Skip to main content

Metabolic engineering approaches for the biosynthesis of antibiotics

Abstract

Background

Antibiotics have been saving countless lives from deadly infectious diseases, which we now often take for granted. However, we are currently witnessing a significant rise in the emergence of multidrug-resistant (MDR) bacteria, making these infections increasingly difficult to treat in hospitals.

Main text

The discovery and development of new antibiotic has slowed, largely due to reduced profitability, as antibiotics often lose effectiveness quickly as pathogenic bacteria evolve into MDR strains. To address this challenge, metabolic engineering has recently become crucial in developing efficient enzymes and cell factories capable of producing both existing antibiotics and a wide range of new derivatives and analogs. In this paper, we review recent tools and strategies in metabolic engineering and synthetic biology for antibiotic discovery and the efficient production of antibiotics, their derivatives, and analogs, along with representative examples.

Conclusion

These metabolic engineering and synthetic biology strategies offer promising potential to revitalize the discovery and development of new antibiotics, providing renewed hope in humanity’s fight against MDR pathogenic bacteria.

Introduction

Antibiotics are one of the most revolutionary discoveries in the twentieth century that have significantly enhanced the lifespan of human beings. Before the discovery of penicillin, which is the first antibiotic to be discovered, infectious diseases accounted for high morbidity and mortality worldwide, resulting in a low average life expectancy of about 47 years [1]. One-third of Europe’s population perished due to pandemics, such as the bubonic plague, from 1347 to 1350. Additionally, until the early 1910s, infectious diseases accounted for 25% of mortality, making them a major cause of death [2]. Penicillin was identified by Alexander Fleming through a serendipitous discovery that an active metabolite from the culture of Penicillium notatum inhibited the growth of a pathogen Staphylococcus. Afterward, the mortality rate due to infectious diseases has decreased sharply to less than 1%, and penicillin could save a lot of wounded soldiers from dying by infections during World War II [3]. Acknowledging the discovery of penicillin and the development of efficient bioprocesses for industrial-scale production, Fleming, Florey and Chain were awarded the Nobel Prize.

Ever since the first discovery of penicillin, numerous antibiotics have been discovered from nature, saving countless patients from infectious diseases. Antibiotics can be largely classified into chemicals [4], metal complexes [5], and peptides [6]. Of particular, the major categories of chemical antibiotics are β-lactams, penicillins, cephalosporins, monobactams, tetracyclines, and quinolones [7]. Antibiotics can also be classified according to their mechanism of action (MoA) such as inhibition of DNA replication, RNA synthesis, protein synthesis, cell wall biosynthesis, cell membrane biosynthesis, or fatty acid synthesis. The different MoAs of antibiotics can serve as a starting point for the discovery of new antibiotics (Table 1) [8].

Table 1 Major antibiotics discovered from nature or synthesized from natural precursors

While antibiotics are essential for treating infectious diseases, inevitable evolution of pathogens has led to the emergence of resistance towards antibiotics, mainly in hospitals. As a result of the emergence of these multidrug-resistant (MDR) bacteria, at least 1.27 million people have died globally, and in 2019, nearly 5 million deaths were reported. In the United States, over 2.8 million patients annually suffer from infections caused by MDR bacteria [9]. One of the major mechanisms for acquiring antibiotic resistance is horizontal gene transfer (HGT). Once genes involved in antibiotic resistance emerge, they can be easily and rapidly transferred to other bacteria through HGT [10]. Due to the lower profitability of antibiotic products resulting from the rapid emergence of new antibiotic-resistant pathogens, pharmaceutical companies have reduced their investment in antibiotic discovery, leading to a decreased pace of new antibiotic development. In particular, we are witnessing a severe natural antibiotic discovery void over the last couple of decades [11]. For these reasons, the development of novel strategies to discover and develop new antibiotics have been rapidly increasing.

This review focuses on antibiotic chemicals that can be produced from biological processes. Recent discovery of natural antibiotics as well as metabolic engineering and synthetic biology strategies for the development of novel antibiotic derivatives are discussed. The readers are guided to some excellent reviews on the discovery and development of new antibiotics as provided here [11,12,13,14,15,16].

Discovery of antibiotics produced from actinomycetes

Importance of actinomycetes in antibiotics discovery and production

As one of the most complex habitats for diverse microorganisms, soil provides a rich ecosystem known as the soil microbiome. Since the discovery of soil bacteria isolated from the roots of legumes [103], the microbial diversity within soil samples have been explored. It is estimated that the maximum number of operational taxonomic units (OTUs) in soil is 52,000 [104]. To survive and thrive in such a highly competitive environment, many soil bacteria have evolved to produce antibiotics that help them outcompete surrounding competitors [105]. Of particular, a significant proportion of antibiotics currently in use were isolated from actinomycetes, a group of filamentous gram-positive bacteria that contributes to the diversity of the soil microbial ecosystem. They are one of the prolific producers of natural products and antibiotics β-lactams, tetracyclines, rifamycins, aminoglycosides, macrolides, and glycopeptides. Therefore, they are widely utilized as chassis strains for the production of a wide array of antibiotics [106]. As shown in Fig. 1, new antibiotics were explosively discovered from actinomycetes during 1940s to 1960s, a period referred to as ‘the golden era of antibiotic discovery’ [107]. Despite the rapid decrease in the discovery of antibiotics from nature, the potential for new antibiotics still remains undiscovered within the genomes of actinomycetes (Fig. 1).

Fig. 1
figure 1

The timeline of antibiotic discoveries approved for clinical use. Representative antibiotic classes are shown; left side of circle indicates the year of discovery for the first chemical belonging to the class. Right side of circle shows the proceeded development way for clinical use. Relevant standards are provided in parentheses. The ‘Golden era’ of antibiotic discovery (from the 1950s to the 1960s) is highlighted in yellow lines. Antibiotics are categorized as follows: green, natural antibiotics; blue, semi-synthetic antibiotics; orange, synthetic antibiotics

Among diverse strains within actinomycetes, the Streptomyces species are reported to produce the most antibiotics (up to 55%) that were discovered from 1945 to 1978. With advancements in sequencing technologies, the cost of sequencing has become more affordable, enabling the analysis of diverse microbial genomes. The availability of a larger volume of genome sequence data for various Streptomyces species has led to the discovery of novel antibiotics that could not be directly identified from nature. For example, Streptomyces coelicolor, Streptomyces avermitilis, Streptomyces griseus, and Saccharopolyspora erythraea each have more than 20 biosynthetic gene clusters (BGCs) encoding secondary metabolites in their genomes. This demonstrates the complex metabolic and regulatory pathways of Streptomyces species and highlights the wide range of secondary metabolites that can be produced in distinct cultural environments. Therefore, the chance of discovery of new antibiotics from the diverse set of actinomycetes are still high. New Streptomyces species capable of producing new antibiotics are still being discovered from soil, as exemplified by the recent discovery of a new type of actinomycete, Streptomyces sp SM01 [108, 109]. Despite the high potential for discovering new antibiotics, industrial-scale production is challenging due to low titers, productivity, and yields [12].

Actinomycetes chassis strains for heterologous production of antibiotics

As natural producers of diverse antibiotics, actinomycetes have been widely employed for the production of many antibiotics at the industrial-scale. While genetic manipulation of actinomycetes is challenging due to their GC-rich genomes and complex morphological and physiological characteristics [110], heterologous production of antibiotics in traditional model microorganisms such as Escherichia coli and Saccharomyces cerevisiae are challenging due to their unfavorable metabolic and regulatory pathways for the expression of large BGCs containing many incompatible genetic elements. Therefore, a number of actinomycetes strains showing favorable characteristics for heterologous BGC expression have been selected as chassis strains for antibiotics production. These chassis candidates have abundant pools of precursors and cofactors required for antibiotics production, relatively well established genome engineering tools, simpler growth conditions, compatible gene expression elements, and high genetic element transformation efficiency. Such strains include S. coelicolor A3(2), Streptomyces albus, Streptomyces avermitilis MA-4680, S. albus J1074, Streptomyces lividans TK24, and Streptomyces venezuelae [111, 112].

Among them, S. albus is one of the most commonly used chassis strains for the heterologous expression of diverse BGCs. The genome of S. albus (6.8 Mbp harboring 5.8 K genes in S. albus J1074) is one of the smallest among Streptomyces species, which allows for higher genetic stability when introducing heterologous BGCs [113, 114]. Growing and screening actinomycetes is a time-consuming and labor-intensive process, necessitating a rapid mutagenesis and screening strategy. Therefore, atmospheric and room temperature plasma, an effective mutagenesis method, was combined with ribosome engineering in the natural salinomycin producer S. albus, resulting in the generation of an overproducer that achieved twice the concentration of salinomycin [115]. S. albus has also been demonstrated as an efficient host for the production of complex terpenoids, making it particularly useful for the functional expression of tailoring enzymes including P450 for terpenoid modification [116]. To construct a more efficient chassis strain, 15 known BGCs were deleted, resulting in enhanced metabolic flux toward the desired products [117]. S. coelicolor is another important chassis strain for the efficient production of many secondary metabolites (e.g., actinorhodin, chloramphenicol, and congocidine). As with S. albus, BGCs encoding pathways for competing secondary metabolites were deleted, and mutations were introduced in genes encoding ribosomal components (i.e., rpoB and rpsL) for the enhanced production of target chemicals [118]. Although engineering large BGCs in actinomycetes remains challenging, new synthetic biology tools and strategies are continuously being developed to facilitate the engineering of these highly potent hosts for industrial-scale antibiotic production.

Methods for screening new antibiotics

Phenotype screening

Despite the abundance of bacterial species in soil, only a small fraction (less than 1%) can be successfully cultured in the laboratory [119]. As a result, only a few of many natural products discovered in nature have been identified as antibiotics. To discover and identify antibiotics among many natural products, a primitive method ‘phenotype screening’ has been employed in the early days. Phenotype screening is an exploratory process that identifies chemicals with antibiotic properties by testing the viability of pathogens when treated with candidate chemicals, based on their observable effects on biochemical activities or MoA, without prior knowledge of the targets [120, 121].

Soil microbiome has been the primary source of antibiotics discovery, but recently, the human microbiome has been gaining interest due to the high chance of encountering pathogen invasions in the respiratory track. For example, a non-ribosomal peptide (NRP) antibiotic, lugdunin, was discovered by phenotype screening from 90 nasal Staphylococci. Staphylococcus lugdunensis IVK28 found in the human nose was identified to produce lugdunin which can kill a representative pathogen Staphylococcus aureus [122].

Phenotypic screening based on target pathogen viability does not provide any insight into the biochemical targets of antibiotic candidates. Therefore, antimicrobial activity screening has shifted from viability tests to specific biochemical target inhibition approaches [123]. However, when the target pathogen (e.g., Mycobacterium tuberculosis) is difficult to test in the lab due to slow growth and biocontainment regulations, the target essential surrogate E. coli (TESEC) platform can be used instead [124]. The TESEC platform is constructed by the deletion of an essential gene in E. coli and replacing it with a functional analog from the target pathogen, linking bacterial growth to the activity of the target enzyme. In this study, high-throughput screening of antibiotic targets was performed in a TESEC platform for M. tuberculosis alanine racemase, leading to the identification of benazepril as an effective antibiotic against M. tuberculosis [124]. As such, phenotypic screening is still being actively used to discover new antibiotics as well as providing insights into the new MoA of new antibiotics [14].

Antibiotic discovery based on mechanisms of action

As discussed above, determining the MoA of an antibiotic is much more challenging than simple discovery of an antibiotic [125]. Some of the major biochemical targets of antibiotics are as follows: essential elements for cell survival, cell wall and cell membrane synthesis, cell membrane permeability, electron transport, purine and purine nucleotide synthesis, DNA synthesis, and protein synthesis [126]. In this subsection, methods for identifying MoAs of antibiotics are discussed.

Bacterial cytological profiling

As a rapid and powerful method for identifying the cellular pathways affected by antibiotics, bacterial cytological profiling (BCP) can distinguish between inhibitors that impact different cellular pathways as well as different targets within the same pathway [125]. Therefore, when similar imaging results are obtained by BCP when two different antibiotics are compared, their MoAs can be considered to be similar [127]. During BCP, bacteria are visualized through fluorescent dye staining methods such as DNA staining with DAPI, SYTOX, and ethidium bromide (EtBr), cell membrane staining with FM4-64, and cell wall staining with crystal violet and calcofluor white. For example, BCP was employed to identify an antibiotic peptide MciZ, which was shown to target FtsZ, a cell mitosis protein, in Bacillus subtilis. Upon treatment with MciZ, BCP shows that B. subtilis cells have shown phenotypes (e.g., undivided cells and abnormal Z-ring distribution) that could also be observed by treatment of FtsZ inhibitors. The loss of the Z-ring can be observed by the length of the DAPI-stained nucleoid, which becomes longer to fill in the area where the Z-ring is lost [128, 129].

In another example, BCP was employed to elucidate the MoA of the analogue of pan-assay interference compounds (PAINS), previously known to contain rhodamine. BCP, FM-6–64, SYTOX-green, and DAPI were employed as fluorescent markers. Upon treatment with the PAINS analogue, inhibition of cell wall synthesis and DNA replication was observed, achieved by repressing thymidylate kinase which is an enzyme responsible for synthesizing pyrimidine DNA bases. This inhibition manifested through filamentation and chromosomal replication defects, as indicated by fluorescence of BCP [130].

Flow cytometry

Flow cytometry is a tool that can quickly and accurately analyze individual microbial cells, even if they cannot be cultivated in the laboratory. Flow cytometry allows the analysis of various chemical and physical phenotypes such as cellular type, viability, and gene expression using fluorescent markers [131]. For example, flow cytometry was employed to unveil the MoA of labdane diterpenes by labeling target bacteria with fluorescence to assess viability [132].

Flow cytometry is also useful for distinguishing live and dead cells by using SYTO9 which stains only live cells and PI which stains only dead cells. Live/dead cell assay by flow cytometry was used to find the best combinations of different antibiotics to enhance the antibacterial property. The combination of antimicrobial peptides, sphistin and sph12-38, with antibiotics such as azithromycin and rifampicin, led to 85.93% reduction in viability of a representative gram-negative pathogen Pseudomonas aeruginosa [133].

CRISPRi

Clustered regularly interspaced short palindromic repeats interference (CRISPRi) employs a catalytically inactive Cas9 endonuclease to specifically repress the transcription of target genes, guided by single guide RNAs (sgRNAs) [134]. CRISPRi is employed to reveal the MoA of an antibiotic by repressing the antibiotic target gene candidates within a pathogen. Since the target genes are mostly essential genes in bacteria, their decreased expression would result in very low cell viability. For example, the MoA of peziculone was analyzed using CRISPRi. When genes involved in cell wall synthesis (i.e., tagB and murB), biofilm formation, and essential metabolic pathways (e.g., fatty acid biosynthesis and protein biosynthesis) were knocked down, the antibiotic sensitivity of S. aureus was increased [135]. In another example, the target gene of irresistin-16, a derivative of SCH-79797, was identified through both BCP and CRISPRi [136]. When essential genes involved in folate metabolism, dfrA (encoding dihydrofolate reductase) and folC (encoding dihydrofolate synthase), are knocked down in B. subtilis, the cell viability was observed to be highly sensitive to antibiotics treatment. Furthermore, irresistin-16 was shown to disrupt membrane integrity, as evidenced by BCP and flow cytometry [136]. As such, the MoA of antibiotics can be effectively elucidated by the combinations of different approaches.

Proteomics

Proteomics is a method for studying complex protein mixtures, such as bacterial lysates or clinical tissue samples containing several thousands of proteins. One of the most fundamental methods of analyzing the proteome of a bacteria is two-dimensional (2D) gel electrophoresis. Another method is iTRAQ®, which relies on the tagging of proteolytically cleaved peptides from different samples, with each sample conjugated to a different tag. The specific proteins found by either 2D gel electrophoresis or iTRAQ® can be subsequently elucidated and quantified by liquid chromatography-mass spectrometry (LC–MS) or matrix-assisted laser desorption-ionization time-of-flight mass spectrometry (MALDI-TOF/MS), leading to the investigation of cellular pathways affected by antibiotic treatment [137, 138]. For instance, 2D gel electrophoresis and MS were employed to observe changes in protein production in Acinetobacter baumannii following treatment with the antibiotic sulbactam. As a result, the levels of essential proteins for cell survival, including the ATP-binding-cassette (ABC) transporter as well as the 30S and 50S ribosomal subunit proteins, were found to be reduced [139]. Also, the proteomic change of S. aureus after lactobionic acid treatment was investigated using iTRAQ®. The analysis of peptides tagged with the reporter marker iTRAQ® through LC–MS/MS revealed disruptions in the cell wall and the membrane integrity, as well as altered ABC transporter levels and cellular energy metabolism [140]. Such proteomic analysis will be useful to provide important insights about the MoA of new antibiotics.

Discovery and activation of cryptic BGCs for the biosynthesis of new antibiotics

Isolation of unculturable bacteria and recovery of products

As aforementioned, a significant number of unculturable bacteria remain elusive and have not yet been comprehensively analyzed. Many of these microorganisms are expected to be valuable for our lives since some of them could recycle various elements (e.g., carbon, nitrogen, and metals) from natural resources, and the others could produce a variety of natural products with unprecedented pharmaceutical activities. Therefore, it is important to isolate and recover these bacteria in order to elucidate the largely unexplored space of antibiotics from nature by reconstruction of the natural habitat as much as possible. This involves providing suitable stimuli such as temperature, osmosis, host conditions, chemical inducers or precursors, and specific interactions with other bacteria in the surroundings [141]. However, no matter how meticulously the culture environment is adjusted, many bacteria still fail to grow, making it challenging to point out the exact problem.

Influence and stimulation of neighboring bacteria can be an important factor for growing some unculturable bacteria. These neighboring bacteria can stimulate the growth of target bacteria by providing diffusible growth factors (e.g., siderophores, cAMP, and acyl-homoserine lactones) or through physical contact, although the exact mechanisms remain largely unknown. Therefore, co-culture with other bacteria found from the same environment might allow the target bacteria to grow in the laboratory conditions. For example, Bacillus marisflavi requires a modified acyl-desferrioxamine siderophore as the growth factor produced from Bacillus megaterium, a helper bacterium inhabiting the same environment. This facilitated the utilization of oxidized iron as an essential nutrient, effectively regulating cell homeostasis and thus promoting growth of B. marisflavi [142]. In another study, co-culture of Micromonospora sp. with Rhodococcus sp. resulted in the production of keycin, a poly-nitroglycosylated anthracycline [143]. It turned out that the anthracycline backbone was first biosynthesized by Micromonospora sp., which was then modified to benzoxocin by Rhodococcus sp., and then further to keycin [143]. A notable feature of keycin is its distinct mechanism apart from that of most anthracyclines. However, the exact mechanism of keycin acting as an antibiotic remains elusive. Co-culture of fungal species can be also useful, as exemplified by the co-culture of two Aspergillus species from mangrove could produce a novel compound called aspergicin, which showed antibacterial activities towards Bacillus proteus, E. coli, S. aureus, and B. subtilis [144].

However, co-culture also has a number of limitations, including challenges in culturing helper bacteria, difficulties in precisely understanding bacterial interactions, and elucidation of suitable bacterial co-culture pairs. This led to the development of the isolation chip (iChip) which is a multichannel device that creates multiple sections divided by semi-permeable membranes in which only a single cell can be isolated. When iChips are buried in the soil, a single bacterial cell can be isolated in each section in the natural habitat, which can then be recovered in the lab. As bacteria are grown in the natural habitat through iChip, a striking 50% of all bacteria from a soil sample could be isolated [145]. As a case study, teixobactin, a newly discovered NRP antibiotic produced by Eleftheria terrae, was identified using the iChip method. Teixobactin shows significant bactericidal activity against MDR gram-positive pathogens without any resistance reported so far [146]. The authors are guided to additional studies on teixobactin in the following literature [147,148,149,150]. Interestingly, a new NRP antibiotic clovibactin could also be discovered from the same bacterial species E. terrae subspecies carolina [148]. Clovibactin was shown to block cell wall biosynthesis, with an unusual structure and MoA, showing no sign of resistance development as well.

Discovery of cryptic BGCs and genome mining

Despite efforts to culture “unculturable” bacteria, a majority of them still cannot be recovered in the lab. In this regard, metagenomics can be employed to comprehensively analyze large amounts of genomic data obtained directly from environmental samples, without the need to culture these elusive microorganisms [151]. The rapid advancement of sequencing technologies has enabled the accumulation of huge metagenomic data that include a significant volume of BGCs for secondary metabolite biosynthesis. As a representative example, S. coelicolor A3(2) was found to have about 7,825 genes involved in > 20 BGCs [108]. Also, 17 BGCs were identified in a marine actinomycete Salinispora tropica, most of which were novel, including the one responsible for the production of a polyene macrolactam salinilactam A [152]. The development of next-generation sequencing (NGS) technologies, along with recent advances in nanopore sequencing and single-cell sequencing, has enabled the rapid sequencing of the genomes of many Streptomyces strains. This has led to the discovery of a significant number of cryptic genes which are not expressed under normal culture conditions. In nature, these cryptic genes are activated only under specific environmental conditions to assist the survival of host cells in diverse environments.

To elucidate the cryptic genes and BGCs from metagenomic data, metagenome mining tools such as MG-RAST, IMG/M, EBI Metagenomics, SILVAngs, MEGAN, QIIME, and Mothur can be employed [153, 154]. For instance, metagenomic analysis was used to predict the genes encoding polyketide synthases and associated enzymes, leading to the discovery of new polyketides that potentially harbor antibacterial properties [151, 155, 156]. The antiSMASH (antibiotics & Secondary Metabolite Analysis Shell) pipeline was first developed in 2011 and is currently the most widely used bioinformatics tool for the prediction of BGCs and their possible products [157]. For example, antiSMASH was used to predict the cryptic BGCs within Streptomyces globisporus SP6C4, a bacterial strain known for its use in suppressing plant diseases. As a result, 15 BGCs were predicted, among which some were shown to produce secondary metabolites with antibacterial activities, providing valuable information for agricultural applications [158]. The antiSMASH pipeline can also be employed to comprehensively screen BGCs from a large volume of database. For example, all the high quality bacterial genomes from GenBank (~ 3000 as of 2014) were analyzed using antiSMASH to search NRPS-encoding gene clusters. As a result, 96 previously unidentified NRPs were predicted to be produced by the identified NRPSs, most of which were C-terminally cyclized peptides. Based on the common structural properties of these peptides, 171 synthetic peptides were designed, nine among which exhibited antibacterial activity against the ESKAPE pathogens (the group of major MDR bacteria including Enterococcus faecium, S. aureus, Klebsiella pneumoniae, A. baumannii, P. aeruginosa and Enterobacter cloacae) and M. tuberculosis [159]. In another example, an NRP-PK hybrid, epifadin, was discovered through the isolation of the BGC by analyzing the genome of the nasal commensal bacterium Staphylococcus epidermidis IVK83 using antiSMASH [160]. Other metagenome mining tools are also available such as BAGEL [161], PRISM [162], RiPPER [163], and TOUCAN [164]. Such approaches enable the discovery of new antibiotics from the metagenomic data without the need to directly culture the bacteria.

Activation of cryptic BGCs

Genetic-level activation

After the elucidation of cryptic BGCs and the predicted products, activation of the cryptic BGCs from the host strain is required to produce, recover, and analyze the resulting chemicals. A representative method of activating BGCs in the native hosts is the insertion of strong constitutive promoters (e.g., ermE* and kasO*) in front of the BGCs by CRISPR/Cas9. For instance, in Streptomyces viridochromogenes, the insertion of the kasO* promoter upstream of the BGC encoding a type II polyketide synthase (PKS) resulted in the production of a previously unknown pentangular type II polyketide with a dihydrobenzo[α]naphthacenequinone core [165].

Other than direct activation of the biosynthetic pathway genes, pathway regulators can be employed to manipulate the expression of cryptic genes, as exemplified in Streptomyces chattanoogensis L10 [166]. By the overexpression of genes encoding three biosynthetic pathway regulators (ChaK, ChaK1, and ChaI) in S. chattanoogensis, the cryptic angucycline gene cluster could be activated, resulting in the successful production of new antibiotics, chattamycin A and B [166]. The large ATP-binding regulator (LAL) regulator family plays an important role in regulating the expression of genes related to type I modular PKSs, and thus can be found within the BGCs of various polyketide antibiotics such as avermectin, salinomycin, and staurosporine [167]. In another example, overexpressing astG1 which encodes a LAL family regulator in Streptomyces sp. XZQH13 found from the cryptic ansatrienin BGC led to the production of hydroxymycotrienins A and thiazinotrienomycin G, previously unachievable ansatrienin antibiotics [168].

Another interesting approach is the deletion of BGCs responsible for the production of already known antibiotics instead of reinforcing the pathways related to unknown compounds [169]. For example, genes responsible for the production of two of the most frequently rediscovered antibiotics, streptothricin and streptomycin, were deleted from the genomes of 11 actinomycetes. As a result, previously unreported variants of antibiotics including thiolactomycin, amicetin, phenanthroviridin, and 5-chloro-3-formylindole could be discovered.

Activation of the cryptic BGC using chemical elicitors

Other than through direct activation of the cryptic BGCs at the genetic-level, they can be sometimes activated upon the addition of specific chemicals due to the complicated metabolic and regulatory pathways of actinomycetes. When bacteria are exposed to antibiotics at sub-inhibitory concentrations, a new secondary metabolite pathway can be activated to generate new products. This induction of BGCs mainly occurs at the transcriptional level and can even lead to various gene expression changes that can lead to many phenotypic changes other than those related to secondary metabolite production. For example, an antibiotic streptomycin was shown to induce the expression of a cryptic type II PKS-related BGC in Microbispora sp. BCCAGE54, leading to the production of tetarimycin B [170]. In addition, alteration of nutrient addition in the culture medium can lead to the induction of cryptic BGCs. A notable example is the discovery of coelichelin produced from S. coelicolor A3(2) by culturing the strain in an iron-deficient condition [171]. Since coelichelin serves as a siderophore, which helps the host cell to acquire iron, culturing the strain in the absence of iron would have led to the enhanced production of coelichelin to help the cell survive in an iron-deficient environment.

Some metabolites produced from neighboring microorganisms can also act as chemical elicitors for cryptic BGCs. Therefore, co-culture of multiple bacterial strains could lead to the observation of unprecedented metabolites that could not have been observed by culturing each bacteria individually. As an example, when Micromonospora sp. UR56 and Actinokineospora sp. EG49 were co-cultured, a number of new metabolites that were not produced by individually culturing the two bacterial strains could be found [172]. Remarkably, all of these products were found to have come from cryptic BGCs that would have been hidden until induced by the co-culture. Among the new chemicals, dimethyl phenazine-1,6-dicarboxylate, phencomycin, and tubermycin demonstrated antibacterial activities [172].

Reconstruction of the cryptic BGCs

For cryptic genes that are difficult to be expressed in native hosts, the genes can be cloned and introduced into heterologous hosts to identify the products that the BGCs can produce. Since it is often difficult to express actinomycetal BGCs in widely used model microorganisms such as E. coli or S. cerevisiae, a number of Streptomyces chassis strains (e.g., S. coelicolor, S. lividans, and S. albus) have been used for facile introduction of actinomycetal cryptic BGCs. For example, as the cryptic ansamycin BGC (type I PKS) could not be activated in the marine actinomycete Streptomyces seoulensis A01 in the laboratory, the cryptic BGC cloned into a plasmid and was introduced in other Streptomyces hosts. As a result, successful heterologous production of ansaseomycins A and B was achieved using the actinomycetal chassis strains S. lividans SBT18 and S. coelicolor M1146 [173]. When the expression of cryptic BGCs in heterologous hosts is combined with genome mining tools (e.g., antiSMASH) for predicting cryptic BGCs from metagenomic data, the speed of discovering new antibiotics can be significantly enhanced.

Enhancing the production levels of antibiotics

Enhanced production of antibiotics by engineering actinomycetes

As previously discussed, actinomycetes are efficient microbial cell factories for the production of diverse antibiotics despite the difficulties of engineering. To facilitate the engineering process and to resolve such difficulties, several metabolic engineering and synthetic biology tools and strategies have been developed [174]. One of the most useful genome engineering tools is the CRISPR system [175, 176]. CRISPR-based genome engineering has allowed insertion and deletion of gene fragments, as well as editing of DNA and RNA bases. CRISPR has been recently adopted for facile engineering of actinomycetes to enhance their production capacities in order to better exploit these remarkable antibiotic producers. For instance, the genome of S. erythraea NRRL 23338, which produces erythromycin (a macrolide produced by type I PKS), was engineered by the CRISPR-Cas9 system [177]. As the expression levels of the ery cluster encoding the tailoring enzymes were low, which served as a significant bottleneck for erythromycin production, strong promoters were inserted in multiple loci within the ery cluster to enhance the transcription levels of the bottleneck genes. As a result, the erythromycin production was enhanced by six-fold when compared to the wild type strain [177]. Thus, the CRISPR-Cas9 system can be used to overexpress bottleneck genes to better streamline the metabolic flux towards the products. CRISPR-Cas9-based genome engineering is also useful for the deletion of major antibiotic pathways in order to redirect the metabolic flux towards other minor secondary metabolites, which would lead to previously undiscovered antibiotics. For example, the BGCs for the production of streptothricin or streptomycin were deleted in 11 actinomycete strains by using CRISPR-Cas9, leading to the discovery of previously unreported antibiotics including tiolactomycin, amicetin, and phenanthroviridin [169]. In addition, CRISPR can be used together with genetic elements of bacteriophage for the integration of large DNA fragments. For instance, the attachment and integration (Att/Int) system from bacteriophage ΦC31 was used together with CRISPR-Cas9 to integrate multiple copies of the large pristinamycin II BGC into the genome of Streptomyces pristinaespiralis [178]. This led to the production of 2.24 g/L of pristinamycin II by shake flask culture [178].

To avoid DNA double-strand breaks as well as the cytotoxic effects of Cas9, CRISPR-base editing system (CRISPR-BEST) was developed and was showcased in a non-model actinomycete Streptomyces collinus Tü365 [179]. The CRISPR-BEST system having a cytidine deaminase could manipulate the kirromycin biosynthetic pathway by inactivating the kirN gene by introducing stop codons within the gene. More recently, an improved base editing system (eSCBE3-ng-Hypa) with improved performance towards high GC DNA sequences, relaxed protospacer adjacent motif (PAM) requirement, and minimal off-target effects was developed in Streptomyces species [180]. The eSCBE3-ng-Hypa system was used to inactivate the competitive pathways within the ave BGC for enhanced production of avermectin B1a in S. avermitilis [180].

Given the complex metabolism and physiology of actinomycetes, it is often challenging to develop engineering strategies to enhance antibiotic production in these strains. Therefore, understanding the relationship between different metabolic and regulatory pathways and antibiotic production through omics analysis can help increase the production of diverse antibiotics. For example, comparative transcriptomic analysis was performed for Actinosynnema pretiosum ATCC 31280, an ansamitocin P-3 (AP-3) producer, to elucidate the cause for excessive mycelial fragmentation during fermentation [181]. As a result, the APASM_4178 gene encoding a subtilisin-like serine peptidase was identified to be responsible for mycelial fragmentation. As mycelial fragmentation had a negative impact on the yield of AP-3, the APASM_4178 gene led to increased cell growth as well as increased production of AP-3 by 43.65% [181]. In another study, metabolomic and transcriptomic analysis of S. avermitilis revealed that triacylglycerol (TAG) accumulated during cell growth was degraded during stationary phase, leading to increased metabolic flux towards acetyl-CoA, reducing equivalents, ATP, and thus polyketides [182]. As the sco6196 gene was shown to be responsible for the degradation of TAG, overexpression of the gene in an industrial S. avermitilis A56 strain resulted in significantly enhanced production of avermectin B1a (9.31 g/L) in a 180-m3 fermenter [182]. Another method of enhancing the production of antibiotics is co-culture of several bacterial strains to activate core BGCs. Co-culture of Vibrio coralliilyticus and Photobacterium galatheae has led to increased production of andrimid and holomycin by 4.3 and 2.7-fold, respectively, when compared to those produced by individually culturing each bacterial strain [183].

Antibiotic production from heterologous model microorganisms

Despite the capability of actinomycetes for the production of high-level antibiotics, they show several problems including difficulties in high-cell-density culture, cumbersome genome engineering, and complex metabolic and regulatory networks. Therefore, model microorganisms including E. coli and S. cerevisiae have been employed for the heterologous production of antibiotics. E. coli is especially known for its well-established genome-scale metabolic models, capability for high-cell-density culture, abundant genome engineering tools, and high growth rate [184, 185]. A classical example of antibiotics produced by metabolically engineered E. coli is erythromycin, which involves a type I PKS. By introducing dexoyerythronolide B synthase (DEBS) from S. erythraea into E. coli, 6-deoxyerythronolide (6-dEB), an aglycone precursor of erythromycin, was produced for the first time by a heterologous host [186]. Further increasing the metabolic flux towards the precursors (acetyl-CoA, propionyl-CoA, and methylmalonyl-CoA) as well as overexpression of genes encoding deoxysugar glycosyltransferase resulted in 4 mg/L of erythromycin A production [187]. Such examples showcase the capability of E. coli to produce macrolide antibiotics which require a mega-sized assembly line of enzymes such as type I PKS or NRPS. Compared with type I PKS, type II PKS had been rather difficult to express in heterologous hosts [188]. The recent identification of a type II PKS from Photorhabdus luminescens led to successful production of aromatic C16 polyketides in E. coli [189, 190]. E. coli capable of heterologous production of aromatic C16 polyketides was particularly useful for the production of non-natural derivatives, which can lead to the development of unprecedented new antibiotics. Carbapenem, a β-lactam antibiotic, could be also produced in E. coli by the introduction of the carABCDE BGC from Pectobacterium carotovorum as well as the removal of key feedback inhibition from glutamate 5-kinase (ProB) responsible for the conversion of glutamate to glutamyl 5-phosphate [191].

Other prokaryotic hosts have also shown to be suitable for antibiotics production. For example, Corynebacterium glutamicum is a gram-positive bacterium capable of efficiently producing food-grade products, but has suffered from reduced cell growth when supplemented with propionate, a precursor for propionyl-CoA and methylmalonyl-CoA. To address this issue, adaptive laboratory evolution was employed to improve the fitness of the host for polyketide production in the presence of propionate, resulting in an 18-fold increase in germicidin production compared to the wild-type C. glutamicum [192]. Pseudomonas species can also be employed for antibiotic production due to its high tolerance towards toxic chemicals, efficient metabolic pathway towards aromatic compounds, well-established genome engineering tools, as well as adaptability to industrial processes [193]; some Pseudomonas species are natural producers of polyketide antibiotics such as 2,4-diacetylphloroglucinol [194] or mupirocin [195]. As malonyl-CoA is a central metabolite for the production of a number of categories of secondary metabolites including polyketides and phenylpropanoids, Pseudomonas taiwanensis was engineered by the deletion of competing pathways, replacing the native 3-ketoacyl-ACP synthase II with that from Pseudomonas putida, and overexpression of acetyl-CoA carboxylase from C. glutamicum [196]. This resulted in the enhanced production of flaviolin, pinosylvin, and resveratrol, and the same strategy can also be applied to the production of antibiotics derived from malonyl-CoA.

Another important heterologous microorganism model for antibiotic production is S. cerevisiae due to its capability of functional expression of tailoring enzymes and well-established genome engineering tools, as well as adaptability to industrial application [197]. For example, penicillin, naturally produced by NRPS in Penicillium chrysogenum, was produced in S. cerevisiae. Co-expression of NRPS and NRPS activator genes (i.e., pcbAB and npgA) along with three additional genes (i.e., pcbC, pclA, and penDE) from P. chrysogenum resulted in the production of 70 ng/mL of benzylpenicillin production [198]. In another study, a type III PKS from Aloe arborescens was introduced in S. cerevisiae for the production of dihydrokalafungin, a precursor of the antibiotic actinorhodin [199]. Along with S. cerevisiae, the non-conventional yeast strain Yarrowia lipolytica can also be used for antibiotic production due to its ability to efficiently produce proteins, its high flux towards acetyl-CoA, and its capacity to accumulate high levels of lipids, which can dissolve hydrophobic chemicals at high concentrations [200]. Leveraging these advantages, Y. lipolytica was engineered by implementing a pyruvate bypass pathway and overexpressing PEX10, which is associated with β-oxidation, leading to a substantial production (35.9 g/L) of triacetic acid lactone (TAL) from glucose [201]. TAL produced from Y. lipolytica can be easily converted into the antibiotic pogostone and its analogs through a one-step chemical conversion [202]. The examples discussed above highlight the potential of engineering model microorganisms for efficient production of antibiotics, providing a viable response to the current lack of high-performance antibiotic production platforms.

Diversification of antibiotics

Engineering PKSs and tailoring enzymes

Diversification of antibiotics by addition of functional groups, modification of the carbon skeleton, and other structural alterations, is an effective strategy for developing new antibiotics with unprecedented properties to combat emerging MDR pathogens [203]. Analogs and derivatives generated through diversification can also facilitate the discovery of novel MoA for treating MDR bacteria or even overcoming physical barriers, such as biofilms. To diversify antibiotics, chemical or biochemical reactions are applied to lead compounds. While chemical reactions have been widely employed for this purpose, the structural complexity of many antibiotics, such as fused polycyclic carbon skeletons and multiple stereocenters, poses significant challenges for chemical synthesis [204]. Sensitive functional groups further restrict reaction conditions to prevent degradation or loss of efficacy. Therefore, synthetic biology has emerged to engineer enzymes and microbial strains that can efficiently produce diverse analogs and derivatives of antibiotics.

To produce diverse derivatives of a lead compound, tailoring enzymes are often employed to add various functional groups. A notable example is the biosynthesis of novel anthraquinones, a group of polycyclic aromatic polyketides, by the introduction of tailoring enzymes in addition to a type II PKS in E. coli [189]. In this study, a type II minimal PKS from P. luminescens that are phylogenetically close to E. coli fatty acid synthases was employed for the efficient biosynthesis of the carbon chain of C16 aromatic polyketides in E. coli. The introduction of an O-methyltransferase to the PKS-harboring strain resulted in the production of a novel methylated anthraquinone termed neomedicamycin, and the introduction of a halogenase to the same strain resulted in the production of a novel chlorinated anthraquinone termed neochaetomycin (Fig. 2A). Particularly, since chlorinated anthraquinones are reported to show enhanced antimicrobial activities as exemplified by chlorinated emodin [205], the production of halogenated polyketide derivatives demonstrate the potential for the development and production of new antibiotics. Glycosylation is another important reaction for improving the property of a lead compound. For example, the introduction of a glycosyltransferase YjiC in an E. coli strain harboring a type I iterative PKS capable of producing an antimicrobial anthraquinone emodin, resulted in the production of an emodin glucoside [206]. The glucoside showed enhanced solubility and stability when compared with those of emodin. Another example is the production of glycosylated erythromycin derivatives. To achieve this, a promiscuous glucosyltransferase EryBV was employed to attach deoxysugars to 6-dEB, resulting in the production of glycosylated erythromycin derivatives [207]. Additionally, ErtBV was capable of glycosylating 6-dEB using diverse deoxysugars including D-allose, D-forosamine, L-noviose, and D-vicenisamine, allowing the production of diverse 6-dEB glycosides [208]. Such ‘plug-and-play’ mode of biosynthesis is useful for the production of diverse derivatives and analogs of a lead compound by the introduction of different combinations of biosynthetic enzymes, showing great potential for the development and production of novel compounds with unprecedented antimicrobial activities.

Fig. 2
figure 2

Strategies for diversifying antibiotics. A Application of tailoring enzymes with type II PKS to diversify polyketide-based antibiotics. B Engineering of type I and III PKSs. Altering substrate-interacting domains in type I PKS changes starter/extender unit selectivity, facilitating the production of novel antibiotics. Production of fluorinated erythromycin using fluoromalonyl-CoA is shown as an example. For certain type III PKSs, substrate promiscuity can be employed to diversify products. Engineering the cavity near the active site of a type III PKS can alter the carbon chain lengths of the products. C As NRPS comprises functional domains, engineering these modular systems can lead to the production of a wide range of non-natural antibiotics

Due to the modular nature of PKS, engineering the PKS itself is another important strategy for the generation of diverse antibiotic derivatives. One notable example is the production of non-natural fluorinated erythromycin analogs. Fluorination is widely used for the chemical modification of drugs to give new pharmacokinetic properties [209, 210]. To produce fluorinated analogs of 6-dEB, the native extender unit for erythromycin biosynthesis, (2S)-methylmalonyl-CoA, should be replaced with fluoromalonyl-CoA. As the cis-AT within the modular PKS is primarily responsible for the gatekeeping of extender units, it was eliminated from the PKS, and a standalone trans-AT engineered for enhanced substrate selectivity towards fluoromalonyl-CoA was introduced instead (Fig. 2B) [211]. Fluoromalonyl-CoA could be produced within the cell by the supply of fluoromalonate, which was converted to fluoromalonyl-CoA by a malonyl-CoA synthetase MatB. In another study, the AT domain of the DEBS module 6 was exchanged with AT from 12 different PKSs, allowing the incorporation of diverse CoA units (e.g., ethylmalonyl-CoA, butylmalonyl-CoA, and benzylmalonyl-CoA) as extender units [212]. In addition to AT domain engineering, modifying other domains within type I PKS modules can improve the production of target chemicals by enhancing inter-modular interactions. For example, as the thioesterase (TE) domain plays a key role in substrate selectivity, site-directed mutagenesis of TE was employed to produce non-natural epimerized hexaketide [213].

Other than swapping domains within the assembly lines, introduction of point mutations at or near the active sites can also lead to the production of diverse compounds. For efficient and rapid editing of large NRPS assembly lines within natural producers, CRISPR-Cas9 gene editing tool was optimized, allowing the production of ten new lipopeptide variants of enduracidin with high yields [214]. Combinatorial engineering of PKS modules is not only used for the production of antibiotics, but also for a diverse portfolio of chemicals, even including small chemicals (e.g., adipic acid, lactones, and ketones.) that have been conventionally produced from petroleum (Fig. 2C) [215,216,217].

Mutagenesis of type III PKSs is another effective strategy for diversifying antibiotics by virtue of their promiscuous substrate specificity [218,219,220]. For example, a type III PKS from Huperzia serrata (HsPKS) accepted a non-natural starter unit, 2-carbamoylbenzoyl-CoA, instead of p-coumaroyl-CoA, producing a new polycyclic alkaloid (2-hydroxypyrido[2,1-a]isoindole-4,6-dione) with two malonyl-CoA molecules [221]. Expanding the active site cavity of HsPKS with the S348G mutation enabled the condensation of three malonyl-CoA molecules, producing another novel alkaloid (1,3-dihydroxy-5H-dibenzo[b,e]azepine-6,11-dione) [221]. Both non-natural alkaloids inhibited the formation of biofilm formation by MRSA. As discussed above, altering the cavity space for polyketide carbon chain elongation effectively diversifies polyketide chain lengths, as also shown in other type III PKS mutants [222, 223]. This shows the potential and versatility of engineering PKS for the production of diverse non-natural chemicals by engineered microbial cell factories.

Engineered PKS and tailoring enzymes are often introduced into heterologous microbial hosts, which frequently lack favorable intracellular conditions for the functional expression of these complex biochemical machineries. For example, as megasynthases such as type I PKSs are not always efficiently and solubly expressed in heterologous hosts due to their large sizes, the efficiency of expression of PKS-coding genes as well as the solubility of PKSs greatly affects the efficiency of polyketide biosynthesis. Also, when different modules within PKSs are recombined and engineered, protein folding and thus solubility can be affected, leading to alteration in the efficiency of polyketide biosynthesis. Therefore, monitoring the solubility and functionality of the engineered PKSs is important. In this regard, a biosensor that can monitor the abundance of misfolded and aggregated proteins was developed by employing promoters of ibpA and fxs that are observed to be highly expressed upon accumulation of misfolded proteins [224]. By the expression of the mCherry gene encoding a fluorescence protein under the ibpA promoter, the solubility of engineered PKSs could be easily monitored. Using the biosensor, a type I modular PKS harboring a hybrid acyltransferase domain showing high solubility and activity could be selected. The combined engineering of PKSs and tailoring enzymes could lead to the generation of a much larger number of new antibiotic candidates, potentially contributing to the development of new antibiotics with no existing resistance.

Semi-synthesis

While synthetic biology and enzyme engineering have been effective for producing diverse antibiotics and their derivatives, the range of reactions enzymes can catalyze still falls short of chemical synthesis. In this regard, semi-synthesis—combining biosynthesis with chemical reactions—can be employed. Semi-synthesis has been successful in developing new antibiotics from previously discovered classical antibiotics. Following the development of methicillin—the first semi-synthetic antibiotic derived from penicillin to combat β-lactamase-producing, penicillin-resistant bacteria—additional semi-synthetic antibiotics, such as ampicillin, amoxicillin, azithromycin, and tigecycline were subsequently developed [225]. One notable example is amoxicillin (a derivative of penicillin G), which is synthesized by attaching p-hydroxyphenylglycine to the amino group of 6-aminopenicillanic acid, the core structure of penicillin. This modification enhances the antibiotic's activity by inhibiting essential enzymes involved in the cross-linking of bacterial cell walls, resulting in a broader spectrum of antibiotic effects compared to penicillin [226]. Additionally, the production of arylomycin derivatives serves as another representative example. To diversify arylomycins, three moieties attached on the macrocyclic tripeptide core, an N-terminal lipopeptide tail, a C-terminal carboxylic acid, and two phenol groups, were modified. One of the derivatives, G0775 showed significanatly improved antibiotic effects against 49 MDR clinical strains of E. coli and K. pneumoniae, 16 MDR A. baumannii strains, 12 MDR P. aeruginosa strains, as well as methicillin-resistant S. aureus and S. epidermidis [227]. Another example is the production of derivatives of chelocardin, an atypical tetracycline produced by Amycolatopsis sulphurea [228]. Chemical modifications of amidochelocardin (2-carboxamid-2-deacetyl-chelocardin) through methylation, acylation, and halogenation resulted in the production of 22 different derivatives. Notably, fluorination at the C7 position led to the significant enhancement of antimicrobial activity (Fig. 3) [228].

Fig. 3
figure 3

Semi-synthesis for further diversification of antibiotics. Biosynthesis of atypical tetracycline, amidochelocardin, through metabolic engineering. Chemical modifications of amidochelocardin enable the production of diverse derivatives, some of which may exhibit enhanced antimicrobial properties

Chemical modification can also lead to the enhancement of pharmacokinetic properties. For example, natamycin is an antibiotic with low toxicity, but its low bioavailability and solubility make it unsuitable for therapeutic use. To improve its solubility, natamycin was modified by attaching various diamines, among which the derivative containing an ethylenediamine moiety showed approximately a ten-fold increase in solubility and also two to eight-fold lower MIC [229]. Additionally, while natamycin lacks antibacterial effects at high concentrations, derivatives containing ethylenediamine or N-(2-fluorobenzyl) ethane-1,2-diamine moieties exhibited significant antibacterial effects.

Vancomycin has been one of the most effective antibiotics for treating complicated skin infections, bloodstream infections, endocarditis, bone and joint infections, and meningitis caused by methicillin-resistant S. aureus (MRSA), one of the most deadly pathogens. However, the emergence of vancomycin resistance has necessitated the development of vancomycin derivatives. To address this, a guanidinium motif was introduced at the vancosamine site in vancomycin, leading to the production of the lipoglycopeptide EVG7, which demonstrated superior antimicrobial activity compared to vancomycin [230]. In addition, a biguanide-vancomycin conjugate, V − C6 − Bg-PhCl, showed superior antibacterial activities towards mycobacteria and the ESKAPE pathogens [231]. Derivatization of caprazamycin is another representative example of semi-synthetic antibiotics. Caprazamycin is an antibiotic derived from Streptomyces sp. MK 730-62F2, which is effective against several mycobacterial species as well as gram-positive and gram-negative bacteria. Acidic treatment of caprazamycins A-G led to the production of caprazene, which was further modified by adding alkylamide, anilide, and ester functional groups. Notably, while caprazene itself lost antibacterial activity, its derivatives restored and even showed higher antibacterial activities than the original caprazamycins [232]. Such examples demonstrate that diversification of known antibiotics to produce a series of non-natural antibiotics can be an effective strategy for combating MDR bacteria. For more in-depth studies on semi-synthetic antibiotics, readers are guided to the following literature [233, 234].

Conclusion

With the increasing threat posed by the emergence of MDR bacteria, the need for the discovery and development of novel antibiotics has grown. Although the rate of new antibiotic discovery had significantly slowed down after the ‘golden era of antibiotic discovery’ during 1940s ~ 1960s, new tools and strategies for the discovery and development of new antibiotics have continued to be reported in various fields, including biology, bioinformatics, synthetic biology, metabolic engineering, systems biology, and synthetic chemistry. As these tools and strategies have become more widely available, the discovery of new antibiotics directly from nature and the prediction of BGCs capable of producing new antibiotics through metagenome sequencing have become more common. As discussed in the above sections, culturing “unculturable” microorganisms for the discovery of new active compounds has greatly expanded the accessible chemical space in nature. Advances in single-cell sequencing technologies and genome mining tools have also allowed exploration of vast BGCs, which undoubtly harbor numerous BGCs capable of producing novel antibiotics with unprecendented properties [235]. Capturing such BGCs and introducing them into heterologous chassis microorganisms for producing them with large quantities has been made possible by advancements in synthetic biology strategies, including large gene cluster assembly, gene expression control, and extensive libraries of standardized biological parts. Activating cryptic BGCs in native producers has also proven useful for the discovery of new antibiotics. Such advancements have greatly facilitated the mining of “microbial dark matters” from nature, which can be leveraged as antibiotics or serve as lead compounds for developing antibiotics [236].

Also, high performance bacterial strains capable of efficient production of antibiotics and their non-natural derivatives have been developed using synthetic biology, facilitating the translation of these new antibiotics to the clinic. State-of-the-art genome engineering tools, such as CRISPR, base editing, and prime editing, along with gene expression manipulation tools like CRISPRi and sRNA, have enabled the tailored construction of microbial cell factories capable of efficiently producing target antibiotics from renewable carbon sources in one-step bioprocesses [237]. With a defined target chemical and metabolic pathway, synthetic biology now enables significant increases in product titers. Improved understanding of metabolic and regulatory pathways in actinomycetes has also made it easier to manipulate these hosts, which are traditionally challenging to engineer but rich in secondary metabolites. Enzyme engineering has also played a crucial role in diversifying antibiotics to introduce new properties. Advanced enzyme and strain engineering strategies, such as automated directed evolution and machine learning-assisted metabolic engineering, will provide an expanded portfolio of reactions available for the synthesis of new antibiotics to combat MDR bacteria.

However, one notable challenge is that testing the efficacy of new antibiotics in animal models or clinical tests with human patients is difficult and time-consuming. This issue could potentially be addressed in the near future by employing ‘organ-on-a-chip’ technology, which mimics the environment of real organs within an animal or human. Using this technology will also allow for a deeper understanding of the MoA of new antibiotics and the physiological change in pathogens within the human body upon antibiotic treatment [238, 239]. Another challenge is that designing biosynthetic enzymes such as PKS or NRPS for the production of desired chemicals is still difficult. Recent advances in protein structure prediction and protein design models, such as AlphaFold3 [240], will eventually allow the creation of designable antibiotics using high-performance enzymes specifically designed to produce the target chemicals with desired chemical structures. The development of machine learning will not only facilitate the discovery of new BGCs from vast metagenomic data, but also allow for the design of antibiotics with specific chemical structures that can precisely interact with the target molecules within pathogens. Such designer antibiotics could be a game-changer in our fight against pathogenic bacteria. Recent applications of deep learning for the prediction of the antibiotic activity and cytotoxicity of millions of chemicals [241] showcase the potential of artificial intelligence (AI) in screening large libraries of antibiotic candidates [242, 243]. Other treatment options for combating MDR bacteria, such as antisense oligonucleotides, antimicrobial peptides, and microbiota-based therapeutics, are also being actively studied [244,245,246]. Continued efforts in developing innovative metabolic engineering and synthetic biology tools and strategies, combined with interdisciplinary collaboration in areas such as in silico enzyme modeling and semi-synthesis, will strengthen humanity's fight against infectious diseases.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

2D:

Two-dimensional

6-dEB:

6-Deoxyerythronolide

ABC:

ATP-binding-cassette

AI:

Artificial intelligence

AP-3:

Ansamitocin P-3

Att/Int:

Attachment and integration

BCP:

Bacterial cytological profiling

BGCs:

Biosynthetic gene clusters

CRISPR-BEST:

CRISPR-base editing system

CRISPRi:

Clustered regularly interspaced short palindromic repeats interference

DEBS:

Dexoyerythronolide B synthase

EtBr:

Ethidium bromide

HGT:

Horizontal gene transfer

iChip:

Isolation chip

LAL:

Large ATP-binding regulator of the LuxR family

LC–MS:

Liquid chromatography-mass spectrometry

MALDI-TOF/MS:

Matrix-assisted laser desorption-ionization time-of-flight mass spectrometry

MDR:

Multidrug-resistant

MoA:

Mechanism of action

MRSA:

Methicillin-resistant S. aureus

NGS:

Next-generation sequencing

NRP:

Non-ribosomal peptide

OTUs:

Operational taxonomic units

PAINS:

Pan-assay interference compounds

PAM:

Protospacer adjacent motif

PKS:

Polyketide synthase

sgRNAs:

Single guide RNAs

TAG:

Triacylglycerol

TAL:

Triacetic acid lactone

TESEC:

Target essential surrogate E. coli

References

  1. Adedeji WA. The treasure called antibiotics. Ann Ib Postgrad Med. 2016;14:56–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. da Ribeiro Cunha B, Fonseca LP, Calado CRC. Antibiotic discovery: where have we come from, where do we go? Antibiotics. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antibiotics8020045.

    Article  Google Scholar 

  3. Smith PW, Watkins K, Hewlett A. Infection control through the ages. Am J Infect Control. 2012;40:35–42.

    Article  PubMed  Google Scholar 

  4. de Souza MC, de Souza Antunes AM. Pipeline of known chemical classes of antibiotics. Antibiotics. 2013;2:500–34.

    Article  Google Scholar 

  5. Frei A, Zuegg J, Elliott AG, Baker M, Braese S, Brown C, Chen F, C GD, Dujardin G, Jung N, et al. Metal complexes as a promising source for new antibiotics. Chem Sci. 2020;11:2627–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Correia A, Weimann A. Protein antibiotics: mind your language. Nat Rev Microbiol. 2021;19:7.

    Article  CAS  PubMed  Google Scholar 

  7. Frei A, Verderosa AD, Elliott AG, Zuegg J, Blaskovich MAT. Metals to combat antimicrobial resistance. Nat Rev Chem. 2023;7:202–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. O’Rourke A, Beyhan S, Choi Y, Morales P, Chan AP, Espinoza JL, Dupont CL, Meyer KJ, Spoering A, Lewis K, et al. Mechanism-of-action classification of antibiotics by global transcriptome profiling. Antimicrob Agents Chemother. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/AAC.01207-19.

    Article  PubMed  PubMed Central  Google Scholar 

  9. About Antimicrobial Resistance. 2024.

  10. Keeling PJ, Palmer JD. Horizontal gene transfer in eukaryotic evolution. Nat Rev Genet. 2008;9:605–18.

    Article  CAS  PubMed  Google Scholar 

  11. Lewis K. The science of antibiotic discovery. Cell. 2020;181:29–45.

    Article  CAS  PubMed  Google Scholar 

  12. Hutchings MI, Truman AW, Wilkinson B. Antibiotics: past, present and future. Curr Opin Microbiol. 2019;51:72–80.

    Article  CAS  PubMed  Google Scholar 

  13. Mohr KI. History of antibiotics research. Curr Top Microbiol Immunol. 2016;398:237–72.

    CAS  PubMed  Google Scholar 

  14. Katz L, Baltz RH. Natural product discovery: past, present, and future. J Ind Microbiol Biotechnol. 2016;43:155–76.

    Article  CAS  PubMed  Google Scholar 

  15. Miethke M, Pieroni M, Weber T, Brönstrup M, Hammann P, Halby L, Arimondo PB, Glaser P, Aigle B, Bode HB, et al. Towards the sustainable discovery and development of new antibiotics. Nat Rev Chem. 2021;5:726–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Paulsel TQ, Williams GJ. Current state-of-the-art toward chemoenzymatic synthesis of polyketide natural products. ChemBioChem. 2023;24:e202300386.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rule AM. American society of health-system pharmacists’ pain management network. J Pain Palliat Care Pharmacother. 2004;18:59–62.

    Article  PubMed  Google Scholar 

  18. Quiros Y, Vicente-Vicente L, Morales AI, López-Novoa JM, López-Hernández FJ. An integrative overview on the mechanisms underlying the renal tubular cytotoxicity of gentamicin. Toxicol Sci. 2011;119:245–56.

    Article  CAS  PubMed  Google Scholar 

  19. Pancu DF, Scurtu A, Macasoi IG, Marti D, Mioc M, Soica C, Coricovac D, Horhat D, Poenaru M, Dehelean C. Antibiotics: conventional therapy and natural compounds with antibacterial activity-a pharmaco-toxicological screening. Antibiotics. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antibiotics10040401.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Edson RS, Keys TF. The aminoglycosides. Streptomycin, kanamycin, gentamicin, tobramycin, amikacin, netilmicin, sisomicin. Mayo Clin Proc. 1983;58:99–102.

    CAS  PubMed  Google Scholar 

  21. Haupt I, Hübener R, Thrum H. Streptothricin F, an inhibitor of protein synthesis with miscoding activity. J Antibiot. 1978;31:1137–42.

    Article  CAS  Google Scholar 

  22. Brogden RN, Pinder RM, Sawyer PR, Speight TM, Avery GS. Tobramycin: a review of its antibacterial and pharmacokinetic properties and therapeutic use. Drugs. 1976;12:166–200.

    Article  CAS  PubMed  Google Scholar 

  23. Papp-Wallace KM, Endimiani A, Taracila MA, Bonomo RA. Carbapenems: past, present, and future. Antimicrob Agents Chemother. 2011;55:4943–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. López-Argüello S, Montaner M, Sayed AR, Oliver A, Bulitta JB, Moya B. Penicillin-binding protein 5/6 acting as a decoy target in Pseudomonas aeruginosa identified by whole-cell receptor binding and quantitative systems pharmacology. Antimicrob Agents Chemother. 2023;67:e0160322.

    Article  PubMed  Google Scholar 

  25. Feigin RD: Feigin & Cherry's textbook of pediatric infectious diseases. 6th edn. Philadelphia, PA: Saunders/Elsevier; 2009.

  26. Cefadroxil monohydrate [https://pubchem.ncbi.nlm.nih.gov/compound/Cefadroxil-monohydrate]

  27. Atherton FR, Hall MJ, Hassall CH, Holmes SW, Lambert RW, Lloyd WJ, Nisbet LJ, Ringrose PS, Westmacott D. Antibacterial properties of alafosfalin combined with cephalexin. Antimicrob Agents Chemother. 1981;20:470–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Duplessis C, Crum-Cianflone NF. Ceftaroline: a new cephalosporin with activity against methicillin-resistant Staphylococcus aureus (MRSA). Clin Med Rev Ther. 2011. https://doiorg.publicaciones.saludcastillayleon.es/10.4137/CMRT.S1637.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Pucci MJ, Boice-Sowek J, Kessler RE, Dougherty TJ. Comparison of cefepime, cefpirome, and cefaclidine binding affinities for penicillin-binding proteins in Escherichia coli K-12 and Pseudomonas aeruginosa SC8329. Antimicrob Agents Chemother. 1991;35:2312–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Weiner CP, Buhimschi C. Drugs for pregnant and lactating women. 2nd ed. Philadelphia: Saunders/Elsevier; 2009.

    Google Scholar 

  31. Alotaibi AF, Mekary RA, Zaidi HA, Smith TR, Pandya A. Safety and efficacy of antibacterial prophylaxis after craniotomy: a decision model analysis. World Neurosurg. 2017;105(906–912):e905.

    Google Scholar 

  32. Fontana R, Aldegheri M, Ligozzi M, Lo Cascio G, Cornaglia G. Interaction of ceftriaxone with penicillin-binding proteins of Escherichia coli in the presence of human serum albumin. J Antimicrob Chemother. 1998;42:95–8.

    Article  CAS  PubMed  Google Scholar 

  33. Nolan RD, Jude DA. The interactions of [14C]cefotetan with penicillin binding proteins of a wide variety of gram-positive and gram-negative species. J Antimicrob Chemother. 1983;11(Suppl):169–77.

    Article  CAS  PubMed  Google Scholar 

  34. Ojkic N, Lilja E, Direito S, Dawson A, Allen RJ, Waclaw B. A roadblock-and-kill mechanism of action model for the DNA-targeting antibiotic ciprofloxacin. Antimicrob Agents Chemother. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/aac.02487-19.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Candel FJ, Peñuelas M. Delafloxacin: design, development and potential place in therapy. Drug Des Devel Ther. 2017;11:881–91.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bhavnani SM, Andes DR. Gemifloxacin for the treatment of respiratory tract infections: in vitro susceptibility, pharmacokinetics and pharmacodynamics, clinical efficacy, and safety. Pharmacotherapy. 2005;25:717–40.

    Article  CAS  PubMed  Google Scholar 

  37. Tran T, Bowman-Carpio L, Buscher N, Ford JJ, Jenkins E, Kalay HN, Nakazono T, Orescan H, Sak R, Shin I, Davidson P. Collaboration in action: measuring and improving contracting performance in the university of California contracting network. Res Manag Rev. 2017;22:28–41.

    PubMed  PubMed Central  Google Scholar 

  38. Mayers DL, Kaye KS, Marchaim D, Ouellette M, Sobel JD. Antimicrobial drug resistance: mechanisms of drug resistance. Cham: Springer International Publishing; 2017.

    Book  Google Scholar 

  39. Watanakunakorn C. Mode of action and in-vitro activity of vancomycin. J Antimicrob Chemother. 1984;14:7–18.

    Article  CAS  PubMed  Google Scholar 

  40. Walker S, Chen L, Hu Y, Rew Y, Shin D, Boger DL. Chemistry and biology of ramoplanin: a lipoglycodepsipeptide with potent antibiotic activity. Chem Rev. 2005;105:449–76.

    Article  CAS  PubMed  Google Scholar 

  41. Marcone GL, Binda E, Reguzzoni M, Gastaldo L, Dalmastri C, Marinelli F. Classification of Actinoplanes sp. ATCC 33076, an actinomycete that produces the glycolipodepsipeptide antibiotic ramoplanin, as Actinoplanes ramoplaninifer sp. nov. Int J Syst Evol Microbiol. 2017;67:4181–8.

    Article  CAS  PubMed  Google Scholar 

  42. Andrade FF, Silva D, Rodrigues A, Pina-Vaz C. Colistin update on its mechanism of action and resistance, present and future challenges. Microorganisms. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/microorganisms8111716.

    Article  PubMed  PubMed Central  Google Scholar 

  43. David JM, Rajasekaran AK. Gramicidin a: a new mission for an old antibiotic. J Kidney Cancer VHL. 2015;2:15–24.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ciesiołka J, Jeżowska-Bojczuk M, Wrzesiński J, Stokowa-Sołtys K, Nagaj J, Kasprowicz A, Błaszczyk L, Szczepanik W. Antibiotic bacitracin induces hydrolytic degradation of nucleic acids. Biochim Biophys Acta. 2014;1840:1782–9.

    Article  PubMed  Google Scholar 

  45. Zheng Q, Wang Q, Wang S, Wu J, Gao Q, Liu W. Thiopeptide antibiotics exhibit a dual mode of action against intracellular pathogens by affecting both host and microbe. Chem Biol. 2015;22:1002–7.

    Article  CAS  PubMed  Google Scholar 

  46. de Crécy-Lagard V, Blanc V, Gil P, Naudin L, Lorenzon S, Famechon A, Bamas-Jacques N, Crouzet J, Thibaut D. Pristinamycin I biosynthesis in Streptomyces pristinaespiralis: molecular characterization of the first two structural peptide synthetase genes. J Bacteriol. 1997;179:705–13.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Artsimovitch I, Seddon J, Sears P. Fidaxomicin is an inhibitor of the initiation of bacterial RNA synthesis. Clin Infect Dis. 2012;55(Suppl 2):S127-131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Thomson AW, Bonham CA, Zeevi A. Mode of action of tacrolimus (FK506): molecular and cellular mechanisms. Ther Drug Monit. 1995;17:584–91.

    Article  CAS  PubMed  Google Scholar 

  49. Liu M, Douthwaite S. Resistance to the macrolide antibiotic tylosin is conferred by single methylations at 23S rRNA nucleotides G748 and A2058 acting in synergy. Proc Natl Acad Sci USA. 2002;99:14658–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Dos Santos AG, Marquês JT, Carreira AC, Castro IR, Viana AS, Mingeot-Leclercq MP, de Almeida RFM, Silva LC. The molecular mechanism of Nystatin action is dependent on the membrane biophysical properties and lipid composition. Phys Chem Chem Phys. 2017;19:30078–88.

    Article  PubMed  Google Scholar 

  51. Kim YH, Park BS, Bhatia SK, Seo HM, Jeon JM, Kim HJ, Yi DH, Lee JH, Choi KY, Park HY, et al. Production of rapamycin in Streptomyces hygroscopicus from glycerol-based media optimized by systemic methodology. J Microbiol Biotechnol. 2014;24:1319–26.

    Article  CAS  PubMed  Google Scholar 

  52. Menninger JR, Otto DP. Erythromycin, carbomycin, and spiramycin inhibit protein synthesis by stimulating the dissociation of peptidyl-tRNA from ribosomes. Antimicrob Agents Chemother. 1982;21:811–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lovmar M, Tenson T, Ehrenberg M. Kinetics of macrolide action: the josamycin and erythromycin cases. J Biol Chem. 2004;279:53506–15.

    Article  CAS  PubMed  Google Scholar 

  54. Usary J, Champney WS. Erythromycin inhibition of 50S ribosomal subunit formation in Escherichia coli cells. Mol Microbiol. 2001;40:951–62.

    Article  CAS  PubMed  Google Scholar 

  55. Lü J, Long Q, Zhao Z, Chen L, He W, Hong J, Liu K, Wang Y, Pang X, Deng Z, Tao M. Engineering the erythromycin-producing strain Saccharopolyspora erythraea HOE107 for the heterologous production of polyketide antibiotics. Front Microbiol. 2020;11:593217.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Heidary M, Ebrahimi Samangani A, Kargari A, Kiani Nejad A, Yashmi I, Motahar M, Taki E, Khoshnood S. Mechanism of action, resistance, synergism, and clinical implications of azithromycin. J Clin Lab Anal. 2022;36:e24427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Champney WS, Burdine R. Azithromycin and clarithromycin inhibition of 50S ribosomal subunit formation in Staphylococcus aureus cells. Curr Microbiol. 1998;36:119–23.

    Article  CAS  PubMed  Google Scholar 

  58. Parnham MJ, Erakovic Haber V, Giamarellos-Bourboulis EJ, Perletti G, Verleden GM, Vos R. Azithromycin: mechanisms of action and their relevance for clinical applications. Pharmacol Ther. 2014;143:225–45.

    Article  CAS  PubMed  Google Scholar 

  59. Symersky J, Osowski D, Walters DE, Mueller DM. Oligomycin frames a common drug-binding site in the ATP synthase. Proc Natl Acad Sci USA. 2012;109:13961–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ghooi RB, Thatte SM. Inhibition of cell wall synthesis–is this the mechanism of action of penicillins? Med Hypotheses. 1995;44:127–31.

    Article  CAS  PubMed  Google Scholar 

  61. Hill P. The production of penicillins in soils and seeds by penicillium chrysogenum and the role of penicillin -lactamase in the ecology of soil bacillus. J Gen Microbiol. 1972;70:243–52.

    Article  CAS  PubMed  Google Scholar 

  62. Zhou J, Cai Y, Liu Y, An H, Deng K, Ashraf MA, Zou L, Wang J. Breaking down the cell wall: Still an attractive antibacterial strategy. Front Microbiol. 2022;13:952633.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Teethaisong Y, Autarkool N, Sirichaiwetchakoon K, Krubphachaya P, Kupittayanant S, Eumkeb G. Synergistic activity and mechanism of action of Stephania suberosa forman extract and ampicillin combination against ampicillin-resistant Staphylococcus aureus. J Biomed Sci. 2014;21:90.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Maiti SN, Phillips OA, Micetich RG, Livermore DM. Beta-lactamase inhibitors: agents to overcome bacterial resistance. Curr Med Chem. 1998;5:441–56.

    Article  CAS  PubMed  Google Scholar 

  65. Mizoguchi J, Suginaka H, Kotani S. Mechanism of synergistic action of a combination of ampicillin and dicloxacillin against a beta-lactamase-producing strain of Citrobacter freundii. Antimicrob Agents Chemother. 1979;16:439–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lima LM, Silva B, Barbosa G, Barreiro EJ. beta-lactam antibiotics: an overview from a medicinal chemistry perspective. Eur J Med Chem. 2020;208:112829.

    Article  CAS  PubMed  Google Scholar 

  67. Manzella JP. Quinupristin-dalfopristin: a new antibiotic for severe gram-positive infections. Am Fam Physician. 2001;64:1863–6.

    CAS  PubMed  Google Scholar 

  68. Tenson T, Lovmar M, Ehrenberg M. The mechanism of action of macrolides, lincosamides and streptogramin B reveals the nascent peptide exit path in the ribosome. J Mol Biol. 2003;330:1005–14.

    Article  CAS  PubMed  Google Scholar 

  69. Thrope BM. Antibacterial activity and post-antibiotic effect of protein synthesis inhibitors. Brook: State University of New York at Stony Brook; 2024.

    Google Scholar 

  70. Chukwudi CU. rRNA binding sites and the molecular mechanism of action of the tetracyclines. Antimicrob Agents Chemother. 2016;60:4433–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Henehan M, Montuno M, De Benedetto A. Doxycycline as an anti-inflammatory agent: updates in dermatology. J Eur Acad Dermatol Venereol. 2017;31:1800–8.

    Article  CAS  PubMed  Google Scholar 

  72. Zhanel GG, Cheung D, Adam H, Zelenitsky S, Golden A, Schweizer F, Gorityala B, Lagacé-Wiens PR, Walkty A, Gin AS, et al. Review of eravacycline, a novel fluorocycline antibacterial agent. Drugs. 2016;76:567–88.

    Article  CAS  PubMed  Google Scholar 

  73. Sarkar P, Yarlagadda V, Ghosh C, Haldar J. A review on cell wall synthesis inhibitors with an emphasis on glycopeptide antibiotics. Medchemcomm. 2017;8:516–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Westly JW. Polyether antibiotics—volume 2: chemistry. 1st ed. Boca Raton: CRC Press; 2019.

    Google Scholar 

  75. Sinzato YK, Gelaleti RB, Volpato GT, Rudge MVC, Herrera E, Damasceno DC. Streptozotocin-induced leukocyte DNA damage in rats. Drug Chem Toxicol. 2020;43:165–8.

    Article  CAS  PubMed  Google Scholar 

  76. Borovinskaya MA, Shoji S, Holton JM, Fredrick K, Cate JHD. A steric block in translation caused by the antibiotic spectinomycin. ACS Chem Biol. 2007;2:545–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Tefas LR, Barbălată C, Tefas C, Tomuță I. Salinomycin-based drug delivery systems: overcoming the hurdles in cancer therapy. Pharmaceutics. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/pharmaceutics13081120.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Uyaguari-Diaz MI, Croxen MA, Luo Z, Cronin KI, Chan M, Baticados WN, Nesbitt MJ, Li S, Miller KM, Dooley D, et al. Human activity determines the presence of integron-associated and antibiotic resistance genes in southwestern British Columbia. Front Microbiol. 2018;9:852.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Spízek J, Rezanka T. Lincomycin, clindamycin and their applications. Appl Microbiol Biotechnol. 2004;64:455–64.

    Article  PubMed  Google Scholar 

  80. AlRabiah H, Allwood JW, Correa E, Xu Y, Goodacre R. pH plays a role in the mode of action of trimethoprim on Escherichia coli. PLoS ONE. 2018;13:e0200272.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Sarathy JP, Gruber G, Dick T. Re-understanding the mechanisms of action of the anti-mycobacterial drug bedaquiline. Antibiotics. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antibiotics8040261.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Levy D, Bluzat A, Seigneuret M, Rigaud JL. Alkali cation transport through liposomes by the antimicrobial fusafungine and its constitutive enniatins. Biochem Pharmacol. 1995;50:2105–7.

    Article  CAS  PubMed  Google Scholar 

  83. Ghiraldi-Lopes LD, Campanerut-Sá PAZ, Evaristo GPC, Meneguello JE, Fiorini A, Baldin VP, de Souza EM, de Lima Scodro RB, Siqueira VLD, Cardoso RF. New insights on ethambutol targets in Mycobacterium tuberculosis. Infect Disord Drug Targets. 2019;19:73–80.

    Article  CAS  PubMed  Google Scholar 

  84. Fernandes P. Fusidic acid: a bacterial elongation factor inhibitor for the oral treatment of acute and chronic staphylococcal infections. Cold Spring Harb Perspect Med. 2016;6:a025437.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Nguyen C, Zhou A, Khan A, Miller JH, Yeh P. Pairwise antibiotic interactions in Escherichia coli: triclosan, rifampicin and aztreonam with nine other classes of antibiotics. J Antibiot. 2016;69:791–7.

    Article  CAS  Google Scholar 

  86. McOsker CC, Fitzpatrick PM. Nitrofurantoin: mechanism of action and implications for resistance development in common uropathogens. J Antimicrob Chemother. 1994;33:23–30.

    Article  CAS  PubMed  Google Scholar 

  87. Yoon D, Moon JH, Cho A, Boo H, Cha JS, Lee Y, Yoo J. Structure-based insight on the mechanism of N-glycosylation inhibition by tunicamycin. Mol Cells. 2023;46:337–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Cholo MC, Mothiba MT, Fourie B, Anderson R. Mechanisms of action and therapeutic efficacies of the lipophilic antimycobacterial agents clofazimine and bedaquiline. J Antimicrob Chemother. 2017;72:338–53.

    Article  CAS  PubMed  Google Scholar 

  89. Silver LL. Fosfomycin: mechanism and resistance. Cold Spring Harb Perspect Med. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a025262.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Tang YZ, Liu YH, Chen JX. Pleuromutilin and its derivatives-the lead compounds for novel antibiotics. Mini Rev Med Chem. 2012;12:53–61.

    Article  CAS  PubMed  Google Scholar 

  91. Timmins GS, Deretic V. Mechanisms of action of isoniazid. Mol Microbiol. 2006;62:1220–7.

    Article  CAS  PubMed  Google Scholar 

  92. Adams RA, Leon G, Miller NM, Reyes SP, Thantrong CH, Thokkadam AM, Lemma AS, Sivaloganathan DM, Wan X, Brynildsen MP. Rifamycin antibiotics and the mechanisms of their failure. J Antibiot. 2021;74:786–98.

    Article  CAS  Google Scholar 

  93. Chrebet GL, Wisniewski D, Perkins AL, Deng Q, Kurtz MB, Marcy A, Parent SA. Cell-based assays to detect inhibitors of fungal mRNA capping enzymes and characterization of sinefungin as a cap methyltransferase inhibitor. J Biomol Screen. 2005;10:355–64.

    Article  CAS  PubMed  Google Scholar 

  94. Gelmetti C. Local antibiotics in dermatology. Dermatol Ther. 2008;21:187–95.

    Article  PubMed  Google Scholar 

  95. Wozel G, Blasum C. Dapsone in dermatology and beyond. Arch Dermatol Res. 2014;306:103–24.

    Article  CAS  PubMed  Google Scholar 

  96. Wang F, Langley R, Gulten G, Dover LG, Besra GS, Jacobs WR Jr, Sacchettini JC. Mechanism of thioamide drug action against tuberculosis and leprosy. J Exp Med. 2007;204:73–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wolf H, Chinali G, Parmeggiani A. Kirromycin, an inhibitor of protein biosynthesis that acts on elongation factor Tu. Proc Natl Acad Sci USA. 1974;71:4910–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Arciszewska K, Pućkowska A, Wróbel A, Drozdowska D. Carbocyclic analogues of distamycin and netropsin. Mini Rev Med Chem. 2019;19:98–113.

    Article  CAS  PubMed  Google Scholar 

  99. Wu Y, Zhang M, Yang Y, Ding X, Yang P, Huang K, Hu X, Zhang M, Liu X, Yu H. Structures and mechanism of chitin synthase and its inhibition by antifungal drug Nikkomycin Z. Cell Discov. 2022;8:129.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Osada H. Discovery and applications of nucleoside antibiotics beyond polyoxin. J Antibiot. 2019;72:855–64.

    Article  CAS  Google Scholar 

  101. Miller SC, Huang R, Sakamuru S, Shukla SJ, Attene-Ramos MS, Shinn P, Van Leer D, Leister W, Austin CP, Xia M. Identification of known drugs that act as inhibitors of NF-kappaB signaling and their mechanism of action. Biochem Pharmacol. 2010;79:1272–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Khoshnood S, Heidary M, Asadi A, Soleimani S, Motahar M, Savari M, Saki M, Abdi M. A review on mechanism of action, resistance, synergism, and clinical implications of mupirocin against Staphylococcus aureus. Biomed Pharmacother. 2019;109:1809–18.

    Article  CAS  PubMed  Google Scholar 

  103. Giri B, Giang PH, Kumari R, Prasad R, Varma A. Microbial diversity in soils. In: Varma A, Buscot F, editors. Microorganisms in soils: roles in genesis and functions. Berlin: Springer; 2005. p. 19–55.

    Chapter  Google Scholar 

  104. Roesch LF, Fulthorpe RR, Riva A, Casella G, Hadwin AK, Kent AD, Daroub SH, Camargo FA, Farmerie WG, Triplett EW. Pyrosequencing enumerates and contrasts soil microbial diversity. ISME J. 2007;1:283–90.

    Article  CAS  PubMed  Google Scholar 

  105. Davies J. What are antibiotics? Archaic functions for modern activities. Mol Microbiol. 1990;4:1227–32.

    Article  CAS  PubMed  Google Scholar 

  106. Genilloud O. Actinomycetes: still a source of novel antibiotics. Nat Prod Rep. 2017;34:1203–32.

    Article  CAS  PubMed  Google Scholar 

  107. Ludwig P, Holzhutter H, Colosimo A. Modelling the reaction mechanism of the reticulocyte lipoxygenase. Biomed Biochim Acta. 1987;46:S241-244.

    CAS  PubMed  Google Scholar 

  108. Bentley SD, Chater KF, Cerdeño-Tárraga AM, Challis GL, Thomson NR, James KD, Harris DE, Quail MA, Kieser H, Harper D, et al. Complete genome sequence of the model actinomycete Streptomyces coelicolor A3(2). Nature. 2002;417:141–7.

    Article  PubMed  Google Scholar 

  109. Maiti PK, Das S, Sahoo P, Mandal S. Streptomyces sp SM01 isolated from Indian soil produces a novel antibiotic picolinamycin effective against multi drug resistant bacterial strains. Sci Rep. 2020;10:10092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Palazzotto E, Tong Y, Lee SY, Weber T. Synthetic biology and metabolic engineering of actinomycetes for natural product discovery. Biotechnol Adv. 2019;37:107366.

    Article  CAS  PubMed  Google Scholar 

  111. Nepal KK, Wang G. Streptomycetes: surrogate hosts for the genetic manipulation of biosynthetic gene clusters and production of natural products. Biotechnol Adv. 2019;37:1–20.

    Article  CAS  PubMed  Google Scholar 

  112. Bu QT, Yu P, Wang J, Li ZY, Chen XA, Mao XM, Li YQ. Rational construction of genome-reduced and high-efficient industrial Streptomyces chassis based on multiple comparative genomic approaches. Microb Cell Fact. 2019;18:16.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Hwang S, Lee Y, Kim JH, Kim G, Kim H, Kim W, Cho S, Palsson BO, Cho BK. Streptomyces as microbial chassis for heterologous protein expression. Front Bioeng Biotechnol. 2021;9:804295.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Zaburannyi N, Rabyk M, Ostash B, Fedorenko V, Luzhetskyy A. Insights into naturally minimised Streptomyces albus J1074 genome. BMC Genomics. 2014;15:97.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Zhang K, Mohsin A, Dai Y, Chen Z, Zhuang Y, Chu J, Guo M. Combinatorial effect of ARTP mutagenesis and ribosome engineering on an industrial strain of Streptomyces albus S12 for enhanced biosynthesis of salinomycin. Front Bioeng Biotechnol. 2019;7:212.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Hu YL, Zhang Q, Liu SH, Sun JL, Yin FZ, Wang ZR, Shi J, Jiao RH, Ge HM. Building Streptomyces albus as a chassis for synthesis of bacterial terpenoids. Chem Sci. 2023;14:3661–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Myronovskyi M, Rosenkränzer B, Nadmid S, Pujic P, Normand P, Luzhetskyy A. Generation of a cluster-free Streptomyces albus chassis strains for improved heterologous expression of secondary metabolite clusters. Metab Eng. 2018;49:316–24.

    Article  CAS  PubMed  Google Scholar 

  118. Gomez-Escribano JP, Bibb MJ. Engineering Streptomyces coelicolor for heterologous expression of secondary metabolite gene clusters. Microb Biotechnol. 2011;4:207–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Pham VH, Kim J. Cultivation of unculturable soil bacteria. Trends Biotechnol. 2012;30:475–84.

    Article  CAS  PubMed  Google Scholar 

  120. Moffat JG, Rudolph J, Bailey D. Phenotypic screening in cancer drug discovery—past, present and future. Nat Rev Drug Discov. 2014;13:588–602.

    Article  CAS  PubMed  Google Scholar 

  121. Terstappen GC, Schlüpen C, Raggiaschi R, Gaviraghi G. Target deconvolution strategies in drug discovery. Nat Rev Drug Discov. 2007;6:891–903.

    Article  CAS  PubMed  Google Scholar 

  122. Zipperer A, Konnerth MC, Laux C, Berscheid A, Janek D, Weidenmaier C, Burian M, Schilling NA, Slavetinsky C, Marschal M, et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature. 2016;535:511–6.

    Article  CAS  PubMed  Google Scholar 

  123. DeVito JA, Mills JA, Liu VG, Agarwal A, Sizemore CF, Yao Z, Stoughton DM, Cappiello MG, Barbosa MD, Foster LA, Pompliano DL. An array of target-specific screening strains for antibacterial discovery. Nat Biotechnol. 2002;20:478–83.

    Article  CAS  PubMed  Google Scholar 

  124. Bongaerts N, Edoo Z, Abukar AA, Song X, Sosa-Carrillo S, Haggenmueller S, Savigny J, Gontier S, Lindner AB, Wintermute EH. Low-cost anti-mycobacterial drug discovery using engineered E. coli. Nat Commun. 2022;13:3905.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Nonejuie P, Burkart M, Pogliano K, Pogliano J. Bacterial cytological profiling rapidly identifies the cellular pathways targeted by antibacterial molecules. Proc Natl Acad Sci USA. 2013;110:16169–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Newton BA. Mechanisms of antibiotic action. Annu Rev Microbiol. 1965;19:209–40.

    Article  CAS  PubMed  Google Scholar 

  127. Nonejuie P, Trial RM, Newton GL, Lamsa A, Ranmali Perera V, Aguilar J, Liu WT, Dorrestein PC, Pogliano J, Pogliano K. Application of bacterial cytological profiling to crude natural product extracts reveals the antibacterial arsenal of Bacillus subtilis. J Antibiot. 2016;69:353–61.

    Article  CAS  Google Scholar 

  128. Araújo-Bazán L, Ruiz-Avila LB, Andreu D, Huecas S, Andreu JM. Cytological profile of antibacterial FtsZ inhibitors and synthetic peptide MciZ. Front Microbiol. 2016;7:1558.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Handler AA, Lim JE, Losick R. Peptide inhibitor of cytokinesis during sporulation in Bacillus subtilis. Mol Microbiol. 2008;68:588–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Montaño ET, Nideffer JF, Sugie J, Enustun E, Shapiro AB, Tsunemoto H, Derman AI, Pogliano K, Pogliano J. Bacterial cytological profiling identifies rhodanine-containing PAINS analogs as specific inhibitors of Escherichia coli thymidylate kinase In Vivo. J Bacteriol. 2021;203:e0010521.

    Article  PubMed  Google Scholar 

  131. Marutescu LG. Current and future flow cytometry applications contributing to antimicrobial resistance control. Microorganisms. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/microorganisms11051300.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Ghosh S, Indukuri K, Bondalapati S, Saikia AK, Rangan L. Unveiling the mode of action of antibacterial labdane diterpenes from Alpinia nigra (Gaertn.) B. L. Burtt seeds. Eur J Med Chem. 2013;66:101–5.

    Article  CAS  PubMed  Google Scholar 

  133. Liu J, Chen F, Wang X, Peng H, Zhang H, Wang KJ. The synergistic effect of mud crab antimicrobial peptides sphistin and Sph(12–38) with antibiotics azithromycin and rifampicin enhances bactericidal activity against Pseudomonas Aeruginosa. Front Cell Infect Microbiol. 2020;10:572849.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Larson MH, Gilbert LA, Wang X, Lim WA, Weissman JS, Qi LS. CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat Protoc. 2013;8:2180–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zhang H, Li X, Liu X, Ji X, Ma X, Chen J, Bao Y, Zhang Y, Xu L, Yang L, Wei X. The usnic acid derivative peziculone targets cell walls of gram-positive bacteria revealed by high-throughput CRISPRi-seq analysis. Int J Antimicrob Agents. 2023;62:106876.

    Article  CAS  PubMed  Google Scholar 

  136. Martin JK 2nd, Sheehan JP, Bratton BP, Moore GM, Mateus A, Li SH, Kim H, Rabinowitz JD, Typas A, Savitski MM, et al. A dual-mechanism antibiotic kills gram-negative bacteria and avoids drug resistance. Cell. 2020;181(1518–1532):e1514.

    Google Scholar 

  137. Tsakou F, Jersie-Christensen R, Jenssen H, Mojsoska B. The role of proteomics in bacterial response to antibiotics. Pharmaceuticals. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph13090214.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Pulido MR, García-Quintanilla M, Gil-Marques ML, McConnell MJ. Identifying targets for antibiotic development using omics technologies. Drug Discov Today. 2016;21:465–72.

    Article  CAS  PubMed  Google Scholar 

  139. Lin CH, Su SC, Ho KH, Hsu YW, Lee KR. Bactericidal effect of sulbactam against Acinetobacter baumannii ATCC 19606 studied by 2D-DIGE and mass spectrometry. Int J Antimicrob Agents. 2014;44:38–46.

    Article  CAS  PubMed  Google Scholar 

  140. Cao J, Zheng Y. iTRAQ-based quantitative proteomic analysis of the antimicrobial mechanism of lactobionic acid against Staphylococcus aureus. Food Funct. 2021;12:1349–60.

    Article  CAS  PubMed  Google Scholar 

  141. Stewart EJ. Growing unculturable bacteria. J Bacteriol. 2012;194:4151–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. D’Onofrio A, Crawford JM, Stewart EJ, Witt K, Gavrish E, Epstein S, Clardy J, Lewis K. Siderophores from neighboring organisms promote the growth of uncultured bacteria. Chem Biol. 2010;17:254–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Adnani N, Chevrette MG, Adibhatla SN, Zhang F, Yu Q, Braun DR, Nelson J, Simpkins SW, McDonald BR, Myers CL, et al. Coculture of marine invertebrate-associated bacteria and interdisciplinary technologies enable biosynthesis and discovery of a new antibiotic, Keyicin. ACS Chem Biol. 2017;12:3093–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Zhu F, Chen G, Chen X, Huang M, Wan X. Aspergicin, a new antibacterial alkaloid produced by mixed fermentation of two marine-derived mangrove epiphytic fungi. Chem Nat Compd. 2011;47:767–9.

    Article  CAS  Google Scholar 

  145. Nichols D, Cahoon N, Trakhtenberg EM, Pham L, Mehta A, Belanger A, Kanigan T, Lewis K, Epstein SS. Use of ichip for high-throughput in situ cultivation of “uncultivable” microbial species. Appl Environ Microbiol. 2010;76:2445–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Ling LL, Schneider T, Peoples AJ, Spoering AL, Engels I, Conlon BP, Mueller A, Schäberle TF, Hughes DE, Epstein S, et al. A new antibiotic kills pathogens without detectable resistance. Nature. 2015;517:455–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shukla R, Lavore F, Maity S, Derks MGN, Jones CR, Vermeulen BJA, Melcrová A, Morris MA, Becker LM, Wang X, et al. Teixobactin kills bacteria by a two-pronged attack on the cell envelope. Nature. 2022;608:390–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Shukla R, Peoples AJ, Ludwig KC, Maity S, Derks MGN, De Benedetti S, Krueger AM, Vermeulen BJA, Harbig T, Lavore F, et al. An antibiotic from an uncultured bacterium binds to an immutable target. Cell. 2023;186(4059–4073):e4027.

    Google Scholar 

  149. Scioli G, Marinaccio L, Bauer M, Kamysz W, Parmar A, Newire E, Singh I, Stefanucci A, Mollica A. New teixobactin analogues with a total lactam ring. ACS Med Chem Lett. 2023;14:1827–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Gunjal VB, Thakare R, Chopra S, Reddy DS. Teixobactin: a paving stone toward a new class of antibiotics? J Med Chem. 2020;63:12171–95.

    Article  CAS  PubMed  Google Scholar 

  151. Garmendia L, Hernandez A, Sanchez MB, Martinez JL. Metagenomics and antibiotics. Clin Microbiol Infect. 2012;18(Suppl 4):27–31.

    Article  CAS  PubMed  Google Scholar 

  152. Udwary DW, Zeigler L, Asolkar RN, Singan V, Lapidus A, Fenical W, Jensen PR, Moore BS. Genome sequencing reveals complex secondary metabolome in the marine actinomycete Salinispora tropica. Proc Natl Acad Sci U S A. 2007;104:10376–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Hiraoka S, Yang CC, Iwasaki W. Metagenomics and bioinformatics in microbial ecology: current status and beyond. Microbes Environ. 2016;31:204–12.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Prayogo FA, Budiharjo A, Kusumaningrum HP, Wijanarka W, Suprihadi A, Nurhayati N. Metagenomic applications in exploration and development of novel enzymes from nature: a review. J Genet Eng Biotechnol. 2020;18:39.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Gillespie DE, Brady SF, Bettermann AD, Cianciotto NP, Liles MR, Rondon MR, Clardy J, Goodman RM, Handelsman J. Isolation of antibiotics turbomycin a and B from a metagenomic library of soil microbial DNA. Appl Environ Microbiol. 2002;68:4301–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Handelsman J. Metagenomics: application of genomics to uncultured microorganisms. Microbiol Mol Biol Rev. 2004;68:669–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Blin K, Shaw S, Augustijn HE, Reitz ZL, Biermann F, Alanjary M, Fetter A, Terlouw BR, Metcalf WW, Helfrich EJN, et al. antiSMASH 7.0: new and improved predictions for detection, regulation, chemical structures and visualisation. Nucleic Acids Res. 2023;51:W46-50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Kim DR, Kwak YS. A genome-wide analysis of antibiotic producing genes in Streptomyces globisporus SP6C4. Plant Pathol J. 2021;37:389–95.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Chu J, Koirala B, Forelli N, Vila-Farres X, Ternei MA, Ali T, Colosimo DA, Brady SF. Synthetic-bioinformatic natural product antibiotics with diverse modes of action. J Am Chem Soc. 2020;142:14158–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Torres Salazar BO, Dema T, Schilling NA, Janek D, Bornikoel J, Berscheid A, Elsherbini AMA, Krauss S, Jaag SJ, Lämmerhofer M, et al. Commensal production of a broad-spectrum and short-lived antimicrobial peptide polyene eliminates nasal Staphylococcus aureus. Nat Microbiol. 2024;9:200–13.

    Article  CAS  PubMed  Google Scholar 

  161. van Heel AJ, de Jong A, Song C, Viel JH, Kok J, Kuipers OP. BAGEL4: a user-friendly web server to thoroughly mine RiPPs and bacteriocins. Nucleic Acids Res. 2018;46:W278–81.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Skinnider MA, Johnston CW, Gunabalasingam M, Merwin NJ, Kieliszek AM, MacLellan RJ, Li H, Ranieri MRM, Webster ALH, Cao MPT, et al. Comprehensive prediction of secondary metabolite structure and biological activity from microbial genome sequences. Nat Commun. 2020;11:6058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Santos-Aberturas J, Chandra G, Frattaruolo L, Lacret R, Pham TH, Vior NM, Eyles TH, Truman AW. Uncovering the unexplored diversity of thioamidated ribosomal peptides in actinobacteria using the RiPPER genome mining tool. Nucleic Acids Res. 2019;47:4624–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Almeida H, Palys S, Tsang A, Diallo AB. TOUCAN: a framework for fungal biosynthetic gene cluster discovery. NAR Genom Bioinform. 2020;2:lqaa098.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Zhang MM, Wong FT, Wang Y, Luo S, Lim YH, Heng E, Yeo WL, Cobb RE, Enghiad B, Ang EL, Zhao H. CRISPR-Cas9 strategy for activation of silent Streptomyces biosynthetic gene clusters. Nat Chem Biol. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nchembio.2341.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Zhou Z, Xu Q, Bu Q, Guo Y, Liu S, Liu Y, Du Y, Li Y. Genome mining-directed activation of a silent angucycline biosynthetic gene cluster in Streptomyces chattanoogensis. ChemBioChem. 2015;16:496–502.

    Article  CAS  PubMed  Google Scholar 

  167. Guan H, Li Y, Zheng J, Liu N, Zhang J, Tan H. Important role of a LAL regulator StaR in the staurosporine biosynthesis and high-production of Streptomyces fradiae CGMCC 4576. Sci China Life Sci. 2019;62:1638–54.

    Article  CAS  PubMed  Google Scholar 

  168. Xie C, Deng JJ, Wang HX. Identification of AstG1, A LAL family regulator that positively controls ansatrienins production in Streptomyces sp. XZQH13. Curr Microbiol. 2015;70:859–64.

    Article  CAS  PubMed  Google Scholar 

  169. Culp EJ, Yim G, Waglechner N, Wang W, Pawlowski AC, Wright GD. Hidden antibiotics in actinomycetes can be identified by inactivation of gene clusters for common antibiotics. Nat Biotechnol. 2019;37:1149–54.

    Article  CAS  PubMed  Google Scholar 

  170. El-Hawary SS, Hassan MHA, Hudhud AO, Abdelmohsen UR, Mohammed R. Elicitation for activation of the actinomycete genome’s cryptic secondary metabolite gene clusters. RSC Adv. 2023;13:5778–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Challis GL, Ravel J. Coelichelin, a new peptide siderophore encoded by the Streptomyces coelicolor genome: structure prediction from the sequence of its non-ribosomal peptide synthetase. FEMS Microbiol Lett. 2000;187:111–4.

    Article  CAS  PubMed  Google Scholar 

  172. M SH, Hassan HM, Mohammed R, M MF, Sayed AM, A AH, S FA, Rateb ME, Alhadrami HA, Abdelmohsen UR. Induction of antibacterial metabolites by co-cultivation of two red-sea-sponge-associated actinomycetes Micromonospora sp. UR56 and Actinokinespora sp. EG49. Mar Drugs. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/md18050243.

    Article  Google Scholar 

  173. Liu SH, Wang W, Wang KB, Zhang B, Li W, Shi J, Jiao RH, Tan RX, Ge HM. Heterologous expression of a cryptic giant type I PKS gene cluster leads to the production of ansaseomycin. Org Lett. 2019;21:3785–8.

    Article  CAS  PubMed  Google Scholar 

  174. Yang D, Eun H, Prabowo CPS, Cho S, Lee SY. Metabolic and cellular engineering for the production of natural products. Curr Opin Biotechnol. 2022;77:102760.

    Article  CAS  PubMed  Google Scholar 

  175. Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346:1258096.

    Article  PubMed  Google Scholar 

  176. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Zhang X, Wang Y, Zhang Y, Wang M. CRISPR/Cas9-mediated multi-locus promoter engineering in ery cluster to improve erythromycin production in Saccharopolyspora erythraea. Microorganisms. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/microorganisms11030623.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Li L, Zheng G, Chen J, Ge M, Jiang W, Lu Y. Multiplexed site-specific genome engineering for overproducing bioactive secondary metabolites in actinomycetes. Metab Eng. 2017;40:80–92.

    Article  CAS  PubMed  Google Scholar 

  179. Tong Y, Whitford CM, Robertsen HL, Blin K, Jørgensen TS, Klitgaard AK, Gren T, Jiang X, Weber T, Lee SY. Highly efficient DSB-free base editing for streptomycetes with CRISPR-BEST. Proc Natl Acad Sci USA. 2019;116:20366–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Wang J, Wang K, Deng Z, Zhong Z, Sun G, Mei Q, Zhou F, Deng Z, Sun Y. Engineered cytosine base editor enabling broad-scope and high-fidelity gene editing in Streptomyces. Nat Commun. 2024;15:5687.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Wu Y, Kang Q, Zhang LL, Bai L. Subtilisin-involved morphology engineering for improved antibiotic production in actinomycetes. Biomolecules. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom10060851.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Wang W, Li S, Li Z, Zhang J, Fan K, Tan G, Ai G, Lam SM, Shui G, Yang Z, et al. Harnessing the intracellular triacylglycerols for titer improvement of polyketides in Streptomyces. Nat Biotechnol. 2020;38:76–83.

    Article  PubMed  Google Scholar 

  183. Buijs Y, Zhang SD, Jørgensen KM, Isbrandt T, Larsen TO, Gram L. Enhancement of antibiotic production by co-cultivation of two antibiotic producing marine Vibrionaceae strains. FEMS Microbiol Ecol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/femsec/fiab041.

    Article  PubMed  Google Scholar 

  184. Yang D, Park SY, Park YS, Eun H, Lee SY. Metabolic engineering of Escherichia coli for natural product biosynthesis. Trends Biotechnol. 2020;38:745–65.

    Article  CAS  PubMed  Google Scholar 

  185. Yang D, Prabowo CPS, Eun H, Park SY, Cho IJ, Jiao S, Lee SY. Escherichia coli as a platform microbial host for systems metabolic engineering. Essays Biochem. 2021;65:225–46.

    Article  CAS  PubMed  Google Scholar 

  186. Pfeifer BA, Admiraal SJ, Gramajo H, Cane DE, Khosla C. Biosynthesis of complex polyketides in a metabolically engineered strain of E. coli. Science. 2001;291:1790–2.

    Article  CAS  PubMed  Google Scholar 

  187. Fang L, Guell M, Church GM, Pfeifer BA. Heterologous erythromycin production across strain and plasmid construction. Biotechnol Prog. 2018;34:271–6.

    Article  CAS  PubMed  Google Scholar 

  188. Yang D, Eun H, Prabowo CPS. Metabolic engineering and synthetic biology approaches for the heterologous production of aromatic polyketides. Int J Mol Sci. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24108923.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Cummings M, Peters AD, Whitehead GFS, Menon BRK, Micklefield J, Webb SJ, Takano E. Assembling a plug-and-play production line for combinatorial biosynthesis of aromatic polyketides in Escherichia coli. PLoS Biol. 2019;17:e3000347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Yang D, Jang WD, Lee SY. Production of carminic acid by metabolically engineered Escherichia coli. J Am Chem Soc. 2021;143:5364–77.

    Article  CAS  PubMed  Google Scholar 

  191. Shomar H, Gontier S, van den Broek NJF, Tejeda Mora H, Noga MJ, Hagedoorn PL, Bokinsky G. Metabolic engineering of a carbapenem antibiotic synthesis pathway in Escherichia coli. Nat Chem Biol. 2018;14:794–800.

    Article  CAS  PubMed  Google Scholar 

  192. Zhan C, Lee N, Lan G, Dan Q, Cowan A, Wang Z, Baidoo EEK, Kakumanu R, Luckie B, Kuo RC, et al. Improved polyketide production in C. glutamicum by preventing propionate-induced growth inhibition. Nat Metab. 2023;5:1127–40.

    Article  CAS  PubMed  Google Scholar 

  193. Martínez-García E, de Lorenzo V. Pseudomonas putida as a synthetic biology chassis and a metabolic engineering platform. Curr Opin Biotechnol. 2024;85:103025.

    Article  PubMed  Google Scholar 

  194. Yang F, Cao Y. Biosynthesis of phloroglucinol compounds in microorganisms–review. Appl Microbiol Biotechnol. 2012;93:487–95.

    Article  CAS  PubMed  Google Scholar 

  195. Gurney R, Thomas CM. Mupirocin: biosynthesis, special features and applications of an antibiotic from a gram-negative bacterium. Appl Microbiol Biotechnol. 2011;90:11–21.

    Article  CAS  PubMed  Google Scholar 

  196. Schwanemann T, Otto M, Wynands B, Marienhagen J, Wierckx N. A Pseudomonas taiwanensis malonyl-CoA platform strain for polyketide synthesis. Metab Eng. 2023;77:219–30.

    Article  CAS  PubMed  Google Scholar 

  197. Siddiqui MS, Thodey K, Trenchard I, Smolke CD. Advancing secondary metabolite biosynthesis in yeast with synthetic biology tools. FEMS Yeast Res. 2012;12:144–70.

    Article  CAS  PubMed  Google Scholar 

  198. Awan AR, Blount BA, Bell DJ, Shaw WM, Ho JCH, McKiernan RM, Ellis T. Biosynthesis of the antibiotic nonribosomal peptide penicillin in baker’s yeast. Nat Commun. 2017;8:15202.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Jakočiūnas T, Klitgaard AK, Kontou EE, Nielsen JB, Thomsen E, Romero-Suarez D, Blin K, Petzold CJ, Gin JW, Tong Y, et al. Programmable polyketide biosynthesis platform for production of aromatic compounds in yeast. Synth Syst Biotechnol. 2020;5:11–8.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Satpute SK, Banat IM, Dhakephalkar PK, Banpurkar AG, Chopade BA. Biosurfactants, bioemulsifiers and exopolysaccharides from marine microorganisms. Biotechnol Adv. 2010;28:436–50.

    Article  CAS  PubMed  Google Scholar 

  201. Markham KA, Palmer CM, Chwatko M, Wagner JM, Murray C, Vazquez S, Swaminathan A, Chakravarty I, Lynd NA, Alper HS. Rewiring Yarrowia lipolytica toward triacetic acid lactone for materials generation. Proc Natl Acad Sci USA. 2018;115:2096–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Yu J, Landberg J, Shavarebi F, Bilanchone V, Okerlund A, Wanninayake U, Zhao L, Kraus G, Sandmeyer S. Bioengineering triacetic acid lactone production in Yarrowia lipolytica for pogostone synthesis. Biotechnol Bioeng. 2018;115:2383–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Yang D, Zhou H, Lee SY. Production of diversified polyketides by metabolic engineering. Biochemistry. 2021;60:3424–6.

    Article  CAS  PubMed  Google Scholar 

  204. Tian DS, Zhang X, Cox RJ. Comparing total chemical synthesis and total biosynthesis routes to fungal specialized metabolites. Nat Prod Rep. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/d4np00015c.

    Article  PubMed  Google Scholar 

  205. Li J, Chen G, Wu H, Webster JM. Identification of two pigments and a hydroxystilbene antibiotic from Photorhabdus luminescens. Appl Environ Microbiol. 1995;61:4329–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Ghimire GP, Koirala N, Pandey RP, Jung HJ, Sohng JK. Modification of emodin and aloe-emodin by glycosylation in engineered Escherihia coli. World J Microbiol Biotechnol. 2015;31:611–9.

    Article  CAS  PubMed  Google Scholar 

  207. Zhang G, Li Y, Fang L, Pfeifer BA. Tailoring pathway modularity in the biosynthesis of erythromycin analogs heterologously engineered in E. coli. Sci Adv. 2015;1:e1500077.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Fang L, Zhang G, El-Halfawy O, Simon M, Brown ED, Pfeifer BA. Broadened glycosylation patterning of heterologously produced erythromycin. Biotechnol Bioeng. 2018;115:2771–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Gillis EP, Eastman KJ, Hill MD, Donnelly DJ, Meanwell NA. Applications of fluorine in medicinal chemistry. J Med Chem. 2015;58:8315–59.

    Article  CAS  PubMed  Google Scholar 

  210. Wang J, Sánchez-Roselló M, Aceña JL, del Pozo C, Sorochinsky AE, Fustero S, Soloshonok VA, Liu H. Fluorine in pharmaceutical industry: fluorine-containing drugs introduced to the market in the last decade (2001–2011). Chem Rev. 2014;114:2432–506.

    Article  CAS  PubMed  Google Scholar 

  211. Sirirungruang S, Ad O, Privalsky TM, Ramesh S, Sax JL, Dong H, Baidoo EEK, Amer B, Khosla C, Chang MCY. Engineering site-selective incorporation of fluorine into polyketides. Nat Chem Biol. 2022;18:886–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Englund E, Schmidt M, Nava AA, Lechner A, Deng K, Jocic R, Lin Y, Roberts J, Benites VT, Kakumanu R, et al. Expanding extender substrate selection for unnatural polyketide biosynthesis by acyltransferase domain exchange within a modular polyketide synthase. J Am Chem Soc. 2023;145:8822–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Koch AA, Hansen DA, Shende VV, Furan LR, Houk KN, Jiménez-Osés G, Sherman DH. A single active site mutation in the pikromycin thioesterase generates a more effective macrocyclization catalyst. J Am Chem Soc. 2017;139:13456–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Thong WL, Zhang Y, Zhuo Y, Robins KJ, Fyans JK, Herbert AJ, Law BJC, Micklefield J. Gene editing enables rapid engineering of complex antibiotic assembly lines. Nat Commun. 2021;12:6872.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Hagen A, Poust S, Rond T, Fortman JL, Katz L, Petzold CJ, Keasling JD. Engineering a polyketide synthase for In Vitro production of adipic acid. ACS Synth Biol. 2016;5:21–7.

    Article  CAS  PubMed  Google Scholar 

  216. Zargar A, Lal R, Valencia L, Wang J, Backman TWH, Cruz-Morales P, Kothari A, Werts M, Wong AR, Bailey CB, et al. Chemoinformatic-guided engineering of polyketide synthases. J Am Chem Soc. 2020;142:9896–901.

    Article  CAS  PubMed  Google Scholar 

  217. Yuzawa S, Mirsiaghi M, Jocic R, Fujii T, Masson F, Benites VT, Baidoo EEK, Sundstrom E, Tanjore D, Pray TR, et al. Short-chain ketone production by engineered polyketide synthases in Streptomyces albus. Nat Commun. 2018;9:4569.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Abe I, Takahashi Y, Noguchi H. Enzymatic formation of an unnatural C(6)-C(5) aromatic polyketide by plant type III polyketide synthases. Org Lett. 2002;4:3623–6.

    Article  CAS  PubMed  Google Scholar 

  219. Morita H, Shimokawa Y, Tanio M, Kato R, Noguchi H, Sugio S, Kohno T, Abe I. A structure-based mechanism for benzalacetone synthase from Rheum palmatum. Proc Natl Acad Sci USA. 2010;107:669–73.

    Article  CAS  PubMed  Google Scholar 

  220. Jez JM, Bowman ME, Noel JP. Expanding the biosynthetic repertoire of plant type III polyketide synthases by altering starter molecule specificity. Proc Natl Acad Sci U S A. 2002;99:5319–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Morita H, Yamashita M, Shi SP, Wakimoto T, Kondo S, Kato R, Sugio S, Kohno T, Abe I. Synthesis of unnatural alkaloid scaffolds by exploiting plant polyketide synthase. Proc Natl Acad Sci USA. 2011;108:13504–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Wanibuchi K, Morita H, Noguchi H, Abe I. Enzymatic formation of an aromatic dodecaketide by engineered plant polyketide synthase. Bioorg Med Chem Lett. 2011;21:2083–6.

    Article  CAS  PubMed  Google Scholar 

  223. Jeya M, Kim TS, Kumar Tiwari M, Li J, Zhao H, Lee JK. A type III polyketide synthase from Rhizobium etli condenses malonyl CoAs to a heptaketide pyrone with unusually high catalytic efficiency. Mol Biosyst. 2012;8:3103–6.

    Article  CAS  PubMed  Google Scholar 

  224. Englund E, Schmidt M, Nava AA, Klass S, Keiser L, Dan Q, Katz L, Yuzawa S, Keasling JD. Biosensor guided polyketide synthases engineering for optimization of domain exchange boundaries. Nat Commun. 2023;14:4871.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Deng JZ. Methicillin/per-6-(4-methoxylbenzyl)-amino-6-deoxy-beta-cyclodextrin 1:1 complex and its potentiation in vitro against methicillin-resistant Staphylococcus aureus. J Antibiot. 2013;66:517–21.

    Article  CAS  Google Scholar 

  226. Roy J. An introduction to pharmaceutical sciences: production, chemistry, techniques and technology. Oxford: Biohealthcare Pub; 2011.

    Book  Google Scholar 

  227. Smith PA, Koehler MFT, Girgis HS, Yan D, Chen Y, Chen Y, Crawford JJ, Durk MR, Higuchi RI, Kang J, et al. Optimized arylomycins are a new class of gram-negative antibiotics. Nature. 2018;561:189–94.

    Article  CAS  PubMed  Google Scholar 

  228. Grandclaudon C, Birudukota NVS, Elgaher WAM, Jumde RP, Yahiaoui S, Arisetti N, Hennessen F, Hüttel S, Stadler M, Herrmann J, et al. Semisynthesis and biological evaluation of amidochelocardin derivatives as broad-spectrum antibiotics. Eur J Med Chem. 2020;188:112005.

    Article  CAS  PubMed  Google Scholar 

  229. Tevyashova AN, Efimova SS, Alexandrov AI, Ghazy E, Bychkova EN, Solovieva SE, Zatonsky GB, Grammatikova NE, Dezhenkova LG, Pereverzeva ER, et al. Semisynthetic amides of polyene antibiotic natamycin. ACS Infect Dis. 2023;9:42–55.

    Article  CAS  PubMed  Google Scholar 

  230. van Groesen E, Mons E, Kotsogianni I, Arts M, Tehrani K, Wade N, Lysenko V, Stel FM, Zwerus JT, De Benedetti S, et al. Semisynthetic guanidino lipoglycopeptides with potent in vitro and in vivo antibacterial activity. Sci Transl Med. 2024;16:4736.

    Article  Google Scholar 

  231. Rahn HP, Liu X, Chosy MB, Sun J, Cegelski L, Wender PA. Biguanide-vancomycin conjugates are effective broad-spectrum antibiotics against actively growing and biofilm-associated gram-positive and gram-negative ESKAPE pathogens and mycobacteria. J Am Chem Soc. 2024;146:22541–52.

    Article  CAS  PubMed  Google Scholar 

  232. Takahashi Y, Igarashi M, Miyake T, Soutome H, Ishikawa K, Komatsuki Y, Koyama Y, Nakagawa N, Hattori S, Inoue K, et al. Novel semisynthetic antibiotics from caprazamycins A-G: caprazene derivatives and their antibacterial activity. J Antibiot. 2013;66:171–8.

    Article  CAS  Google Scholar 

  233. Stojković D, Petrović J, Carević T, Soković M, Liaras K. Synthetic and semisynthetic compounds as antibacterials targeting virulence traits in resistant strains: a narrative updated review. Antibiotics. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antibiotics12060963.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Goel B, Tripathi N, Bhardwaj N, Pal Singh I, Jain SK. Semisynthesis: an essential tool for antibiotics drug discovery. ChemistrySelect. 2024;9:e202400554.

    Article  CAS  Google Scholar 

  235. Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, Leonhardt H, Heyn H, Hellmann I, Enard W. Comparative analysis of single-cell RNA sequencing methods. Mol Cell. 2017;65(631–643):e634.

    Google Scholar 

  236. Kingwell K. Microbial “dark matter” yields new antibiotic. Nat Rev Drug Discov. 2023;22:872.

    Article  CAS  PubMed  Google Scholar 

  237. Nielsen J, Keasling JD. Engineering cellular metabolism. Cell. 2016;164:1185–97.

    Article  CAS  PubMed  Google Scholar 

  238. Sollier J, Basler M, Broz P, Dittrich PS, Drescher K, Egli A, Harms A, Hierlemann A, Hiller S, King CG, et al. Revitalizing antibiotic discovery and development through in vitro modelling of in-patient conditions. Nat Microbiol. 2024;9:1–3.

    Article  CAS  PubMed  Google Scholar 

  239. Li X, Tian T. Recent advances in an organ-on-a-chip: biomarker analysis and applications. Anal Methods. 2018;10:3122–30.

    Article  Google Scholar 

  240. Abramson J, Adler J, Dunger J, Evans R, Green T, Pritzel A, Ronneberger O, Willmore L, Ballard AJ, Bambrick J, et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature. 2024;630:493–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Wong F, Zheng EJ, Valeri JA, Donghia NM, Anahtar MN, Omori S, Li A, Cubillos-Ruiz A, Krishnan A, Jin W, et al. Discovery of a structural class of antibiotics with explainable deep learning. Nature. 2024;626:177–85.

    Article  CAS  PubMed  Google Scholar 

  242. Zheng EJ, Valeri JA, Andrews IW, Krishnan A, Bandyopadhyay P, Anahtar MN, Herneisen A, Schulte F, Linnehan B, Wong F, et al. Discovery of antibiotics that selectively kill metabolically dormant bacteria. Cell Chem Biol. 2024;31(712–728):e719.

    Google Scholar 

  243. Wong F, de la Fuente-Nunez C, Collins JJ. Leveraging artificial intelligence in the fight against infectious diseases. Science. 2023;381:164–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. MacNair CR, Rutherford ST, Tan M-W. Alternative therapeutic strategies to treat antibiotic-resistant pathogens. Nat Rev Microbiol. 2024;22:262–75.

    Article  CAS  PubMed  Google Scholar 

  245. King AM, Zhang Z, Glassey E, Siuti P, Clardy J, Voigt CA. Systematic mining of the human microbiome identifies antimicrobial peptides with diverse activity spectra. Nat Microbiol. 2023;8:2420–34.

    Article  CAS  PubMed  Google Scholar 

  246. Torres MDT, Brooks EF, Cesaro A, Sberro H, Gill MO, Nicolaou C, Bhatt AS, de la Fuente-Nunez C. Mining human microbiomes reveals an untapped source of peptide antibiotics. Cell. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2024.07.027.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Research Foundation of Korea(NRF) grants funded by the Korea government(MSIT) (RS-2024–00398252 and RS-2024–00440975).

Author information

Authors and Affiliations

Authors

Contributions

G.Y, J.N: Conceptualization, Writing, Reviewing, Investigation, Data Curation, Visualization Y.J, H.Y: Conceptualization, Writing the original draft, Investigation, D.Y: Conceptualization, Writing, Revewing, Investigation, Supervision, Project administraiton, Funding acquisition. All authors reviewed and approved the final manuscript.

Corresponding author

Correspondence to Dongsoo Yang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yook, G., Nam, J., Jo, Y. et al. Metabolic engineering approaches for the biosynthesis of antibiotics. Microb Cell Fact 24, 35 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12934-024-02628-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12934-024-02628-2

Keywords