal of feedback inhibition, increasing precursor supply, blocking of competing pathways, and other techniques (Figure 1a, step 4) [59]. The large number of genome editing tools and techniques available for E. coli [60,61] allow quick and precise manipulation of this organism for biotechnological applications.

In the context of NP biosynthesis in E. coli, metabolic engineering has been used to build chassis strains such as HM0079 and BAP1, that are optimized to support the synthesis of complex NPs by expressing phosphopantetheine transferase, an enzyme crucial for the function of the polyketide synthases and nonribosomal peptide synthetases [9,10]. BAP1 and its derivatives are further optimized to support increased pools of building blocks for polyketide biosynthesis, in particular erythromycin and its analogs [62]. Also, genetically recoded strains that allow the incorporation of non-canonical amino acids by amber stop codon repression into enzymes and RiPPs are intriguing chassis strains for NP biosynthesis [63]. With such a recoded strain Kakkar et al. [64] recently generated unnatural variants of the lantibiotics nisin and lacticin 481, demonstrating its applicability for the direct modification of peptide NPs.

Metabolic engineering is furthermore employed to alleviate specific pathway bottlenecks. In a recent effort, Shomar et al. [65] engineered a strain with elevated pools of precursors and increased β-lactam tolerance through the timed arrest of fatty acid synthesis (FAS). When FAS arrest is induced, E. coli cells stop growing—becoming tolerant to the β-lactams they produce—but remain metabolically active for many generations and are able to yield higher titers. This engineered strain can serve as a powerful platform for carbapenem production and diversification through the co-expression of tailoring enzymes.

Another example of classic metabolic engineering is how Fang et al. [66] increased the production of the glycosylated macrolide antibiotic erythromycin A by 400-fold by knocking out the pathways competing with deoxysugar incorporation. Notably, the large BGC (>50 kB) was cloned into two bacterial artificial chromosomes, each encoding a discrete stage (scaffolding or tailoring) of erythromycin biosynthesis. Such separation enables systematic manipulation of either stage of the biosynthetic process to generate new analogs.

Strategies to unlock new antimicrobial variants

Along with the search for novel antibiotics, a promising approach to combat AMR is the chemical diversification of known antimicrobial scaffolds. Heterologous expression in genetically tractable hosts such as E. coli facilitates the generation of new bioactive derivatives via precursor-directed biosynthesis, rational engineering, and combinatorial biosynthesis.

Precursor-directed biosynthesis exploits the inherent substrate promiscuity of many enzymes in NP pathways. The incorporation of alternative building blocks can thus be achieved by restricting defined nutrients and/or by feeding substrates of choice [67]. With the latter approach, new erythromycin analogs were generated by feeding synthetic building blocks that mimicked native biosynthesis intermediates [68]. Furthermore, expression in a heterologous host may already provide alternative building blocks, resulting in the production of analogs with altered biological activities. For example, the expression of a stand-alone actinobacterial type III polyketide synthase in E. coli resulted in the production of antimicrobial adipostatins A and B and nine additional alkylresorcinols, some with enhanced activity against AMR pathogens [69].

Rational mutagenesis of active site residues alters the substrate scope of an enzyme and enables the production of specific derivatives. For example, it was recently demonstrated that fluorinated building blocks can be incorporated into macrolide antibiotics by using engineered polyketide synthases [70,71]. Similarly, the lyngbyatoxin-producing nonribosomal peptide synthetase was engineered towards the production of pharmaceutically relevant indolactam variants [64,72,73].

Combinatorial biosynthesis enables the generation of NP analogs by genetic manipulation of biosynthetic pathways, for instance by mixing and matching of non-cognate enzymes. Fang et al. [74] diversified the bioactivity of erythromycin analogs by incorporating engineered deoxysugar pathways within their E. coli heterologous system. Three of these analogs were active against an erythromycin-resistant strain of Bacillus subtilis. Similarly, lipidation can be used to modulate the biological activity of peptide-based antimicrobials. Zhao et al. [75] employed a hybrid approach combining E. coli-based expression and processing of disulfide-containing peptides with in vitro lipidation. The resulting non-natural lipo-lanthipeptides exhibited bactericidal activity against several human pathogens through membrane disruption—a mode of action different from the parent peptides.

Combinatorial biosynthesis and random mutagenesis techniques have the potential to become one of the largest sources of new antimicrobial compounds. As they require efficient screening techniques, we will discuss these in detail in the following section.

Screening is a fundamental step in the discovery of new antimicrobials. Once a BGC has been successfully expressed, its products need to be characterized, ideally through a combination of bioactivity testing and analytical chemistry (Figure 1a, step 3). Characterization can be carried out at a single-colony level, where extracts or whole cells from clonal cultures are analyzed for NP production, or in bulk, where large libraries are screened in parallel with high-throughput techniques. In the following section, we will briefly discuss some of the most widely used methods with E. coli-based platforms (Figure 2).

Single-colony screening

Nowadays, HPLC and MS are the standard techniques for the chemical identification and characterization of heterologously expressed NPs. Herein, extracts of E. coli strains (or CFPS reactions) expressing the target BGC are compared with the host background to identify newly produced molecules (Figure 2a). Scale-up of the fermentation and compound purification then allow structural characterization via high-resolution MS (HRMS) and NMR [76,77]. MS and NMR spectra enable the rapid identification of the produced NPs (dereplication) [78]. Complementary to that, agar plate bioassays (Figure 2b) are often used to screen E. coli transformants (or their extracts) for antimicrobial activity—an approach largely unchanged over almost a century [79]. The main disadvantages of this method are the high limits of detection in terms of potency and quantity of the metabolites. Furthermore, plate bioassays do not provide any insight into the metabolic profile and therefore cannot be used for the chemical identification of bioactive compounds. Fortunately, plate bioassays can be easily combined with direct-colony MALDI-ToF MS [80,81] to overcome the latter, by replicating the bacterial colonies on a MALDI target plate (Figure 2b,c). This was elegantly showcased in a recent study on lanthibiotics [81], where researchers could directly link amino acid changes to the bioactivity of the peptide variants in a single experiment. Other MS techniques for direct metabolic profiling of live cultures exist [82,83], but they have not been used in combination with bioactivity assays yet.

Parallel high-throughput screening

One of the most common and powerful high-throughput techniques used in E. coli is phage display (Figure 2d), although it is only suitable for peptide NPs. Herein, a library of peptides—fused to a phage coat protein—is cloned into the host and the newly assembled virions will display the peptides of interest on their surface. By combining this step with systems such as ELISA or streptavidin-biotin–based co-immobilization, large libraries (>106 phages) can be screened against biological targets and enriched for variants with increased binding affinity [84]. This technique was applied to screen and enrich libraries of lanthibiotics using lipid II as binding target [85], as well as a library of artificial lanthipeptides that bind specifically to human urokinase plasminogen activator (uPA), an important factor in tumor progression [86].

Several new approaches treat bacterial death as the target phenotype, leveraging the dissonance between antibiotics discovery and bacterial host systems. Herein, a library of natural or synthetic AMP candidates is expressed in a bacterial host, and the grown cultures are sequenced to identify the least frequent—and thus the most potent—antimicrobial sequences (Figure 2e). E. coli can therefore serve as a chassis for the development of self-inhibition screening platforms for antibiotics against Gram-negative bacteria. Surface localized antimicrobial display (SLAY) was the first technique to apply this approach, screening 800 000 peptides in a single tube [87]. A library encoding random 20-mer peptides was expressed on the E. coli cell surface and the pool of clones was sequenced pre- and post-induction of expression. The initial screening revealed 7968 hits with potential antimicrobial activity. Interestingly, these peptides possessed an unprecedented range of physicochemical properties different from those of naturally occurring AMPs, which are commonly positively charged and hydrophobic. Of the 22 hits chosen for experimental validation, most proved to be active against E. coli, Acinetobacter baumannii and Pseudomonas aeruginosa in the low µM range. Similarly, Koch et al. [88] screened a library of 10 663 naturally occurring peptides with a self-inhibition screen, in which >1000 sequences were active. From these hits, 15 were chemically synthesized and 10 peptides inhibited E. coli at a minimal inhibitory concentration in the upper nM to low µM range. In addition, autotoxicity-based approaches were used to conduct comprehensive mutational studies of the lasso peptide klebsidin [89] and a proline-rich AMP, oncocin [90], providing new insight into their mechanism of action and enabling sequence-activity mapping.

Whole-cell biosensors also emerged as a powerful tool to detect the structure and report the titer of a given NP with high sensitivity and specificity [91]. The basic principle relies on metabolite-sensing proteins, such as allosteric transcription factors, which are activated upon binding of effector molecules and in turn control the expression of an actuator part (e.g. fluorescent reporters, regulatory switches, or selection markers) [92] (Figure 2f). A well-known example of a natural antibiotic biosensor is the erythromycin resistance repressor protein (MphR). This transcription factor controls the transcription of a gene cassette responsible for resistance to macrolide antibiotics and is derepressed by several macrolides with varying degrees of efficiency. Kasey et al. [93] transformed E. coli with libraries of rational and random MphR variants to reveal macrolide biosensors with altered inducer specificities. Contrary to wild-type MphR, some variants were induced by pikromycin and roxithromycin, revealing remarkable plasticity for engineering new sensitivities and selectivity in this transcription factor [93]. Li et al. [94] further shifted the specificity of MphR towards clarithromycin, while Miller et al. [95] improved its sensitivity towards erythromycin. One recent example of how biosensors can be applied to improving antibiotic production, is the work of Johnston et al. [34]. The authors exploited the sensitivity of the Rho-dependent λtR1 terminator to the antibiotic bicyclomycin to apply selective pressure in a phage-assisted continuous evolution experiment (PACE) and generate more efficient variants of the bicyclomycin pathway. Here, higher amounts of bicyclomycin led to increased production of infectious phage particles and thus PACE gradually selected for pathway variants that yielded higher titers of antibiotics. Similarly, d'Oelsnitz et al. [96] developed highly specific and sensitive alkaloid biosensors, which were directly employed to evolve an engineered simplified pathway for tetrahydropapaverine, an important precursor to pharmaceutical compounds. Biosensors may also be useful for the metabolic engineering of producer strains. The FapR-based biosensor engineered by Kalkreuter et al., for instance, detects malonyl-CoA and its C2-substituted derivatives, which serve as extender units in the synthesis of polyketide antibiotics such as merochlorin B, splenocin B and macrolides [97]. The utility of whole-cell biosensors for the selection and engineering of potent specialized antimicrobials is expected to expand as more transcription factors are being characterized.

With the rapid development and increasing accessibility of high-quality genome sequencing and DNA synthesis, the field of NP discovery through genome mining and heterologous expression is moving quickly [98]. New pathway cloning strategies facilitate fast or even automated cloning and refactoring of BGCs for experimental characterization [38,42,99,100]. Several approaches have been used to assemble libraries of natural, engineered, or even combinatorial pathway variants [101–103], yet they remain to be applied for the discovery of antimicrobial NPs. Lastly, for the bioactivity screening step, the field is moving towards medium- and high-throughput approaches that enable parallel screening of larger pathway libraries. Herein, adaptations of the two approaches recently developed by Yang et al. [104] and Sarkar et al. [105] could be particularly interesting for antimicrobial or antiparasitic discovery. In both studies, E. coli was used as the expression host for a set of NP pathway variants, as well as the biological target protein(s). Inhibition of the target protein(s) was coupled to the survival of the host and thus enabled the rapid detection of bioactive NPs [104,105]. Furthermore, whole-cell biosensor approaches reporting antibiotic-induced stress reactions as used with other bacteria [106,107] could be further developed for applications in E. coli.

As already highlighted by the ‘pressure test' commissioned by the U.S. Defense Advanced Research Projects Agency in 2018, E. coli and its cell-free lysates are exquisite platforms for the generation of target compounds, including antimicrobials [108]. This is particularly true for peptides, with many approaches and screening methods available that allow fast product characterization and massive parallelization. Since the expression of large biosynthetic machineries such as nonribosomal peptide synthetases and some types of polyketide synthases remains challenging, the further development of tailored host strains and cell-free platforms might offer solutions to expand the range of NPs produced with E. coli. Thus, it may gain even more importance for antimicrobial discovery and sustainable production in the future.

  • New antimicrobial drugs are needed to combat emerging AMR pathogens. Antimicrobial drug development is deeply rooted in the discovery and exploitation of NPs.

  • E. coli is a powerful and versatile workhorse for the heterologous expression of enzymes and biosynthetic pathways and has been heavily used to discover and produce numerous antimicrobial drugs.

  • Recent advances in pathway cloning and refactoring, and new screening technologies, allow rapid and parallelizable prototyping of biosynthetic pathways in E. coli. This will accelerate the discovery of new leads for drug development and the sustainable biosynthesis of antimicrobial drugs.

The authors declare that there are no competing interests associated with the manuscript.

R.I., N.S. and K.H. collected literature. R.I. and N.S. wrote the manuscript. All authors edited the manuscript, read, and approved the final version.

K.H. is grateful for funding from the European Union's Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 893122 and from the Federation European Biochemical Societies under the Excellence award agreement No 21.07043.

AMPs

antimicrobial peptides

AMR

antimicrobial-resistant

BGCs

biosynthetic gene clusters

CFPS

cell-free protein synthesis

DiPaC

direct pathway cloning

FAS

fatty acid synthesis

NPs

natural products

PACE

phage-assisted continuous evolution experiment

PK

polyketides

1
Murray
,
C.J.
,
Ikuta
,
K.S.
,
Sharara
,
F.
,
Swetschinski
,
L.
,
Robles Aguilar
,
G.
,
Gray
,
A.
et al (
2022
)
Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis
.
Lancet
399
,
629
655
2
Newman
,
D.J.
and
Cragg
,
G.M.
(
2020
)
Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019
.
J. Nat. Prod.
83
,
770
803
3
Atanasov
,
A.G.
,
Zotchev
,
S.B.
,
Dirsch
,
V.M.
,
Orhan
,
I.E.
Banach
,
M.
,
Rollinger
,
J.M.
et al (
2021
)
Natural products in drug discovery: advances and opportunities
.
Nat. Rev. Drug Discov.
20
,
200
216
4
Zhang
,
J.J.
,
Tang
,
X.
and
Moore
,
B.S.
(
2019
)
Genetic platforms for heterologous expression of microbial natural products
.
Nat. Prod. Rep.
36
,
1313
1332
5
Meng
,
X.
,
Fang
,
Y.
,
Ding
,
M.
,
Zhang
,
Y.
,
Jia
,
K.
,
Li
,
Z.
et al (
2022
)
Developing fungal heterologous expression platforms to explore and improve the production of natural products from fungal biodiversity
.
Biotechnol. Adv.
54
,
107866
6
Zhang
,
H.
,
Fang
,
L.
,
Osburne
,
M.S.
and
Pfeifer
,
B.A.
(
2016
)
The continuing development of E. coli as a heterologous host for complex natural product biosynthesis
.
Methods Mol. Biol.
1401
,
121
134
7
Sahdev
,
S.
,
Khattar
,
S.K.
and
Saini
,
K.S.
(
2008
)
Production of active eukaryotic proteins through bacterial expression systems: a review of the existing biotechnology strategies
.
Mol. Cell. Biochem.
307
,
249
264
8
Urlacher
,
V.B.
and
Girhard
,
M.
(
2019
)
Cytochrome P450 monooxygenases in biotechnology and synthetic biology
.
Trends Biotechnol.
37
,
882
897
9
Pfeifer,
,
B.A.
,
Admiraal,
,
S.J.
,
Gramajo,
,
H.
,
Cane,
,
D.E.
and
Khosla,
,
C.
(
2001
)
Biosynthesis of complex polyketides in a metabolically engineered strain of E. coli
.
Science
291
,
1790
292
10
Gruenewald
,
S.
,
Mootz
,
H.D.
,
Stehmeier
,
P.
and
Stachelhaus
,
T.
(
2004
)
In vivo production of artificial nonribosomal peptide products in the heterologous host Escherichia coli
.
Appl. Environ. Microbiol.
70
,
3282
3291
11
Cronan
,
J.E.
(
2014
) Escherichia coli as an experimental organism. In
eLS
,
John Wiley & Sons, Ltd (Ed.)
12
Tietz
,
J.I.
,
Schwalen
,
C.J.
,
Patel
,
P.S.
,
Maxson
,
T.
,
Blair
,
P.M.
,
Tai
,
H.C.
et al (
2017
)
A new genome-mining tool redefines the lasso peptide biosynthetic landscape
.
Nat. Chem. Biol.
13
,
470
478
13
van Heel
,
A.J.
,
de Jong
,
A.
,
Song
,
C.
,
Viel
,
J.H.
,
Kok
,
J.
and
Kuipers
,
O.P.
(
2018
)
BAGEL4: a user-friendly web server to thoroughly mine RiPPs and bacteriocins
.
Nucleic Acids Res.
46
,
W278
W281
14
Merwin
,
N.J.
,
Mousa
,
W.K.
,
Dejong
,
C.A.
,
Skinnider
,
M.A.
,
Cannon
,
M.J.
,
Li
,
H.
et al (
2020
)
DeepRiPP integrates multiomics data to automate discovery of novel ribosomally synthesized natural products
.
Proc. Natl Acad. Sci. U.S.A.
117
,
371
380
15
Blin
,
K.
,
Shaw
,
S.
,
Kloosterman
,
A.M.
,
Charlop-Powers
,
Z.
,
van Wezel
,
G.P.
,
Medema
,
M.H.
et al (
2021
)
antiSMASH 6.0: improving cluster detection and comparison capabilities
.
Nucleic Acids Res.
49
,
W29
W35
16
Li
,
J.
,
Zhang
,
L.
and
Liu
,
W.
(
2018
)
Cell-free synthetic biology for in vitro biosynthesis of pharmaceutical natural products
.
Synth. Syst. Biotechnol.
3
,
83
89
17
Liu
,
T.
,
Mazmouz
,
R.
and
Neilan
,
B.A.
(
2018
)
An in vitro and in vivo study of broad-range phosphopantetheinyl transferases for heterologous expression of cyanobacterial natural products
.
ACS Synth. Biol.
7
,
1143
1151
18
Liu
,
R.
,
Zhang
,
Y.
,
Zhai
,
G.
,
Fu
,
S.
,
Xia
,
Y.
,
Hu
,
B.
et al (
2020
)
A cell-free platform based on nisin biosynthesis for discovering novel lanthipeptides and guiding their overproduction in vivo
.
Adv. Sci.
7
,
2001616
.
19
Biswas
,
S.
,
De Gonzalo C V.
,
G.
,
Repka
,
L.M.
and
van der Donk
,
W.A.
(
2017
)
Structure-activity relationships of the S-linked glycocin sublancin
.
ACS Chem. Biol.
12
,
2965
2969
20
Shi
,
Y.
,
Yang
,
X.
,
Garg
,
N.
and
van der Donk
,
W.A.
(
2011
)
Production of lantipeptides in Escherichia coli
.
J. Am. Chem. Soc.
133
,
2338
2341
21
Choudhary
,
P.
and
Rao
,
A.
(
2021
)
SELECT-GLYCOCIN: a recombinant microbial system for expression and high-throughput screening of glycocins
.
Glycoconj. J.
38
,
233
250
22
Zhang
,
Q.
,
Yu
,
Y.
,
Vélasquez
,
J.E.
and
van der Donk
,
W.A.
(
2012
)
Evolution of lanthipeptide synthetases
.
Proc. Natl Acad. Sci. U.S.A.
109
,
18361
1836
23
Singh
,
M.
,
Chaudhary
,
S.
and
Sareen
,
D.
(
2020
)
Roseocin, a novel two-component lantibiotic from an actinomycete
.
Mol. Microbiol.
113
,
326
337
24
Zhang
,
Y.
,
Hong
,
Z.
,
Zhou
,
L.
,
Zhang
,
Z.
,
Tang
,
T.
,
Guo
,
E.
et al (
2022
)
Biosynthesis of gut-microbiota-derived lantibiotics reveals a subgroup of S8 family proteases for class III leader removal
.
Angew. Chem. Int. Ed.
61
,
e202114414
25
Pane
,
K.
,
Cafaro
,
V.
,
Avitabile
,
A.
,
Torres
,
M.D.T.
,
Vollaro
,
A.
,
de Gregorio
,
E.
et al (
2018
)
Identification of novel cryptic multifunctional antimicrobial peptides from the human stomach enabled by a computational-experimental platform
.
ACS Synth. Biol.
7
,
2105
2115
26
Gaglione
,
R.
,
Dell'Olmo
,
E.
,
Bosso
,
A.
,
Chino
,
M.
,
Pane
,
K.
,
Ascione
,
F.
et al (
2017
)
Novel human bioactive peptides identified in apolipoprotein B: evaluation of their therapeutic potential
.
Biochem. Pharmacol.
130
,
34
50
27
Cesaro
,
A.
,
Torres
,
M.D.T.
,
Gaglione
,
R.
,
Dell'Olmo
,
E.
,
di Girolamo
,
R.
,
Bosso
,
A.
et al (
2022
)
Synthetic antibiotic derived from sequences encrypted in a protein from human plasma
.
ACS Nano
16
,
1880
1895
28
Autelitano
,
D.J.
,
Rajic
,
A.
,
Smith
,
A.I.
,
Berndt
,
M.C.
,
Ilag
,
L.L.
and
Vadas
,
M.
(
2006
)
The cryptome: a subset of the proteome, comprising cryptic peptides with distinct bioactivities
.
Drug. Discov. Today
11
,
306
314
29
Ueki
,
N.
,
Someya
,
K.
,
Matsuo
,
Y.
,
Wakamatsu
,
K.
and
Mukai
,
H.
(
2007
)
Cryptides: functional cryptic peptides hidden in protein structures
.
Biopolymers
88
,
190
198
30
Mulani
,
M.S.
,
Kamble
,
E.E.
,
Kumkar
,
S.N.
,
Tawre
,
M.S.
and
Pardesi
,
K.R.
(
2019
)
Emerging strategies to combat ESKAPE pathogens in the era of antimicrobial resistance: a review
.
Front. Microbiol.
10
,
539
.
31
Kaunietis,
,
A.
,
Buivydas,
,
A.
,
Čitavičius,
,
D.J.
and
Kuipers,
,
O.P.
(
2019
)
Heterologous biosynthesis and characterization of a glycocin from a thermophilic bacterium
.
Nat. Commun.
10
,
1115
32
Li
,
Y.
,
Han
,
Y.
,
Zeng
,
Z.
,
Li
,
W.
,
Feng
,
S.
and
Cao
,
W.
(
2021
)
Discovery and bioactivity of the novel lasso peptide microcin Y
.
J. Agric. Food Chem.
69
,
8758
8767
33
Ongley
,
S.E.
,
Bian
,
X.
,
Zhang
,
Y.
,
Chau
,
R.
,
Gerwick
,
W.H.
,
Müller
,
R.
et al (
2013
)
High-titer heterologous production in E. coli of lyngbyatoxin, a protein kinase C activator from an uncultured marine cyanobacterium
.
ACS Chem. Biol.
8
,
1888
1893
34
Johnston
,
C.W.
,
Badran
,
A.H.
and
Collins
,
J.J.
(
4202
)
Continuous bioactivity-dependent evolution of an antibiotic biosynthetic pathway
.
Nat. Commun.
202
,
4202
.
35
Zhang
,
H.
,
Wang
,
Y.
,
Wu
,
J.
,
Skalina
,
K.
and
Pfeifer
,
B.A.
(
2010
)
Complete biosynthesis of erythromycin A and designed analogs using E. coli as a heterologous host
.
Chem. Biol.
17
,
1232
1240
36
Cummings
,
M.
,
Peters
,
A.D.
,
Whitehead
,
G.F.S.
,
Menon
,
B.R.K.
,
Micklefield
,
J.
,
Webb
,
S.J.
et al (
2019
)
Assembling a plug-and-play production line for combinatorial biosynthesis of aromatic polyketides in Escherichia coli
.
PLoS Biol.
17
,
e3000347
37
Liu
,
X.
,
Hua
,
K.
,
Liu
,
D.
,
Wu
,
Z.L.
,
Wang
,
Y.
,
Zhang
,
H.
et al (
2020
)
Heterologous biosynthesis of type II polyketide products using E. coli
.
ACS Chem. Biol.
15
,
1177
1183
38
Greunke
,
C.
,
Duell
,
E.R.
,
D'Agostino
,
P.M.
,
Glöckle
,
A.
,
Lamm
,
K.
and
Gulder
,
T.A.M.
(
2018
)
Direct pathway cloning (DiPaC) to unlock natural product biosynthetic potential
.
Metab. Eng.
47
,
334
345
39
D'Agostino
,
P.M.
and
Gulder
,
T.A.M.
(
2018
)
Direct pathway cloning combined with sequence- and ligation-independent cloning for fast biosynthetic gene cluster refactoring and heterologous expression
.
ACS Synth. Biol.
7
,
1702
1708
40
Fu
,
J.
,
Bian
,
X.
,
Hu
,
S.
,
Wang
,
H.
,
Huang
,
F.
,
Seibert
,
P.M.
et al (
2012
)
Full-length RecE enhances linear-linear homologous recombination and facilitates direct cloning for bioprospecting
.
Nat. Biotechnol.
30
,
440
446
41
Jiang
,
W.
and
Zhu
,
T.F.
(
2016
)
Targeted isolation and cloning of 100-kb microbial genomic sequences by Cas9-assisted targeting of chromosome segments
.
Nat. Protoc.
11
,
960
975
42
Zhang
,
J.J.
,
Tang
,
X.
,
Zhang
,
M.
,
Nguyen
,
D.
,
Moore
,
B.S.
,
Davies
,
J.E.
et al (
2022
)
Broad-Host-Range expression reveals native and host regulatory elements that influence heterologous antibiotic production in gram-negative bacteria
.
mBio
8
,
e01291-17
43
Yin
,
J.
,
Hoffmann
,
M.
,
Bian
,
X.
,
Tu
,
Q.
,
Yan
,
F.
,
Xia
,
L.
et al (
2015
)
Direct cloning and heterologous expression of the salinomycin biosynthetic gene cluster from Streptomyces albus DSM41398 in Streptomyces coelicolor A3(2)
.
Sci. Rep.
5
,
15081
.
44
Alam
,
K.
,
Hao
,
J.
,
Zhang
,
Y.
and
Li
,
A.
(
2021
)
Synthetic biology-inspired strategies and tools for engineering of microbial natural product biosynthetic pathways
.
Biotechnol. Adv.
49
,
107759
.
45
Engler
,
C.
,
Gruetzner
,
R.
,
Kandzia
,
R.
and
Marillonnet
,
S.
(
2009
)
Golden gate shuffling: a one-pot DNA shuffling method based on type IIs restriction enzymes
.
PLoS One
4
,
e5553
46
Ren
,
H.
,
Hu
,
P.
and
Zhao
,
H.
(
2017
)
A plug-and-play pathway refactoring workflow for natural product research in Escherichia coli and Saccharomyces cerevisiae
.
Biotechnol. Bioeng.
114
,
1847
1854
47
Ren
,
H.
,
Biswas
,
S.
,
Ho
,
S.
,
van der Donk
,
W.A.
and
Zhao
,
H.
(
2018
)
Rapid discovery of glycocins through pathway refactoring in Escherichia coli
.
ACS Chem. Biol.
13
,
2966
2972
48
Chong
,
S.
(
2014
)
Overview of cell-free protein synthesis: historic landmarks, commercial systems, and expanding applications
.
Curr. Protoc. Mol. Biol.
108
,
16.30.1
16.30.11
49
Carlson
,
E.D.
,
Gan
,
R.
,
Hodgman
,
C.E.
and
Jewett
,
M.C.
(
2012
)
Cell-free protein synthesis: Applications come of age
.
Biotechnol. Adv.
30
,
1185
1194
50
Silverman
,
A.D.
,
Karim
,
A.S.
and
Jewett
,
M.C.
(
2020
)
Cell-free gene expression: an expanded repertoire of applications
.
Nat. Rev. Genet.
21
,
151
170
51
Karim
,
A.S.
,
Dudley
,
Q.M.
,
Juminaga
,
A.
,
Yuan
,
Y.
,
Crowe
,
S.A.
,
Heggestad
,
J.T.
et al (
2020
)
In vitro prototyping and rapid optimization of biosynthetic enzymes for cell design
.
Nat. Chem. Biol.
16
,
912
919
52
Dudley
,
Q.M.
,
Karim
,
A.S.
,
Nash
,
C.J.
and
Jewett
,
M.C.
(
2020
)
In vitro prototyping of limonene biosynthesis using cell-free protein synthesis
.
Metab. Eng.
61
,
251
260
53
Valliere
,
M.A.
,
Korman
,
T.P.
,
Arbing
,
M.A.
and
Bowie
,
J.U.
(
2020
)
A bio-inspired cell-free system for cannabinoid production from inexpensive inputs
.
Nat. Chem. Biol.
16
,
1427
1433
54
Ozaki
,
T.
,
Yamashita
,
K.
,
Goto
,
Y.
,
Shimomura
,
M.
,
Hayashi
,
S.
,
Asamizu
,
S.
et al (
2017
)
Dissection of goadsporin biosynthesis by in vitro reconstitution leading to designer analogues expressed in vivo
.
Nat. Commun.
8
,
14207
55
Fleming
,
S.R.
,
Bartges
,
T.E.
,
Vinogradov
,
A.A.
,
Kirkpatrick
,
C.L.
,
Goto
,
Y.
,
Suga
,
H.
et al (
2019
)
Flexizyme-enabled benchtop biosynthesis of thiopeptides
.
J. Am. Chem. Soc.
141
,
758
762
56
Vinogradov
,
A.A.
,
Shimomura
,
M.
,
Goto
,
Y.
,
Ozaki
,
T.
,
Asamizu
,
S.
,
Sugai
,
Y.
et al (
2020
)
Minimal lactazole scaffold for in vitro thiopeptide bioengineering
.
Nat. Commun.
11
,
2272
57
Si
,
Y.
,
Kretsch
,
A.M.
,
Daigh
,
L.M.
,
Burk
,
M.J.
and
Mitchell
,
D.A.
(
2021
)
Cell-free biosynthesis to evaluate lasso peptide formation and enzyme-substrate tolerance
.
J. Am. Chem. Soc.
143
,
5917
5927
58
Dong
,
H.
,
Li
,
J.
,
Liu
,
H.
,
Lu
,
S.
,
Wu
,
J.
,
Zhang
,
Y.
et al (
2022
)
Design and ribosomal incorporation of noncanonical disulfide-directing motifs for the development of multicyclic peptide libraries
.
J. Am. Chem. Soc.
144
,
5116
5125
59
Yang
,
D.
,
Park
,
S.Y.
,
Park
,
Y.S.
,
Eun
,
H.
and
Lee
,
S.Y.
(
2020
)
Metabolic engineering of Escherichia coli for natural product biosynthesis
.
Trends Biotechnol.
38
,
745
765
60
Dong
,
H.
,
Cui
,
Y.
and
Zhang
,
D.
(
2021
)
CRISPR/cas technologies and their applications in Escherichia coli
.
Front. Bioeng. Biotechnol.
9
,
762676
61
Zeng
,
Y.
,
Hong
,
Y.
,
Azi
,
F.
,
Liu
,
Y.
,
Chen
,
Y.
,
Guo
,
C.
et al (
2022
)
Advanced genome-editing technologies enable rapid and large-scale generation of genetic variants for strain engineering and synthetic biology
.
Curr. Opin. Microbiol.
69
,
102175
62
Zhang
,
H.
,
Skalina
,
K.
,
Jiang
,
M.
and
Pfeifer
,
B.A.
(
2012
)
Improved E. coli erythromycin a production through the application of metabolic and bioprocess engineering
.
Biotechnol. Prog.
28
,
292
296
63
Amiram
,
M.
,
Haimovich
,
A.D.
,
Fan
,
C.
,
Wang
,
Y.S.
,
Aerni
,
H.R.
,
Ntai
,
I.
et al (
2015
)
Evolution of translation machinery in recoded bacteria enables multi-site incorporation of nonstandard amino acids
.
Nat. Biotechnol.
33
,
1272
1279
64
Kakkar
,
N.
,
Perez
,
J.G.
,
Liu
,
W.R.
,
Jewett
,
M.C.
and
van der Donk
,
W.A.
(
2018
)
Incorporation of nonproteinogenic amino acids in class I and II lantibiotics
.
ACS Chem. Biol.
13
,
951
957
65
Shomar
,
H.
,
Gontier
,
S.
,
van den Broek
,
N.J.F.
,
Mora H
,
T.
,
Noga
,
M.J.
,
Hagedoorn
,
P.L.
et al (
2018
)
Metabolic engineering of a carbapenem antibiotic synthesis pathway in Escherichia coli
.
Nat. Chem. Biol.
14
,
794
800
66
Fang
,
L.
,
Guell
,
M.
,
Church
,
G.M.
and
Pfeifer
,
B.A.
(
2018
)
Heterologous erythromycin production across strain and plasmid construction
.
Biotechnol. Prog.
34
,
271
276
67
Li
,
G.
and
Lou
,
H.X.
(
2018
)
Strategies to diversify natural products for drug discovery
.
Med. Res. Rev.
38
,
1255
1294
68
Harvey
,
C.J.B.
,
Puglisi
,
J.D.
,
Pande
,
V.S.
,
Cane
,
D.E.
and
Khosla
,
C.
(
2012
)
Precursor directed biosynthesis of an orthogonally functional erythromycin analogue: selectivity in the ribosome macrolide binding pocket
.
J. Am. Chem. Soc.
134
,
12259
12265
69
Hou
,
L.
,
Li
,
Y.
,
Wu
,
Q.
,
Li
,
M.
,
Older
,
E.A.
,
Tang
,
X.
et al (
2021
)
Discovery of anti-infective adipostatins through bioactivity-guided isolation and heterologous expression of a type III polyketide synthase
.
Bioorg. Chem.
112
,
104925
70
Rittner
,
A.
,
Joppe
,
M.
,
Schmidt
,
J.J.
,
Mayer
,
L.M.
,
Heid
,
E.
,
Sherman
,
D.H.
et al (
2021
)
Directed biosynthesis of fluorinated polyketides
.
bioRxiv
2021.07.30.454469.
71
Sirirungruang
,
S.
,
Ad
,
O.
,
Privalsky
,
T.M.
,
Ramesh
,
S.
,
Sax
,
J.L.
,
Dong
,
H.
et al (
2022
)
Engineering site-selective incorporation of fluorine into polyketides
.
Nat. Chem. Biol.
18
,
886
893
72
Soeriyadi
,
A.H.
,
Ongley
,
S.E.
,
Kehr
,
J.
,
Pickford
,
R.
,
Dittmann
,
E.
and
Neilan
,
B.A.
(
2022
)
Tailoring enzyme stringency masks the multispecificity of a lyngbyatoxin (indolactam alkaloid) nonribosomal peptide synthetase
.
Chembiochem
23
,
e202100574
73
Wals
,
K.
and
Ovaa
,
H.
(
2014
)
Unnatural amino acid incorporation in E. coli: current and future applications in the design of therapeutic proteins
.
Front. Chem.
2
,
15
.
74
Fang
,
L.
,
Zhang
,
G.
,
El-Halfawy
,
O.
,
Simon
,
M.
,
Brown
,
E.D.
and
Pfeifer
,
B.A.
(
2018
)
Broadened glycosylation patterning of heterologously produced erythromycin
.
Biotechnol. Bioeng.
115
,
2771
2777
75
Zhao
,
X.
,
Xu
,
Y.
,
Viel
,
J.H.
and
Kuipers
,
O.P.
(
2021
)
Semisynthetic macrocyclic lipo-lanthipeptides display antimicrobial activity against bacterial pathogens
.
ACS Synth. Biol.
10
,
1980
1991
76
Pinu
,
F.
and
Villas-Boas
,
S.
(
2017
)
Extracellular microbial metabolomics: the state of the art
.
Metabolites
7
,
43
77
Pinu
,
F.
,
Villas-Boas
,
S.
and
Aggio
,
R.
(
2017
)
Analysis of intracellular metabolites from microorganisms: quenching and extraction protocols
.
Metabolites
7
,
53
78
Ito
,
T.
and
Masubuchi
,
M.
(
2014
)
Dereplication of microbial extracts and related analytical technologies
.
J. Antibiot.
67
,
353
360
79
Heatley
,
N.G.
(
1944
)
A method for the assay of penicillin
.
Biochem. J.
38
,
61
65
80
Alatoom
,
A.A.
,
Cunningham
,
S.A.
,
Ihde
,
S.M.
,
Mandrekar
,
J.
and
Patel
,
R.
(
2011
)
Comparison of direct colony method versus extraction method for identification of gram-positive cocci by use of Bruker biotyper matrix-assisted laser desorption ionization-time of flight mass spectrometry
.
J. Clin. Microbiol.
49
,
2868
2873
81
Si
,
T.
,
Tian
,
Q.
,
Min
,
Y.
,
Zhang
,
L.
,
Sweedler J
,
V.
,
van der Donk
,
W.A.
et al (
2018
)
Rapid screening of lanthipeptide analogs via in-colony removal of leader peptides in Escherichia coli
.
J. Am. Chem. Soc.
140
,
11884
11888
82
Watrous
,
J.
,
Roach
,
P.
,
Heath
,
B.
,
Alexandrov
,
T.
,
Laskin
,
J.
and
Dorrestein
,
P.C.
(
2013
)
Metabolic profiling directly from the petri dish using nanospray desorption electrospray ionization imaging mass spectrometry
.
Anal. Chem.
85
,
10385
10391
83
Hsu
,
C.C.
,
Elnaggar
,
M.S.
,
Peng
,
Y.
,
Fang
,
J.
,
Sanchez
,
L.M.
,
Mascuch
,
S.J.
et al (
2013
)
Real-time metabolomics on living microorganisms using ambient electrospray ionization flow-probe
.
Anal. Chem.
85
,
7014
7018
84
Jaroszewicz,
,
W.
,
Morcinek-Orłowska,
,
J.
,
Pierzynowska,
,
K.
,
Gaffke,
,
L.
and
Wȩgrzyn
,
G.
(
2022
)
Phage display and other peptide display technologies
.
FEMS Microbiol. Rev.
46
,
fuab052
85
Hetrick
,
K.J.
,
Walker
,
M.C.
and
van der Donk
,
W.A.
(
2018
)
Development and application of yeast and phage display of diverse lanthipeptides
.
ACS Cent. Sci.
4
,
458
467
86
Urban
,
J.H.
,
Moosmeier
,
M.A.
,
Aumüller
,
T.
,
Thein
,
M.
,
Bosma
,
T.
,
Rink
,
R.
et al (
2017
)
Phage display and selection of lanthipeptides on the carboxy-terminus of the gene-3 minor coat protein
.
Nat. Commun.
8
,
1500
87
Tucker
,
A.T.
,
Leonard
,
S.P.
,
DuBois
,
C.D.
,
Knauf
,
G.A.
,
Cunningham
,
A.L.
,
Wilke
,
C.O.
et al (
2018
)
Discovery of next-generation antimicrobials through bacterial self-screening of surface-displayed peptide libraries
.
Cell
172
,
618
628.e13
88
Koch
,
P.
,
Schmitt
,
S.
,
Cardner
,
M.
,
Beerenwinkel
,
N.
,
Panke
,
S.
and
Held
,
M.
(
2022
)
Discovery of antimicrobials by massively parallelized growth assays (Mex)
.
Sci. Rep.
12
,
4097
89
Hills
,
E.
,
Woodward
,
T.J.
,
Fields
,
S.
and
Brandsen
,
B.M.
(
2022
)
Comprehensive mutational analysis of the lasso peptide klebsidin
.
ACS Chem. Biol.
17
,
998
1010
90
Dejong
,
M.P.
,
Ritter
,
S.C.
,
Fransen
,
K.A.
,
Tresnak
,
D.T.
,
Golinski
,
A.W.
and
Hackel
,
B.J.
(
2021
)
A platform for deep sequence-activity mapping and engineering antimicrobial peptides
.
ACS Synth. Biol.
10
,
2689
2704
91
Mitchler
,
M.M.
,
Garcia
,
J.M.
,
Montero
,
N.E.
and
Williams
,
G.J.
(
2021
)
Transcription factor-based biosensors: a molecular-guided approach for natural product engineering
.
Curr. Opin. Biotechnol.
69
,
172
181
92
Ding
,
N.
,
Zhou
,
S.
and
Deng
,
Y.
(
2021
)
Transcription-factor-based biosensor engineering for applications in synthetic biology
.
ACS Synth. Biol.
10
,
911
922
93
Kasey
,
C.M.
,
Zerrad
,
M.
,
Li
,
Y.
,
Cropp
,
T.A.
and
Williams
,
G.J.
(
2018
)
Development of transcription factor-based designer macrolide biosensors for metabolic engineering and synthetic biology
.
ACS Synth. Biol.
7
,
227
239
94
Li
,
Y.
,
Reed
,
M.
,
Wright
,
H.T.
,
Cropp
,
T.A.
and
Williams
,
G.J.
(
2021
)
Development of genetically encoded biosensors for reporting the methyltransferase-dependent biosynthesis of semisynthetic macrolide antibiotics
.
ACS Synth. Biol.
10
,
2520
2531
95
Miller
,
C.A.
,
Ho
,
J.M.
,
Parks
,
S.E.
and
Bennett
,
M.R.
(
2021
)
Macrolide biosensor optimization through cellular substrate sequestration
.
ACS Synth. Biol.
10
,
258
264
96
d'Oelsnitz
,
S.
,
Kim
,
W.
,
Burkholder
,
N.T.
,
Javanmardi
,
K.
,
Thyer
,
R.
,
Zhang
,
Y.
et al (
2022
)
Using fungible biosensors to evolve improved alkaloid biosynthesis
.
Nat. Chem. Biol.
18
,
981
989
97
Kalkreuter
,
E.
,
Keeler
,
A.M.
,
Malico
,
A.A.
,
Bingham
,
K.S.
,
Gayen
,
A.K.
and
Williams
,
G.J.
(
2019
)
Development of a genetically encoded biosensor for detection of polyketide synthase extender units in Escherichia coli
.
ACS Synth. Biol.
8
,
1391
1400
98
Bauman
,
K.D.
,
Butler
,
K.S.
,
Moore
,
B.S.
and
Chekan
,
J.R.
(
2021
)
Genome mining methods to discover bioactive natural products
.
Nat. Prod. Rep.
38
,
2100
2129
99
Zhang
,
J.J.
and
Moore
,
B.S.
(
2020
)
Site-directed mutagenesis of large biosynthetic gene clusters via oligonucleotide recombineering and CRISPR/Cas9 targeting
.
ACS Synth. Biol.
9
,
1917
1922
100
Zhang
,
Y.
,
Chen
,
H.
,
Zhang
,
Y.
,
Yin
,
H.
,
Zhou
,
C.
and
Wang
,
Y.
(
2021
)
Direct RBS engineering of the biosynthetic gene cluster for efficient productivity of violaceins in E. coli
.
Microb. Cell Factories
20
,
38
101
Cleto
,
S.
and
Lu
,
T.K.
(
2017
)
An engineered synthetic pathway for discovering nonnatural nonribosomal peptides in Escherichia coli
.
mBio
8
,
e01474-17
102
Burkhart
,
B.J.
,
Kakkar
,
N.
,
Hudson
,
G.A.
,
van der Donk
,
W.A.
and
Mitchell
,
D.A.
(
2017
)
Chimeric leader peptides for the generation of non-natural hybrid RiPP products
.
ACS Cent. Sci.
3
,
629
638
103
Feng
,
J.
,
Li
,
R.
,
Zhang
,
S.
,
Bu
,
Y.
,
Chen
,
Y.
,
Cui
,
Y.
et al (
2020
)
Bioretrosynthesis of functionalized N-heterocycles from glucose via one-pot tandem collaborations of designed microbes
.
Adv. Sci.
7
,
2001188
104
Yang
,
X.
,
Lennard
,
K.R.
,
He
,
C.
,
Walker
,
M.C.
,
Ball
,
A.T.
,
Doigneaux
,
C.
et al (
2018
)
A lanthipeptide library used to identify a protein-protein interaction inhibitor
.
Nat. Chem. Biol.
14
,
375
380
105
Sarkar
,
A.
,
Kim
,
E.Y.
,
Jang
,
T.
,
Hongdusit
,
A.
,
Kim
,
H.
,
Choi
,
J.M.
et al (
2021
)
Microbially guided discovery and biosynthesis of biologically active natural products
.
ACS Synth. Biol.
10
,
1505
1519
106
Lautenschläger
,
N.
,
Popp
,
P.F.
and
Mascher
,
T.
(
2020
)
Development of a novel heterologous β-lactam-specific whole-cell biosensor in Bacillus subtilis
.
J. Biol. Eng.
14
,
21
107
Yin
,
J.
,
Cheng
,
D.
,
Zhu
,
Y.
,
Liang
,
Y.
and
Yu
,
Z.
(
2022
)
Development of a whole-cell biosensor for detection of antibiotics targeting bacterial cell envelope in Bacillus subtilis
.
Appl. Microbiol. Biotechnol.
106
,
789
798
108
Casini
,
A.
,
Chang
,
F.Y.
,
Eluere
,
R.
,
King
,
A.M.
,
Young
,
E.M.
,
Dudley
,
Q.M.
et al (
2018
)
A pressure test to make 10 molecules in 90 days: external evaluation of methods to engineer biology
.
J. Am. Chem. Soc.
140
,
4302
4316
109
Yi
,
L.
,
Luo
,
L.
and
,
X.
(
2018
)
Efficient exploitation of multiple novel bacteriocins by combination of complete genome and peptidome
.
Front. Microbiol.
9
,
1567
110
Jin
,
X.
,
Yao
,
J.
,
Fan
,
H.
,
Che
,
Y.
,
Pan
,
J.
,
Zhang
,
L.
et al (
2019
)
Heterologous expression and purification of BtCspB, a novel cold-shock protein-like bacteriocin from Bacillus thuringiensis BRC-ZYR2
.
World J. Microbiol. Biotechnol.
35
,
23
111
Yang
,
Y.
,
Chen
,
F.
,
Chen
,
H.Y.
,
Peng
,
H.
,
Hao
,
H.
and
Wang
,
K.J.
(
2020
)
A novel antimicrobial peptide scyreprocin from Mud crab scylla paramamosain showing potent antifungal and anti-biofilm activity
.
Front. Microbiol.
11
,
1589
112
Horbal
,
L.
,
Marques
,
F.
,
Nadmid
,
S.
,
Mendes M
,
V.
and
Luzhetskyy
,
A.
(
2018
)
Secondary metabolites overproduction through transcriptional gene cluster refactoring
.
Metab. Eng.
49
,
299
315
113
Liu
,
T.
,
Mazmouz
,
R.
,
Ongley
,
S.E.
,
Chau
,
R.
,
Pickford
,
R.
,
Woodhouse
,
J.N.
et al (
2017
)
Directing the heterologous production of specific cyanobacterial toxin variants
.
ACS Chem. Biol.
12
,
2021
2029
114
Liu
,
T.
,
Mazmouz
,
R.
,
Pearson
,
L.A.
and
Neilan
,
B.A.
(
2019
)
Mutagenesis of the microcystin tailoring and transport proteins in a heterologous cyanotoxin expression system
.
ACS Synth. Biol.
8
,
1187
1194
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).