Skip to main content
mSystems logoLink to mSystems
. 2023 Oct 19;8(6):e00835-23. doi: 10.1128/msystems.00835-23

Metabolic modeling predicts unique drug targets in Borrelia burgdorferi

Peter J Gwynne 1,2,, Kee-Lee K Stocks 1,2, Elysse S Karozichian 1,2, Aarya Pandit 1,2, Linden T Hu 1,2
Editor: Anthony Fodor3
PMCID: PMC10734484  PMID: 37855615

ABSTRACT

The Lyme disease bacterium Borrelia burgdorferi is extremely host dependent, with a small genome and correspondingly limited metabolism. As such, it is an excellent candidate for the development of targeted, narrow-spectrum antimicrobials. To accelerate drug discovery in this fastidious bacterium, in silico genome-scale metabolic modeling was used to construct a map of B. burgdorferi’s metabolism. This map was used to predict essential genes and enzymes; experimental data validated these predicted hits as viable drug targets. Repurposing existing small-molecule inhibitors, it is shown that inhibition of two predicted essential enzymes (pyridoxal kinase and serine hydroxymethyltransferase) selectively kills B. burgdorferi in culture. Thus, the essential processes identified here represent targets for the development of narrow-spectrum antimicrobials. This pipeline, pairing in silico discovery with validation in culture, may be useful for other genetically intractable pathogens.

IMPORTANCE

Lyme disease is often treated using long courses of antibiotics, which can cause side effects for patients and risks the evolution of antimicrobial resistance. Narrow-spectrum antimicrobials would reduce these risks, but their development has been slow because the Lyme disease bacterium, Borrelia burgdorferi, is difficult to work with in the laboratory. To accelerate the drug discovery pipeline, we developed a computational model of B. burgdorferi’s metabolism and used it to predict essential enzymatic reactions whose inhibition prevented growth in silico. These predictions were validated using small-molecule enzyme inhibitors, several of which were shown to have specific activity against B. burgdorferi. Although the specific compounds used are not suitable for clinical use, we aim to use them as lead compounds to develop optimized drugs targeting the pathways discovered here.

KEYWORDS: antimicrobial agents, Lyme disease, drug discovery, host-pathogen interactions, metabolic modeling

INTRODUCTION

Borrelia burgdorferi has a small genome of ~1.5 Mbp (13) and, as a result, is highly dependent on its tick and vertebrate hosts for the provision of many metabolites (46). Small genomes by necessity have few redundant pathways or enzymes and a higher proportion of essential genes. The minimal genome of B. burgdorferi, therefore, makes it an attractive candidate for the development of narrow-spectrum antibiotics targeting these essential genes.

While the bacterium does not live independently of its hosts in the wild, it is culturable in the laboratory. The organism is extremely fastidious, however, and requires a complex and undefined growth medium. The standard in vitro growth medium Barbour-Stoenner-Kelly (BSK) contains numerous undefined components: yeast extract, bovine serum albumin, and rabbit serum (7, 8). Traditional metabolic and genetic studies are further hindered by the species’ slow growth rate (9) and a complex and unstable genome (10, 11). To circumvent the challenges of in vitro metabolic studies, we have developed a genome-scale metabolic model for B. burgdorferi B31. Genome-scale metabolic modeling permits the creation and analysis of in silico simulations of metabolism, as predicted by comparison of an organism’s protein coding sequences with those of known metabolic enzymes (12). Similar models are often used for the optimization of pathways and strains in the bioproduction of high-value metabolites (1315) but have also been applied to the study of pathogenesis and host-pathogen interactions, particularly in intractable organisms (16, 17).

In addition to describing the basic metabolic capacity of the organism, such models can be used to predict essential processes. The prediction of essential reactions by genome-scale metabolic models has allowed for the identification of drug targets and the design of novel drugs in both prokaryotic (18) and eukaryotic (19) systems. Comparison of the new B. burgdorferi metabolic model with existing models for well-characterized pathogens (Escherichia coli and Staphylococcus aureus) permits the selection of targeted antimicrobials with specific activity against B. burgdorferi.

Current clinical guidelines recommend treatment of Lyme borreliosis with antibiotics (penicillins, tetracyclines, or macrolides) with risks of nosocomial infections such as Clostridiodes difficile due to disruption of the native microbiome (20, 21). Although antimicrobial resistance is rarely described in Borrelia, the use of broad-spectrum antibiotics [and, in particular, extended treatment regimens (22)] may also drive enduring resistance in off-target bacteria (23). Lyme disease prophylaxis is currently limited to the use of antibiotics within a narrow window after a high-risk tick bite (24). An alternative means of reducing infections is the reduction of carriage in wild animal reservoirs. Previous studies treating small mammals with doxycycline were highly effective in reducing Borrelia and Anaplasma infections (25, 26), but concerns around antimicrobial resistance have prevented widespread implementation.

The identification and use of narrow-spectrum antibiotics [such as the recently rediscovered hygromycin A (27)] with specific activity against the Lyme disease Borrelia would minimize the potential harms of prolonged broad-spectrum antibiotic use. In addition to reducing the risk of antimicrobial resistance in off-target commensals, targeted anti-borrelials could also be used prophylactically by high-risk individuals or to reduce infections by eradication of the disease from its wild animal reservoirs. We describe the construction, analysis, and validation of a metabolic model for B. burgdorferi for the prediction of critical reactions which are targets for the development of narrow-spectrum anti-borrelials.

MATERIALS AND METHODS

Metabolic model construction

The complete B. burgdorferi B31 genome [GenBank assembly GCF_000008695.2 (2, 3)] of a ~900-kbp chromosome and 22 plasmids was used throughout. The metabolic model iBB151 was constructed using RAVEN 2.5.3 (28), chosen for its ability to integrate models derived from multiple sources. Reconstruction and analysis were performed in MATLAB R2020a. In absence of a scaffold model from a close genetic relative, three models were created by different methods and integrated. Two models were derived from existing metabolic databases KEGG (RAVEN command getKEGGModelForOrganism) and MetaCyc (RAVEN command getMetaCycModelForOrganism). The KEGG database identifies metabolic functions from coding sequences based on similarity to characterized enzymes (29). KEGG contains automated annotations of over 5,000 genomes, including that of B. burgdorferi B31, with associated reactions and metabolites. MetaCyc is a curated database of metabolic enzymes, reactions, and metabolites (30). These curated enzyme sequences were queried using the B. burgdorferi B31 genome to generate de novo a draft model structure.

The RAVEN 2.5 package also includes a series of hidden Markov models (HMMs) trained using HMMER3 (31) on nonredundant protein sequences derived from the KEGG database (version 91.0) and arranged in nonredundant clusters by CD-HIT (32). Three HMMs were used to generate draft models based on similarity to KEGG orthologues (RAVEN command getKEGGModelForOrganism) with redundancy cutoffs of 50%, 90%, and 100%. These three models were merged first with each other and then the KEGG model (RAVEN command mergeModels) and, finally, with the MetaCyc model (RAVEN command combineMetaCycKEGGModels) to produce the combined draft model containing 378 metabolic reactions. Manual curation removed duplicates, unconnected reactions, and obvious mis-annotations and added processes described experimentally but not predicted by any of the pipelines (33). With few transporters described or predicted in B. burgdorferi, transport and exchange reactions were added to allow every reaction in the model to carry flux (as determined by the RAVEN command hasFlux). Memote (34) was used to confirm stoichiometric consistency. The final Memote score was 87%.

Biomass reaction

In silico “growth” is represented by flux through a pseudo-reaction which consumes known biomass precursors. Biomass composition has not been extensively studied in B. burgdorferi. In the absence of a well-studied close relative of B. burgdorferi, the biomass objective function was derived from that of that of E. coli [as described in the model iAF1260 (35)], with modifications based on available literature. Total lipid composition and contribution to dry weight were adjusted based on experimental data (36), and lipopolysaccharide was removed from the biomass calculations. The contribution of the remaining major biomass constituents (DNA, RNA, protein, and peptidoglycan) were adjusted to account for the higher amount of lipid in B. burgdorferi dry weight. Codon utilization (amino acid composition) and GC content were determined from the genome using CoCoPUTs (37). Metal ions were omitted, as were any cofactors not predicted elsewhere in the model including thiamin, which is not detectable in B. burgdorferi (38).

Analysis of iBB151

Analysis was performed using the COBRA toolbox v3.0 (39) to determine flux through each reaction while maximizing for a single objective function. For all analyses, the objective function was the reaction EX_BM, the export of biomass. Essential reactions were identified using the Cobra function singleRxnDeletion, which sequentially inactivates each reaction and attempts to solve the model for the objective function. Any inactivation which produces an insoluble model (i.e., a growth rate of 0) is classed as essential. Synthetic lethal single and double gene deletions were similarly predicted using singleGeneDeletion and doubleGeneDeletion. For comparison with other organisms, the same method was used on existing E. coli and S. aureus metabolic models iML1515 (40) and iYS854 (41).

Minimum inhibitory concentrations

Inhibitors of predicted essential reactions were identified by literature search. These were tested against B. burgdorferi by growth BSK II medium, composed of bovine serum albumin (50.00 g/L), CMRL-1066 [US Biologicals (9.80 g/L)], HEPES (6.60 g/L), peptone (5.60 g/L), dextrose (5.60 g/L), sodium bicarbonate (2.44 g/L), yeast extract (2.20 g/L), sodium pyruvate (1.00 g/L), sodium citrate (0.90 g/L), N-acetyl glucosamine (0.50 g/L), and 6.2% rabbit serum. Media were filter sterilized, and the pH was adjusted to 7.6 before the addition of gelatin to 1.4% and sterile water to 1 L and stored at −20°C. The B. burgdorferi B31 isolate used throughout this study was an infectious strain, cultured from the ear of an infected C3H-HeJ mouse. By PCR plasmid typing, this strain possessed all B31 plasmids except lp5, cp32-6, and cp32-9, which carry no metabolic genes either described previously or predicted in the modeling above.

Minimum inhibitory concentration (MIC) was determined for three drugs: cycloserine (Thermo Scientific) at 0–256 μg/mL, theophylline (TCI Chemicals), and pemetrexed hydrate (TCI Chemicals) at 0–8 mg/mL. Cycloserine was diluted 1:500 from a stock (in water) of 128 mg/mL: an equivalent volume of water was added to no-drug controls. Theophylline and pemetrexed were dissolved directly into BSK or tryptic soy broth (TSB) medium, which was then filter sterilized with a 0.22-µm nitrocellulose syringe filter. Dilution series were inoculated with 1 × 105 cells/mL B. burgdorferi B31 from an early stationary phase (1 × 108 cells/mL) culture. Cultures were grown in volumes of 200 µL in uncoated flat-bottom 96-well plates at 32°C with 1% CO2 for 5–7 days, until drug-free controls reached at least 1 × 108 cells/mL. Cells were directly enumerated using a Petroff-Hausser counter (Hausser Scientific). E. coli MG1655 and S. aureus 502a were cultured in TSB (Becton Dickinson): tryptone (17.0 g/L), soytone (3.0 g/L), glucose (2.5 g/L), sodium chloride (5.0 g/L), and dipotassium phosphate (2.5 g/L). Overnight cultures were diluted 1:1,000 into fresh media containing the same dilutions series of drug and grown overnight (18 hours) at 37°C. Growth was measured by optical density at 600 nm (OD600) using a plate reader (Biotek Synergy H1). The MIC was defined as the lowest concentration at which growth was reduced by 90% or more compared to an untreated control. Growth data presented are the average of biological triplicates in all cases.

Measurement of bacterial viability

Triplicate B. burgdorferi B31 cultures were grown to exponential phase (6 × 106–2 × 107) or stationary phase (>9 × 107). Five-hundred-microliter culture was pelleted (10,000 g, 5 minutes) and resuspended in 500-µL fresh BSK containing theophylline (8.0 and 1.0 mg/mL), pemetrexed (4.0 and 0.5 mg/mL), or doxycycline (1.0 µg/mL). Untreated controls were resuspended in BSK only. All conditions were incubated for 20 hours at 32°C with 1% CO2. For enumeration of colony-forming units, cultures were plated in overlay agars onto BSK composed as above but for the omission of gelatin and the addition of agarose to 0.65% to solidify the media. Overlays that were 1.8 mL were plated onto 5-mL plates in 60-mm Petri dishes. Treated cultures were diluted 10−5 or 10−6 into a final volume of 900 µL, to which 900-µL 1.7% low-melting point agarose (Apex) was added. The lower limit of detection was, therefore, 2 × 105 cells or a 99.8% reduction from a 1 × 108 starting culture.

For live/dead staining, 100-µL cell culture was mixed with 100-µL BacLight LIVE/DEAD stain (Invitrogen L7007) and incubated in the dark at room temperature for 15 minutes. In addition to the untreated control, a heat-killed control culture was pelleted as above and resuspended in 500-µL saline (0.9% NaCl, pH 7.4). Two-hundred-microliter cell suspension was incubated at 70°C for 10 minutes. Five-microliter stained cells were visualized on slides using an Echo Revolve fluorescence microscope fitted with darkfield, FITC (Ex: 470/40, Em: 525/50) and TXRED (Ex: 560/40, Em: 630/75) filter cubes. In addition to the untreated control, a heat-killed control culture was pelleted as above and resuspended in 500-µL saline (0.9% NaCl, pH 7.4). Two-hundred-microliter cell suspension was incubated at 70°C for 10 minutes. The FITC channel detected SYTO 9-stained (viable) cells, while the TXRED channel detected propidium iodide-stained (dead) cells. FITC and TXRED images were contrast enhanced and merged (in ImageJ), and fields were counted until a minimum of 200 cells were counted in a single channel. The same images were then counted for the second color, with percentage killing calculated as TXRED cells/(TXRED cells + FITC cells) × 100.

RESULTS

iBB151, the metabolic model of Borrelia burgdorferi

iBB151, an in silico model of B. burgdorferi metabolism, was generated by combining three genome annotation pipelines with experimental data from the literature (Fig. 1). B. burgdorferi B31 has a small genome, with a chromosome of <1 Mbp and plasmids totaling a further 600 kbp, and the model created is correspondingly small, describing only 208 metabolic reactions. A total of 151 genes were annotated here, representing 10.5% of the predicted protein coding sequences of the genome. Reaction and gene annotations are cross-referenced in the “Rxns” tab of the model file, supplemental file 4. This small model is reflective of a genome with a high proportion of hypothetical genes with no similarity outside of the genus and a significant proportion of protein-coding genes devoted to (nonmetabolic) surface lipoproteins (4244). A total of 388 reactions (metabolic, transport, exchange, and biomass reactions) are modeled in iBB151. The previous E. coli and S. aureus models used as controls here model 2,713 and 743 reactions, respectively. The size and complexity of metabolic models reflect the size of the target genome but are also dependent on the degree of conservation with well-characterized reference genomes, as annotations are based on homology to known enzymes. Given the evolutionary divergence of the spirochetes and limited experimental data (33), the value of combining multiple annotations is demonstrated by Fig. 1. Only 71 reactions were predicted by all three annotation methods, while each pipeline contributed numerous unique reactions. The model is available in Microsoft excel format in the supplemental information (supplemental file S4), along with image files depicting model overviews (files S2 and S3). An SBML (version 3.1) file is available at https://github.com/pjgwynne/iBB151.

Fig 1.

Fig 1

Generation and analysis of the genome-scale metabolic model of B. burgdorferi iBB151. Predicted coding sequences from the genome were assigned enzymatic functions by comparison to two databases (KEGG and MetaCyc) and by a de novo annotation using pre-trained HMMs. The curated model mapped a total of 208 enzymatic reactions to 151 genes and was used to predict reaction essentiality by flux balance analysis (FBA). Eighty-five transport reactions provide extracellular metabolites (hollow circles), to 213 cytoplasmic reactions involving 232 cytoplasmic metabolites (blue circles). Thirty-two biomass metabolites (pink circles) produced by 77 essential reactions (pink lines) are consumed in the biomass reaction. A larger, labeled version of the inset metabolic map is available in supplemental file S2.

The overall architecture of the model features two compartments, extracellular and cytoplasmic. Insufficient experimental data exist for B. burgdorferi to confidently predict localization to the periplasmic space. Extracellular metabolites mimic the growth medium and are transported into the cell by transport reactions. Cytoplasmic reactions convert metabolites into biomass metabolites, which are consumed by a biomass reaction to model growth. Many major metabolic pathways are missing from B. burgdorferi. As in previous predictions (1, 2), no enzymes of the tricarboxylic cycle, oxidative phosphorylation, or fatty acid or sterol synthesis are found. Amino acid metabolism is largely absent, as is the urea cycle and polyamine synthesis. Energy is generated through glycolysis, with lactic acid the terminal product (file S3: ‘RXNS’: BB_0087). As demonstrated by experimental data (45, 46) glucose (BB_0730), mannose (BB_0407), glycerol (BB_0241)_, and disaccharides including chitobiose (BB_0620) can be utilized via glycolysis. Other predictions also match available experimental data: N-acetyl glucosamine and N-acetyl mannosamine can be used interchangeably (BB_0644) (47). Acetyl-CoA initiates a partial mevalonate pathway (BB_0685) essential for the synthesis of peptidoglycan precursor undecaprenyl phosphate (48). The arginine deiminase system produces ornithine (BB_0842) (49), which is the peptidoglycan crosslinking diamino acid (50, 51). Again, matching recent experimental data, two glycerol-3-phosphate dehydrogenases were predicted: one using NADH as a cofactor (BB_0368) (52) and one using FADH (BB_0243) (53).

Prediction of essential reactions

Essential reactions were predicted by sequential inactivation of each reaction and solving with growth (reaction EX_BM) as the objective function. Growth rate of the wild type (WT) model in complete media was 0.274 mmol/gDW/h, with all single reaction deletions either having no effect or completely preventing growth. Seventy-seven single reactions were essential for growth, representing 37.0% of the 208 metabolic reactions described. A similar percentage of transport reactions were essential (32/85, 37.6%). The growth rate and growth ratio (growth rate of mutant/growth rate of wild type) after inactivation of each individual reaction are available in supplemental file S5, “SRD,” which lists the growth rate of a model in which each reaction is deleted. Essential reactions are those where the model cannot be solved, and the growth rate is 0. As expected in a small model, 37% is a relatively high percentage of essential reactions. Similar screens of metabolic reactions using models of E. coli (40) and S. aureus (41) predicted 227/1,552 (14.6%) and 435/1,455 (29.8%) essential reactions, respectively. A complete list of essential reactions is included in the supplemental information (file S5: SRD).

Transport processes are poorly understood in B. burgdorferi. Although a small number of transport reactions were predicted by the genome reconstruction pipeline, most were added manually to allow all predicted reactions to carry flux. Thus, 85 transport reactions were required to allow every reaction in the model to function; the complete list of these can be found in supplemental file S4, RXNS tab. Transport of 85 metabolites allowed the full function of the model, but only 32 transport reactions were found to be required for the production of biomass in the above screen of essential reactions. These essential nutrients reflect those which have been described experimentally, including most amino acids (54), lipid precursors (55), and nucleotides (56) as well as a number of cofactors and vitamins for which B. burgdorferi has no predicted synthetic pathways.

A transposon mutagenesis screen was previously completed in B. burgdorferi (57). Although the unsaturated library cannot confidently predict gene essentiality, any gene with a transposon insertion can be assumed to be nonessential for growth in complex medium. Overall, the previous library featured insertions in 45.5% of protein-coding genes (57). Of the 151 genes annotated in iBB151, only 25.8% were previously inactivated in the transposon library, likely reflecting the immutability of core metabolic genes. The transposon library is cross-referenced to the predictions of essentiality in iBB151 in supplemental file S5, SGD. Most of the transposon mutants were found in genes predicted to be nonessential: mutability of the predicted nonessential genes was 38.8%. Growth rates and growth ratios (growth rate of deletion/growth rate of wild type) of each mutant are available in supplemental file S5, SGD tab. As above, a mutant with a growth rate less than the WT (growth ratio <1) is impaired. A mutant resulting in a growth ratio of 0 is lethal.

Only six predicted essential genes (6/66, 9.1% mutability) (BB_0377, BB_0527, BB_0585, BB_0605, BB_0767, and BB_0841) had transposon insertions. Three of these insertions were at the end of the gene (insertion ratio ≥0.97) and thus may not have been inactivating insertions. BB_0377 encodes ribosylhomocysteine lyase, which is essential to avoid the accumulation of waste product S-ribosyl homocysteine in silico. In vivo, this product may be excreted or detoxified elsewhere in the absence of the BB_0377 gene product. BB_0841 is part of the pathway producing the cell wall diamino acid ornithine. This gene may be mutable in cultured Borrelia, where ornithine can be imported from the growth medium, but is essential in silico without this compensatory transport reaction. The product of BB_0605 is predicted to be essential for cell wall maturation but was previously shown to have five transposon insertions. This gene is evidently mis-annotated in iBB151, but no alternative prediction was found.

Pairs of lethal gene deletions may represent targets for combination therapies but are also illustrative of important pathway intersections and rare examples of metabolic redundancy. For instance, BB_0137 and BB_0593 are nonessential individually but lethal in combination. These two genes encode fatty acid ligases producing acyl-CoA and acyl-acyl carrier protein (ACP), respectively. Lethality in the double deletion highlights the importance of fatty acid scavenging. A grid of double gene deletions is shown in supplemental file S5, “DGD.” Unlike the single reaction or single gene deletions, many double gene deletions result in intermediate growth phenotypes (growth ratios >0 but <1).

Identification of anti-borrelial targets

Many of the 77 reactions predicted as essential for growth are the targets of existing antibiotics. These include cell wall synthesis (58) (12 reactions), aminoacyl tRNA ligases (59) (19 reactions), and alanine racemase (60). Others such as the mevalonate pathway (61) (eight reactions) and glutamate racemase (62) have been proposed as targets for the development of novel antibiotics. This in silico screen also predicts the efficacy of compounds targeted at specific borrelial pathways. Adenosylhomocysteine nucleosidase has been extensively characterized as a unique target for the development of drugs with specific activity against B. burgdorferi (63, 64). Similarly, the essentiality of a mammalian-type mevalonate pathway for the synthesis of undecaprenyl phosphate is consistent with prior studies of this pathway (65).

In order to identify enzymes whose inactivation is lethal only in B. burgdorferi, the predicted essential reactions from iBB151 were compared with those of iML1515 (40) (E. coli) and iYS854 (41) (S. aureus). Twenty-one reactions were predicted to be essential only in B. burgdorferi. A further seven were predicted to be partially specific: essential in only one of the comparator species. These 28 potential narrow-spectrum drug targets are listed in Table 1.

TABLE 1.

Selected essential enzymatic reactions in B. burgdorferia

B. burgdorferi (iBB151) E. coli (iML1515) S. aureus (iYS853)
Pathway Reaction ID Full name Reaction Gene Deletion growth ratio Reaction ID Deletion growth ratio Reaction ID Deletion growth ratio
ARG_D Arginine deiminase L-arginine + H2O ⇌ ammonium + L-citrulline BB_0841 0.00 nf ARGDr 1.00
SAH_N Adenosyl-
homocysteine nucleosidase
S-adenosylhomocysteine + H2O → adenine + S-ribosyl-L-homocysteine BB_0375/BB_0588/BB_I06 0.00 AHCYSNS 0.00 HCYSNS 1.00
PYDX_K Pyridoxal kinase ATP + pyridoxal → ADP + H+ + pyridoxal 5'-phosphate BB_0768 0.00 PDXK 1.00 PYDXK_1 1.00
Folates METTHF_DH Methylene-THF dehydrogenase Methylene-THF + NADP+ ⇌ methenyl-THF + NADPH BB_0026 0.00 MTHFD 0.99 MTHFD 0.98
MENTHF_CH Methylene-THF cyclohydrolase Methenyl-THF + H2O ⇌ formyl-THF + H+ BB_0026 0.00 MTHFC 0.99 MTHFC 0.98
SER_HMT Serine hydroxymethyl- transferase L-serine + THF ⇌ glycine + methylene-THF + H2O BB_0601 0.00 GHMT2r 0.99 GHMT2r 0.98
Glycolysis GAP_DH GAP dehydrogenase Glyceraldehyde 3-phosphate + NAD+ + phosphate ⇌ 3-phospho-glyceroyl-phosphate + NADH+ H+ BB_0057 0.00 GAPD 0.87 GAPD_1 0.00
PGLYC_K Phosphoglycerate kinase ATP + 3-phospho-glycerate ⇌ ADP + 3-phospho-glyceroyl-phosphate BB_0055/BB_0056 0.00 PGK 0.87 PGK 0.00
PGLYC_M Phosphoglycerate mutase 2-Phospho-glycerate ⇌ 3-phospho-glycerate BB_0658 0.00 PGM 0.93 PGM 0.78
ENO Enolase 2-Phospho-D-glycerate ⇌ phosphoenolpyruvate + H2O BB_0337 0.00 ENO 0.92 ENO 0.78
Lipids UGLUC_E UDP-glucose epimerase UDP-glucose ⇌ UDP-galactose BB_0444 0.00 UDPG4E 1.00 UDPG4E 1.00
PC_S Phosphatidyl- choline synthase CDP-diacylglycerol + choline → CMP + phosphatidylcholine + H+ BB_0249 0.00 nf nf
DAG_GT Diacylglycerol galactosyl- transferase CDP-diacylglycerol + UDP-galactose ⇌ galactosyldiacylglycerol + UDP BB_0454 0.00 nf nf
CHO_GT Cholesterol galactosyl- transferase Cholesterol + UDP-galactose ⇌ galactosylcholesterol BB_0572 0.00 nf nf
Mevalonate ACTCOA_AT Acetyl CoA acetyltransferase Two acetyl-CoA ⇌ acetoacetyl-CoA + CoA BB_0685 0.00 ACACT1r 0.99 ACACT1r 0.00
HMGCOA_S Hydroxymethyl-CoA synthase Acetoacetyl-CoA + acetyl CoA + H2O ⇌ (S)-3-hydroxy-3-methylglutaryl-CoA + CoA + H+ BB_0683 0.00 nf HMGCOAS
MEV_K Mevalonate kinase ATP + mevalonate ⇌ ADP + mevalonate 5-phosphate + H+ BB_0688 0.00 nf MEVK1_1,/MEVK2/MEVK3/MEVK4b 0.00
PMEV_K Phospho-mevalonate kinase ATP + mevalonate 5-phosphate ⇌ ADP + mevalonate diphosphate BB_0687 0.00 nf PMEVK 0.00
PPMEV_DC Mevalonate diphosphate decarboxylase ATP + mevalonate diphosphate → ADP + CO2 + isopentenyl diphosphate + phosphate BB_0686 0.00 nf DPMVD 0.00
Nucleotides IPP_I Isopentenyl diphosphate isomerase Isopentenyl diphosphate ⇌ dimethylallyl diphosphate BB_0684 0.00 IPDDI 1.00 IPDDI 0.00
DCYT_K Deoxycytidine kinase 2'-Deoxycytidine + ATP → dCMP + ADP + H+ BB_0239/BB_0128/BB_0791/BB_0015 0.00 nf nf
DADE_K Deoxyadenosine kinase ATP + 2'-deoxyadenosine → ADP + dAMP + H+ BB_0239/BB_0791/BB_0015 0.00 nf DADNK_1 1.00
AMP_K Adenylate kinase ATP +AMP → 2 ADP BB_0417 0.00 ADK1 1.00 ADK1/ADK2_1/ADKdc 1.00
DAMP_K Deoxyadenylate kinase ATP + dAMP → ADP + dADP BB_0417 0.00 DADK 1.00 DADK 1.00
TMP_K Thymidylate kinase dTMP + ATP → ADP + dTDP BB_0417/BB_0793 0.00 DTMPK 1.00 DTMPK 0.00
CMP_K Cytidylate kinase CMP + ATP → ADP + CDP BB_0417/BB_0128/BB_0819 0.00 CYTK1 1.00 CYTK1/CYTK1_1c 1.00
DCMP_K Deoxycytidylate kinase dCMP + ATP → ADP + dCDP BB_0417 0.00 CYTK2 1.00 CYTK2 0.00
DGMP_K Deoxyguanylate kinase dGMP + ATP → ADP + dGDP BB_0417 0.00 DGK1 1.00 DGK1/2 1.00
a

Reactions are predicted to be essential when an in silico deletion shows zero flux through the biomass reaction (growth ratio deletion:wild type = 0). Essential reactions highlighted bold. Most of the reactions shown are uniquely essential in B. burgdorferi, although some are also essential in E. coli or S. aureus. NF, reaction not found in organism: deletion growth ratio cannot be calculated (–) where reaction is not found. Complete list of essential reactions is found in supplemental file S5, “SRD.”

b

Any one of four mevalonate kinase reactions is required for growth.

c

No growth defect even if all homologs of adenylate/cytidylate kinase were deleted.

Enzyme inhibitors prevent growth of B. burgdorferi

Four reactions with known small molecule inhibitors were assayed for growth inhibition in each of the three species. Cycloserine targets alanine racemase (60), an enzyme predicted to be essential for cell wall biosynthesis in all three species investigated. Bromopyruvate is an inhibitor of several enzymes in the glycolytic Embden-Meyerhof-Parnas (EMP) pathway (66), which is known to be essential in S. aureus (67) and is the only known ATP-generating pathway in B. burgdorferi (6). In E. coli, the Entner-Doudouroff glycolytic pathway and fatty acid beta oxidation are alternatives to EMP glycolysis, both generating acetyl-CoA to drive ATP production via the tricarboxylic acid cycle. Like B. burgdorferi, S. aureus lacks either of these alternatives (68).

Theophylline and pemetrexed target pyridoxal kinase (69) and serine hydroxymethltransferase (70), respectively. Both enzymes are predicted to be uniquely essential to B. burgdorferi: E. coli and S. aureus possess redundant pathways for the synthesis of the essential products pyridoxal phosphate and methylene-tetrahydrofolate.

Experimentally determined minimum inhibitory concentrations (MICs; Table 2) match the predictions of reaction essentiality (Fig. 2). Cycloserine effectively inhibits all three species with similar MIC. The experimental MIC of cycloserine is fourfold higher in B. burgdorferi in than the other species. This variation may be due to nonmetabolic differences: transport, efflux, or degradation of the drug may vary between the species. Bromopyruvate inhibits the growth of EMP glycolysis-dependent species B. burgdorferi (MIC = 128 µg/mL) and S. aureus (MIC = 64 µg/mL) at concentrations which do not inhibit E. coli (MIC >512 µg/mL). Both theophylline (MIC = 1 mg/mL) and pemetrexed (MIC = 0.5 mg/mL) have specific activity against B. burgdorferi without inhibiting growth of either E. coli or S. aureus in the concentration range tested (MIC >8 mg/mL).

TABLE 2.

Predicted growth ratios (growth rate of knockout/growth rate of wild type, where 0 = no growth) and experimentally determined MIC for four reactions across three speciesa

Species (model) Reaction (ID) Predicted growth ratio (KO/WT) Compound MIC
B. burgdorferi (iBB151) Alanine racemase (ALA_R) 0 D-cycloserine 256 µg/mL
E. coli (iML1515) Alanine racemase (ALAR) 0 D-cycloserine 64 µg/mL
S. aureus (iYS854) Alanine racemase (ALAR) 0 D-cycloserine 64 µg/mL
B. burgdorferi (iBB151) GAPDH (GAP_DH) 0 Bromopyruvate 128 µg/mL
E. coli (iML1515) GAPDH (GAPD) 0.87 Bromopyruvate >512 µg/mL
S. aureus (iYS854) GAPDH (GAPD_1) 0 Bromopyruvate 64 µg/mL
B. burgdorferi (iBB151) Pyridoxal kinase (PYDX_K) 0 Theophylline 1 mg/mL
E. coli (iML1515) Pyridoxal kinase (PYDXK) 1 Theophylline >8 mg/mL
S. aureus (iYS854) Pyridoxal kinase (PYDXK_1) 1 Theophylline >8 mg/mL
B. burgdorferi (iBB151) Serine hydroxymethyltransferase (SER_HMT) 0 Pemetrexed 0.5 mg/mL
E. coli (iML1515) Serine hydroxymethyltransferase (GHMT2r) 0.98 Pemetrexed >8 mg/mL
S. aureus (iYS854) Serine hydroxymethyltransferase (GHMT2r) 0.98 Pemetrexed >8 mg/mL
a

Inhibitory concentrations were reached (highlighted bold) where in silico analysis predicted gene essentiality.

Fig 2.

Fig 2

Inhibition of growth of three bacterial species by four small molecule inhibitors. D-cycloserine (A) prevents growth of all three species at similar concentrations. Bromopyruvate (B) inhibits the glycolysis-dependent species, B. burgdorferi and S. aureus. Both pemetrexed (C) and theophylline (D) selectively inhibit B. burgdorferi. B. burgdorferi growth quantified by direct cell counts (cells/mL); E. coli and S. aureus growth, by optical density (OD600): values for all three were normalized to growth of an untreated control grown in parallel. Black line represents 10% of the growth of untreated controls: the cutoff for determination of MIC.

Enzyme inhibitors reduce viability in established B. burgdorferi cultures

Estimation of viability in B. burgdorferi is complicated by the fact that the species forms neither turbid cultures nor traditional colonies. We determined the effect of candidate drugs on stationary-phase B. burgdorferi using three complementary methods (Fig. 3). Enumeration of “colonies” (visible growth in solid media overlays) after serial dilution (3A) revealed reduction of >99.8% in viable cell counts with theophylline at 8 mg/mL (8× the MIC) and reduction by 84% at the MIC, 1 mg/mL. Pemetrexed had less bactericidal efficacy: even at 8× the MIC viability was only reduced ~72%.

Fig 3.

Fig 3

Killing of Borrelia burgdorferi by 20-h exposure to antibiotics. Cells were grown to stationary phase and incubated at 32°C for 20 hours with drugs (theophylline, T, and pemetrexed, P) at their MIC or eight times their MIC. Viability was normalized to that of untreated (UT) cells incubated in media only. After treatment, cells were diluted onto solid media overlay plates for enumeration of colony-forming units (A) or directly live/dead stained (B). Colonies were counted at 14 days: the lower limit of detection (pink line) was 2 × 105 cells or a 99.8% reduction from 1 × 108. At 8 mg/mL, all theophylline-treated cultures fell below the LOD. For live/dead staining, treated cells were immediately stained with SYTO 9 (Em 503 nm; shown blue) and propidium iodide (Em 615 nm; shown pink): all cells are stained by SYTO 9, while only dead cells with permeable membranes are stained by propidium iodide. Pink and blue cells were counted to yield % viable counts (B). P-values shown were calculated by one-way analysis of variance with Dunnett’s test: P-values not shown were >0.05. Example images in C show untreated (UT), heat-killed (HK), and theophylline (T 8.0)- and pemetrexed (P 4.0)-treated cells. Scale bars, 25 µm.

We also used a direct staining method to measure viability immediately after treatment. Propidium iodide is a membrane-impermeable dye and only stains cells in which membrane integrity has been lost, usually as a result of cell death. SYTO 9 is membrane permeable and stains viable cells in the absence of propidium iodide. This method produces lower estimates of killing than the CFU counting, but microscopy did reveal morphological changes after some treatments. Despite largely retaining membrane integrity, pemetrexed-treated cells were morphologically different (Fig. 3C), appearing longer and with a less regular helical shape. Greater viability in the direct staining assay may suggest that cell death occurs via a mechanism in which membrane integrity is not compromised or that treatment induces the formation of an intact and viable but slow-growing subpopulation (which appears dead in outgrowth assays but not by staining) in addition to killing some of the treated Borrelia. Both methods agree that treatment with theophylline at 8× the MIC has a significant bactericidal effect on stationary-phase B. burgdorferi. The lower concentrations of theophylline and pemetrexed at either concentration were less effective.

DISCUSSION

The model generated here represents the most complete synthesis of predictive and experimental data available to date. While iBB151 may not completely describe the metabolism of B. burgdorferi, it was sufficient to generate experimentally validated predictions. The model produced here annotated 208 reactions to 151 genes. Although many of these genes are predicted by other pipelines such as KEGG (29) or the NCBI’s prokaryotic genome annotation pipeline (71), the integration of multiple such pipelines with experimental data makes iBB151 more comprehensive than any single existing database. Two pipelines cross-referenced the B. burgdorferi genome to existing annotation databases. The MetaCyc pipeline looked for homology to any enzyme in the MetaCyc database, whereas the HMM pipeline probed the KEGG database. The MetaCyc metabolism database is manually curated and, therefore, smaller than the automated KEGG, and accordingly, our MetaCyc pipeline predicted fewest reactions. The KEGG database includes automated annotations of over 9,000 organisms, providing a larger (but less curated) set of enzyme sequences for the HMM pipeline. Our third annotation, the KEGG pipeline, pulled directly from KEGG’s automated annotation (72) of the B. burgdorferi genome. As the HMM pipeline is based on KEGG’s database, it is unsurprising that these two pipelines generated models with significant overlap. Each pipeline annotated a different but overlapping set of reactions, although 71 of 208 reactions were predicted by all three pipelines. These 71 are likely well conserved between Borrelia and other better-described species and thus predicted with greater confidence.

Prediction of enzymatic functions by homology to other sequences is confounded by the evolutionary divergence of the spirochetes, which leaves a high proportion of the genome annotated only as hypothetical proteins. Complete characterization of these proteins will require further experimental approaches, but B. burgdorferi is a fastidious organism with a slow growth rate and complex genome; these factors combine to make traditional studies of metabolism challenging. By developing and analyzing the novel in silico metabolic network described here, 28 anti-borrelial drug targets were predicted. Existing small molecule drugs were repurposed to target two of these B. burgdorferi enzymes and shown to have selective activity in culture.

The prediction of essential reactions is highly dependent on the availability of extracellular metabolites, analogous to an in silico growth medium. In vivo, the essentiality of reactions predicted by modeling here could be overcome by the uptake of products from a rich extracellular environment. For instance, the phospholipid synthases are predicted to be essential but could become nonessential due to passive uptake where the environment is abundant in phospholipids (55). The production of the cell wall diamino acid ornithine by arginine deiminase (BB_0841) is predicted essential here, but the enzyme has previously been inactivated both singly (49) and as part of a transposon library (57). Thus, the prediction of arginine deiminase essentiality is likely to reflect only the requirement for ornithine, which can be sourced either through the arginine deiminase system or by direct uptake from the environment. A modification of the model to supply ornithine as an extracellular metabolite renders the reaction nonessential. Similarly, there are examples of genes dispensable in culture but essential during infection, such as the nicotinamidase pncA (BB_E22) (73, 74). Matching in vitro data, BB_E22 is predicted nonessential here, due to the presence of nicotinamide mononucleotide (NMN) in the synthetic growth medium. However, a double gene deletion of BB_E22 and nicotinamide mononucleotide transport (transport_59) is lethal—nicotinamide mononucleotide provides an alternative substrate for NAD synthesis via NMN adenyltransferase. This double deletion effectively recreates the BB_E22 deletion in an environment with no extracellular NMN—poor availability of NAD/NMN in mammalian hosts may be the reason for in vivo essentiality of BB_E22. These examples illustrate the importance of the in silico growth medium in prediction of essential reactions. Modification of the growth medium based on nutrient availability in the various niches of the infective cycle may yield site-specific predictions of gene essentiality.

Many of the 77 genes predicted as essential represent candidate targets for the development of novel anti-borrelial antibiotics. Repurposing four existing inhibitors of related enzymes, predictions were validated in culture and the inhibitors shown to have activity against B. burgdorferi, with three of those exhibiting a degree of specificity. D-cycloserine is used as part of combination therapies against M. tuberculosis, but the other compounds tested are unsuitable for clinical use. Although tested extensively against cultured cancer cells, bromopyruvate has not been studied in clinical trials: its simple structure and broad range of cellular targets (75) raise concerns of off-target toxicity. Theophylline is rarely used for its original indication against asthma due to a narrow therapeutic index and reported toxicity (76). Like many anticancer drugs, pemetrexed also has a narrow therapeutic window and has significant associated toxicity (77). While the inhibitory concentrations determined here for theophylline and pemetrexed (1.0 mg/mL and 0.5 mg/mL) are high, efficacy is likely to be improved by modification of these repurposed drugs (both developed to target mammalian enzyme homologs) to bind their B. burgdorferi enzyme targets more efficiently and specifically. These modifications, and others to improve bioavailability and activity, will be the focus of future work. The ability of theophylline to kill even stationary-phase Borrelia suggests the possibility that the targeting of core metabolism may be effective even in established or persistent infections, where traditional antibiotics have limited efficacy.

The study of core metabolism is especially important in host-dependent pathogens such as B. burgdorferi, where the complex metabolic interactions between pathogen and host underpin persistence and pathogenesis. Furthermore, the in silico approach to drug target identification described here could be of particular value when applied to other fastidious organisms for which experimental approaches are difficult. The metabolic model generated may also facilitate further study of the core metabolism of B. burgdorferi and the complex interactions between the bacterium and its various animal hosts. Four essential reactions are validated here in vitro, and the two candidates for the development of specific anti-borrelial drugs will be further investigated to design more effective and tolerable inhibitors. Development of these targets into narrow-spectrum antimicrobials could reduce the incidence of Lyme disease by reservoir-targeted elimination of B. burgdorferi or as prophylaxis in high-risk groups.

Contributor Information

Peter J. Gwynne, Email: peter.gwynne@tufts.edu.

Anthony Fodor, University of North Carolina at Charlotte, Charlotte, North Carolina, USA.

SUPPLEMENTAL MATERIAL

The following material is available online at https://doi.org/10.1128/msystems.00835-23.

File S1. msystems.00835-23-s0001.docx.

Legends describing the other supplemental files.

DOI: 10.1128/msystems.00835-23.SuF1
File S2. msystems.00835-23-s0002.pdf.

Metabolic model overview showing essential reactions.

DOI: 10.1128/msystems.00835-23.SuF2
File S3. msystems.00835-23-s0003.pdf.

Metabolic model overview highlighting major pathways.

DOI: 10.1128/msystems.00835-23.SuF3
File S4. msystems.00835-23-s0004.xlsx.

Metabolic model in Excel format.

DOI: 10.1128/msystems.00835-23.SuF4
File S5. msystems.00835-23-s0005.xlsx.

Predictions of essential reactions and genes.

DOI: 10.1128/msystems.00835-23.SuF5

ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

  • 1. Schutzer SE, Fraser-Liggett CM, Casjens SR, Qiu W-G, Dunn JJ, Mongodin EF, Luft BJ. 2011. Whole-genome sequences of thirteen isolates of Borrelia burgdorferi. J Bacteriol 193:1018–1020. doi: 10.1128/JB.01158-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Fraser CM, Casjens S, Huang WM, Sutton GG, Clayton R, Lathigra R, White O, Ketchum KA, Dodson R, Hickey EK, Gwinn M, Dougherty B, Tomb JF, Fleischmann RD, Richardson D, Peterson J, Kerlavage AR, Quackenbush J, Salzberg S, Hanson M, van Vugt R, Palmer N, Adams MD, Gocayne J, Weidman J, Utterback T, Watthey L, McDonald L, Artiach P, Bowman C, Garland S, Fuji C, Cotton MD, Horst K, Roberts K, Hatch B, Smith HO, Venter JC. 1997. Genomic sequence of a Lyme disease spirochaete, Borrelia burgdorferi. Nature 390:580–586. doi: 10.1038/37551 [DOI] [PubMed] [Google Scholar]
  • 3. Casjens S, Palmer N, van Vugt R, Huang WM, Stevenson B, Rosa P, Lathigra R, Sutton G, Peterson J, Dodson RJ, Haft D, Hickey E, Gwinn M, White O, Fraser CM. 2000. A bacterial genome in flux: the twelve linear and nine circular extrachromosomal DNAs in an infectious isolate of the Lyme disease spirochete Borrelia burgdorferi. Mol Microbiol 35:490–516. doi: 10.1046/j.1365-2958.2000.01698.x [DOI] [PubMed] [Google Scholar]
  • 4. Kerstholt M, Netea MG, Joosten LAB. 2020. Borrelia burgdorferi hijacks cellular metabolism of immune cells: consequences for host defense. Ticks Tick Borne Dis 11:101386. doi: 10.1016/j.ttbdis.2020.101386 [DOI] [PubMed] [Google Scholar]
  • 5. Caimano MJ, Drecktrah D, Kung F, Samuels DS. 2016. Interaction of the Lyme disease spirochete with its tick vector. Cell Microbiol 18:919–927. doi: 10.1111/cmi.12609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Corona A, Schwartz I, Conway T, Cohen P. 2015. Borrelia burgdorferi: carbon metabolism and the tick-mammal enzootic cycle. Microbiol Spectr 3. doi: 10.1128/microbiolspec.MBP-0011-2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Pollack RJ, Telford SR, Spielman A. 1993. Standardization of medium for culturing Lyme disease spirochetes. J Clin Microbiol 31:1251–1255. doi: 10.1128/jcm.31.5.1251-1255.1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Barbour AG. 1984. Isolation and cultivation of Lyme disease spirochetes. Yale J Biol Med 57:521–525. [PMC free article] [PubMed] [Google Scholar]
  • 9. Heroldová M, Němec M, Hubálek Z. 1998. Growth parameters of Borrelia burgdorferi sensu stricto at various temperatures. Zentralblatt für Bakteriologie 288:451–455. doi: 10.1016/S0934-8840(98)80058-3 [DOI] [PubMed] [Google Scholar]
  • 10. Casjens SR, Gilcrease EB, Vujadinovic M, Mongodin EF, Luft BJ, Schutzer SE, Fraser CM, Qiu W-G. 2017. Plasmid diversity and phylogenetic consistency in the Lyme disease agent Borrelia burgdorferi. BMC Genomics 18:165. doi: 10.1186/s12864-017-3553-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Schwan TG, Burgdorfer W, Garon CF. 1988. Changes in infectivity and plasmid profile of the Lyme disease spirochete, Borrelia burgdorferi, as a result of in vitro cultivation. Infect Immun 56:1831–1836. doi: 10.1128/iai.56.8.1831-1836.1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gu C, Kim GB, Kim WJ, Kim HU, Lee SY. 2019. Current status and applications of genome-scale metabolic models. Genome Biol 20:121. doi: 10.1186/s13059-019-1730-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Yang JE, Park SJ, Kim WJ, Kim HJ, Kim BJ, Lee H, Shin J, Lee SY. 2018. One-step fermentative production of aromatic polyesters from glucose by metabolically engineered Escherichia coli strains. Nat Commun 9:79. doi: 10.1038/s41467-017-02498-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kim M, Yi JS, Lakshmanan M, Lee D-Y, Kim B-G. 2016. Transcriptomics-based strain optimization tool for designing secondary metabolite overproducing strains of Streptomyces coelicolor. Biotechnol Bioeng 113:651–660. doi: 10.1002/bit.25830 [DOI] [PubMed] [Google Scholar]
  • 15. Mishra P, Lee N-R, Lakshmanan M, Kim M, Kim B-G, Lee D-Y. 2018. Genome-scale model-driven strain design for dicarboxylic acid production in Yarrowia lipolytica. BMC Syst Biol 12:12. doi: 10.1186/s12918-018-0542-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Borah K, Kearney J-L, Banerjee R, Vats P, Wu H, Dahale S, Manjari Kasibhatla S, Joshi R, Bonde B, Ojo O, Lahiri R, Williams DL, McFadden J. 2020. GSMN-ML- a genome scale metabolic network reconstruction of the obligate human pathogen Mycobacterium leprae. PLoS Negl Trop Dis 14:e0007871. doi: 10.1371/journal.pntd.0007871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Driscoll TP, Verhoeve VI, Guillotte ML, Lehman SS, Rennoll SA, Beier-Sexton M, Rahman MS, Azad AF, Gillespie JJ. 2017. Wholly Rickettsia! reconstructed metabolic profile of the quintessential bacterial parasite of eukaryotic cells. mBio 8:e00859-17. doi: 10.1128/mBio.00859-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Cesur MF, Siraj B, Uddin R, Durmuş S, Çakır T. 2019. Network-based metabolism-centered screening of potential drug targets in Klebsiella pneumoniae at genome scale. Front Cell Infect Microbiol 9:447. doi: 10.3389/fcimb.2019.00447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Agren R, Mardinoglu A, Asplund A, Kampf C, Uhlen M, Nielsen J. 2014. Identification of anticancer drugs for hepatocellular carcinoma through personalized genome‐scale metabolic modeling. Mol Syst Biol 10:721. doi: 10.1002/msb.145122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Modi SR, Collins JJ, Relman DA. 2014. Antibiotics and the gut microbiota. J Clin Invest 124:4212–4218. doi: 10.1172/JCI72333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Aldeyab MA, Kearney MP, Scott MG, Aldiab MA, Alahmadi YM, Darwish Elhajji FW, Magee FA, McElnay JC. 2012. An evaluation of the impact of antibiotic stewardship on reducing the use of high-risk antibiotics and its effect on the incidence of Clostridium difficile infection in hospital settings. J Antimicrob Chemother 67:2988–2996. doi: 10.1093/jac/dks330 [DOI] [PubMed] [Google Scholar]
  • 22. Costelloe C, Metcalfe C, Lovering A, Mant D, Hay AD. 2010. Effect of antibiotic prescribing in primary care on antimicrobial resistance in individual patients: systematic review and meta-analysis. BMJ 340:c2096. doi: 10.1136/bmj.c2096 [DOI] [PubMed] [Google Scholar]
  • 23. Löfmark S, Jernberg C, Jansson JK, Edlund C. 2006. Clindamycin-induced enrichment and long-term persistence of resistant Bacteroides spp. and resistance genes. J Antimicrob Chemother 58:1160–1167. doi: 10.1093/jac/dkl420 [DOI] [PubMed] [Google Scholar]
  • 24. Nguyen CT, Cifu AS, Pitrak D. 2022. Prevention and treatment of Lyme disease. JAMA 327:772. doi: 10.1001/jama.2021.25302 [DOI] [PubMed] [Google Scholar]
  • 25. Dolan MC, Schulze TL, Jordan RA, Dietrich G, Schulze CJ, Hojgaard A, Ullmann AJ, Sackal C, Zeidner NS, Piesman J. 2011. Elimination of Borrelia burgdorferi and Anaplasma phagocytophilum in rodent reservoirs and Ixodes scapularis ticks using a doxycycline hyclate-laden bait. Am J Trop Med Hyg 85:1114–1120. doi: 10.4269/ajtmh.2011.11-0292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Dolan MC, Schulze TL, Jordan RA, Schulze CJ, Ullmann AJ, Hojgaard A, Williams MA, Piesman J. 2017. Evaluation of doxycycline-laden oral bait and topical fipronil delivered in a single bait box to control Ixodes scapularis (Acari: Ixodidae) and reduce Borrelia burgdorferi and Anaplasma phagocytophilum infection in small mammal reservoirs and host-seeking ticks. J Med Entomol 54:403–410. doi: 10.1093/jme/tjw194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Leimer N, Wu X, Imai Y, Morrissette M, Pitt N, Favre-Godal Q, Iinishi A, Jain S, Caboni M, Leus IV, Bonifay V, Niles S, Bargabos R, Ghiglieri M, Corsetti R, Krumpoch M, Fox G, Son S, Klepacki D, Polikanov YS, Freliech CA, McCarthy JE, Edmondson DG, Norris SJ, D’Onofrio A, Hu LT, Zgurskaya HI, Lewis K. 2021. A selective antibiotic for Lyme disease. Cell 184:5405–5418. doi: 10.1016/j.cell.2021.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Wang H, Marcišauskas S, Sánchez BJ, Domenzain I, Hermansson D, Agren R, Nielsen J, Kerkhoven EJ. 2018. RAVEN 2.0: a versatile toolbox for metabolic network reconstruction and a case study on Streptomyces coelicolor. PLOS Comput Biol 14:e1006541. doi: 10.1371/journal.pcbi.1006541 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. 2016. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res 44:D457–62. doi: 10.1093/nar/gkv1070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Caspi R, Billington R, Keseler IM, Kothari A, Krummenacker M, Midford PE, Ong WK, Paley S, Subhraveti P, Karp PD. 2020. The MetaCyc database of metabolic pathways and enzymes - a 2019 update. Nucleic Acids Res 48:D445–D453. doi: 10.1093/nar/gkz862 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Finn RD, Clements J, Eddy SR. 2011. HMMER web server: interactive sequence similarity searching. Nucleic Acids Res 39:W29–37. doi: 10.1093/nar/gkr367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Li W, Godzik A. 2006. Cd-hit: a fast program for clustering and comparing large sets of protein or nucleotide sequences. Bioinformatics 22:1658–1659. doi: 10.1093/bioinformatics/btl158 [DOI] [PubMed] [Google Scholar]
  • 33. Gherardini FC, Dulebohn DP, Bourret TJ, Richards CL. 2021. Metabolism and physiology of Borrelia, p 131–180. In Lyme disease and relapsing fever spirochetes: genomics, molecular biology, host interactions and disease pathogenesis. Caister Academic Press. doi: 10.21775/9781913652616.06 [DOI] [Google Scholar]
  • 34. Lieven C, Beber ME, Olivier BG, Bergmann FT, Ataman M, Babaei P, Bartell JA, Blank LM, Chauhan S, Correia K, Diener C, Dräger A, Ebert BE, Edirisinghe JN, Faria JP, Feist AM, Fengos G, Fleming RMT, García-Jiménez B, Hatzimanikatis V, van Helvoirt W, Henry CS, Hermjakob H, Herrgård MJ, Kaafarani A, Kim HU, King Z, Klamt S, Klipp E, Koehorst JJ, König M, Lakshmanan M, Lee D-Y, Lee SY, Lee S, Lewis NE, Liu F, Ma H, Machado D, Mahadevan R, Maia P, Mardinoglu A, Medlock GL, Monk JM, Nielsen J, Nielsen LK, Nogales J, Nookaew I, Palsson BO, Papin JA, Patil KR, Poolman M, Price ND, Resendis-Antonio O, Richelle A, Rocha I, Sánchez BJ, Schaap PJ, Sheriff RSM, Shoaie S, Sonnenschein N, Teusink B, Vilaça P, Vik JO, Wodke JAH, Xavier JC, Yuan Q, Zakhartsev M, Zhang C. 2020. MEMOTE for standardized genome-scale metabolic model testing. Nat Biotechnol 38:272–276. doi: 10.1038/s41587-020-0477-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Feist AM, Henry CS, Reed JL, Krummenacker M, Joyce AR, Karp PD, Broadbelt LJ, Hatzimanikatis V, Palsson BØ. 2007. A genome-scale metabolic reconstruction for Escherichia coli K-12 MG1655 that accounts for 1260 ORFs and thermodynamic information. Mol Syst Biol 3:121. doi: 10.1038/msb4100155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Stübs G, Fingerle V, Wilske B, Göbel UB, Zähringer U, Schumann RR, Schröder NWJ. 2009. Acylated cholesteryl galactosides are specific antigens of Borrelia causing Lyme disease and frequently induce antibodies in late stages of disease. J Biol Chem 284:13326–13334. doi: 10.1074/jbc.M809575200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Alexaki A, Kames J, Holcomb DD, Athey J, Santana-Quintero LV, Lam PVN, Hamasaki-Katagiri N, Osipova E, Simonyan V, Bar H, Komar AA, Kimchi-Sarfaty C. 2019. Codon and codon-pair usage tables (CoCoPUTs): facilitating genetic variation analyses and recombinant gene design. J Mol Biol 431:2434–2441. doi: 10.1016/j.jmb.2019.04.021 [DOI] [PubMed] [Google Scholar]
  • 38. Zhang K, Bian J, Deng Y, Smith A, Nunez RE, Li MB, Pal U, Yu A-M, Qiu W, Ealick SE, Li C. 2016. Lyme disease spirochaete Borrelia burgdorferi does not require thiamin. Nat Microbiol 2:16213. doi: 10.1038/nmicrobiol.2016.213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Heirendt L, Arreckx S, Pfau T, Mendoza SN, Richelle A, Heinken A, Haraldsdóttir HS, Wachowiak J, Keating SM, Vlasov V, Magnusdóttir S, Ng CY, Preciat G, Žagare A, Chan SHJ, Aurich MK, Clancy CM, Modamio J, Sauls JT, Noronha A, Bordbar A, Cousins B, El Assal DC, Valcarcel LV, Apaolaza I, Ghaderi S, Ahookhosh M, Ben Guebila M, Kostromins A, Sompairac N, Le HM, Ma D, Sun Y, Wang L, Yurkovich JT, Oliveira MAP, Vuong PT, El Assal LP, Kuperstein I, Zinovyev A, Hinton HS, Bryant WA, Aragón Artacho FJ, Planes FJ, Stalidzans E, Maass A, Vempala S, Hucka M, Saunders MA, Maranas CD, Lewis NE, Sauter T, Palsson BØ, Thiele I, Fleming RMT. 2019. Creation and analysis of biochemical constraint-based models using the COBRA toolbox V.3.0. Nat Protoc 14:639–702. doi: 10.1038/s41596-018-0098-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Monk JM, Lloyd CJ, Brunk E, Mih N, Sastry A, King Z, Takeuchi R, Nomura W, Zhang Z, Mori H, Feist AM, Palsson BO. 2017. iML1515, a knowledgebase that computes Escherichia coli traits. Nat Biotechnol 35:904–908. doi: 10.1038/nbt.3956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Seif Y, Monk JM, Mih N, Tsunemoto H, Poudel S, Zuniga C, Broddrick J, Zengler K, Palsson BO, Wallqvist A. 2019. A computational knowledge-base elucidates the response of Staphylococcus aureus to different media types. PLoS Comput Biol 15:e1006644. doi: 10.1371/journal.pcbi.1006644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Mongodin EF, Casjens SR, Bruno JF, Xu Y, Drabek EF, Riley DR, Cantarel BL, Pagan PE, Hernandez YA, Vargas LC, Dunn JJ, Schutzer SE, Fraser CM, Qiu W-G, Luft BJ. 2013. Inter- and intra-specific pan-genomes of Borrelia burgdorferi sensu lato: genome stability and adaptive radiation. BMC Genomics 14:693. doi: 10.1186/1471-2164-14-693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Brisson D, Drecktrah D, Eggers CH, Samuels DS. 2012. Genetics of Borrelia burgdorferi. Annu Rev Genet 46:515–536. doi: 10.1146/annurev-genet-011112-112140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Schwartz I, Margos G, Casjens SR, Qiu W-G, Eggers CH. 2021. Multipartite genome of Lyme disease Borrelia: structure, variation and prophages. Curr Issues Mol Biol 42:409–454. doi: 10.21775/cimb.042.409 [DOI] [PubMed] [Google Scholar]
  • 45. Hoon-Hanks LL, Morton EA, Lybecker MC, Battisti JM, Samuels DS, Drecktrah D. 2012. Borrelia burgdorferi malQ mutants utilize disaccharides and traverse the enzootic cycle. FEMS Immunol Med Microbiol 66:157–165. doi: 10.1111/j.1574-695X.2012.00996.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. von Lackum K, Stevenson B. 2005. Carbohydrate utilization by the Lyme borreliosis spirochete, Borrelia Burgdorferi. FEMS Microbiol Lett 243:173–179. doi: 10.1016/j.femsle.2004.12.002 [DOI] [PubMed] [Google Scholar]
  • 47. Schneider EM, Rhodes RG. 2018. N-acetylmannosamine (ManNAc) supports the growth of Borrelia burgdorferi in the absence of N-acetylglucosamine (GlcNAc). FEMS Microbiol Lett 365. doi: 10.1093/femsle/fny243 [DOI] [PubMed] [Google Scholar]
  • 48. Van Laar TA, Lin Y-H, Miller CL, Karna SLR, Chambers JP, Seshu J. 2012. Effect of levels of acetate on the mevalonate pathway of Borrelia burgdorferi. PLoS ONE 7:e38171. doi: 10.1371/journal.pone.0038171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Richards CL, Raffel SJ, Bontemps-Gallo S, Dulebohn DP, Herbert TC, Gherardini FC, Coburn J. 2022. The arginine deaminase system plays distinct roles in Borrelia burgdorferi and Borrelia hermsii. PLOS Pathog 18:e1010370. doi: 10.1371/journal.ppat.1010370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Beck G, Benach JL, Habicht GS. 1990. Isolation, preliminary chemical characterization, and biological activity of Borrelia burgdorferi peptidoglycan. Biochem Biophys Res Commun 167:89–95. doi: 10.1016/0006-291x(90)91734-a [DOI] [PubMed] [Google Scholar]
  • 51. Jutras BL, Lochhead RB, Kloos ZA, Biboy J, Strle K, Booth CJ, Govers SK, Gray J, Schumann P, Vollmer W, Bockenstedt LK, Steere AC, Jacobs-Wagner C. 2019. Borrelia burgdorferi peptidoglycan is a persistent antigen in patients with Lyme arthritis. Proc Natl Acad Sci U S A 116:13498–13507. doi: 10.1073/pnas.1904170116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Drecktrah D, Hall LS, Crouse B, Schwarz B, Richards C, Bohrnsen E, Wulf M, Long B, Bailey J, Gherardini F, Bosio CM, Lybecker MC, Samuels DS. 2022. The glycerol-3-phosphate dehydrogenases GpsA and GlpD constitute the oxidoreductive metabolic linchpin for Lyme disease spirochete host infectivity and persistence in the tick. PLOS Pathog 18:e1010385. doi: 10.1371/journal.ppat.1010385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Ramos D, Lasseter AG, Richards CL, Schwarz B, Ghosh S, Victoria B, Bosio CM, Gherardini FC, Jewett MW. 2022. Riboflavin salvage by Borrelia burgdorferi supports carbon metabolism and is essential for survival in the tick vector. Mol Microbiol 118:443–456. doi: 10.1111/mmi.14977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Groshong AM, Dey A, Bezsonova I, Caimano MJ, Radolf JD. 2017. Peptide uptake is essential for Borrelia burgdorferi viability and involves structural and regulatory complexity of its oligopeptide transporter. mBio 8:e02047-17. doi: 10.1128/mBio.02047-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Gwynne PJ, Clendenen LH, Turk S-P, Marques AR, Hu LT. 2022. Antiphospholipid autoantibodies in Lyme disease arise after scavenging of host phospholipids by Borrelia burgdorferi. J Clin Invest 132:e152506. doi: 10.1172/JCI152506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Jain S, Sutchu S, Rosa PA, Byram R, Jewett MW. 2012. Borrelia burgdorferi harbors a transport system essential for purine salvage and mammalian infection. Infect Immun 80:3086–3093. doi: 10.1128/IAI.00514-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Lin T, Gao L, Zhang C, Odeh E, Jacobs MB, Coutte L, Chaconas G, Philipp MT, Norris SJ, Stevenson B. 2012. Analysis of an ordered, comprehensive STM mutant library in infectious Borrelia burgdorferi: insights into the genes required for mouse infectivity. PLOS ONE 7:e47532. doi: 10.1371/journal.pone.0047532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Zapun A, Contreras-Martel C, Vernet T. 2008. Penicillin-binding proteins and β-lactam resistance. FEMS Microbiol Rev 32:361–385. doi: 10.1111/j.1574-6976.2007.00095.x [DOI] [PubMed] [Google Scholar]
  • 59. Yanagisawa T, Lee JT, Wu HC, Kawakami M. 1994. Relationship of protein structure of isoleucyl-tRNA synthetase with pseudomonic acid resistance of Escherichia coli. a proposed mode of action of pseudomonic acid as an inhibitor of isoleucyl-tRNA synthetase. J Biol Chem 269:24304–24309. [PubMed] [Google Scholar]
  • 60. Lambert MP, Neuhaus FC. 1972. Mechanism of d-cycloserine action: alanine racemase from Escherichia coli W1. J Bacteriol 110:978–987. doi: 10.1128/jb.110.3.978-987.1972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Wilding EI, Brown JR, Bryant AP, Chalker AF, Holmes DJ, Ingraham KA, Iordanescu S, So CY, Rosenberg M, Gwynn MN. 2000. Identification, evolution, and essentiality of the mevalonate pathway for isopentenyl diphosphate biosynthesis in gram-positive cocci. J Bacteriol 182:4319–4327. doi: 10.1128/JB.182.15.4319-4327.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Fisher SL. 2008. Glutamate racemase as a target for drug discovery. Microb Biotechnol 1:345–360. doi: 10.1111/j.1751-7915.2008.00031.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Chakraborti M, Schlachter S, Primus S, Wagner J, Sweet B, Carr Z, Cornell KA, Parveen N. 2020. Evaluation of nucleoside analogs as antimicrobials targeting unique enzymes in Borrelia burgdorferi. Pathogens 9:678. doi: 10.3390/pathogens9090678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Cornell KA, Primus S, Martinez JA, Parveen N. 2009. Assessment of methylthioadenosine/S-adenosylhomocysteine nucleosidases of Borrelia burgdorferi as targets for novel antimicrobials using a novel high-throughput method. J Antimicrob Chemother 63:1163–1172. doi: 10.1093/jac/dkp129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Van Laar TA, Hole C, Rajasekhar Karna SL, Miller CL, Reddick R, Wormley FL, Seshu J. 2016. Statins reduce spirochetal burden and modulate immune responses in the C3H/HeN mouse model of Lyme disease. Microbes Infect 18:430–435. doi: 10.1016/j.micinf.2016.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Shoshan MC. 2012. 3-bromopyruvate: targets and outcomes. J Bioenerg Biomembr 44:7–15. doi: 10.1007/s10863-012-9419-2 [DOI] [PubMed] [Google Scholar]
  • 67. Chaudhuri RR, Allen AG, Owen PJ, Shalom G, Stone K, Harrison M, Burgis TA, Lockyer M, Garcia-Lara J, Foster SJ, Pleasance SJ, Peters SE, Maskell DJ, Charles IG. 2009. Comprehensive identification of essential Staphylococcus aureus genes using transposon-mediated differential hybridisation (TMDH). BMC Genomics 10:291. doi: 10.1186/1471-2164-10-291 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Parsons JB, Frank MW, Subramanian C, Saenkham P, Rock CO. 2011. Metabolic basis for the differential susceptibility of gram-positive pathogens to fatty acid synthesis inhibitors. Proc Natl Acad Sci U S A 108:15378–15383. doi: 10.1073/pnas.1109208108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Lainé-Cessac P, Cailleux A, Allain P. 1997. Mechanisms of the inhibition of human erythrocyte pyridoxal kinase by drugs. Biochem Pharmacol 54:863–870. doi: 10.1016/s0006-2952(97)00252-9 [DOI] [PubMed] [Google Scholar]
  • 70. Scaletti E, Jemth A-S, Helleday T, Stenmark P. 2019. Structural basis of inhibition of the human serine hydroxymethyltransferase SHMT2 by antifolate drugs. FEBS Lett 593:1863–1873. doi: 10.1002/1873-3468.13455 [DOI] [PubMed] [Google Scholar]
  • 71. Li W, O’Neill KR, Haft DH, DiCuccio M, Chetvernin V, Badretdin A, Coulouris G, Chitsaz F, Derbyshire MK, Durkin AS, Gonzales NR, Gwadz M, Lanczycki CJ, Song JS, Thanki N, Wang J, Yamashita RA, Yang M, Zheng C, Marchler-Bauer A, Thibaud-Nissen F. 2021. RefSeq: expanding the prokaryotic genome annotation pipeline reach with protein family model curation. Nucleic Acids Res 49:D1020–D1028. doi: 10.1093/nar/gkaa1105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Kanehisa M, Sato Y, Morishima K. 2016. BlastKOALA and GhostKOALA: KEGG tools for functional characterization of genome and metagenome sequences. J Mol Biol 428:726–731. doi: 10.1016/j.jmb.2015.11.006 [DOI] [PubMed] [Google Scholar]
  • 73. Jewett MW, Jain S, Linowski AK, Sarkar A, Rosa PA. 2011. Molecular characterization of the Borrelia burgdorferi in vivo-essential protein PncA. Microbiology (Reading) 157:2831–2840. doi: 10.1099/mic.0.051706-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Purser JE, Lawrenz MB, Caimano MJ, Howell JK, Radolf JD, Norris SJ. 2003. A plasmid-encoded nicotinamidase (PncA) is essential for infectivity of Borrelia burgdorferi in a mammalian host. Mol Microbiol 48:753–764. doi: 10.1046/j.1365-2958.2003.03452.x [DOI] [PubMed] [Google Scholar]
  • 75. Ehrke E, Arend C, Dringen R. 2015. 3-bromopyruvate inhibits glycolysis, depletes cellular glutathione, and compromises the viability of cultured primary rat astrocytes. J Neurosci Res 93:1138–1146. doi: 10.1002/jnr.23474 [DOI] [PubMed] [Google Scholar]
  • 76. Wood AJJ, Weinberger M, Hendeles L. 1996. Theophylline in asthma. N Engl J Med 334:1380–1388. doi: 10.1056/NEJM199605233342107 [DOI] [PubMed] [Google Scholar]
  • 77. Niyikiza C, Baker SD, Seitz DE, Walling JM, Nelson K, Rusthoven JJ, Stabler SP, Paoletti P, Calvert AH, Allen RH. 2002. Homocysteine and methylmalonic acid: markers to predict and avoid toxicity from pemetrexed therapy 1. Mol Cancer Ther 1:545–552. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

File S1. msystems.00835-23-s0001.docx.

Legends describing the other supplemental files.

DOI: 10.1128/msystems.00835-23.SuF1
File S2. msystems.00835-23-s0002.pdf.

Metabolic model overview showing essential reactions.

DOI: 10.1128/msystems.00835-23.SuF2
File S3. msystems.00835-23-s0003.pdf.

Metabolic model overview highlighting major pathways.

DOI: 10.1128/msystems.00835-23.SuF3
File S4. msystems.00835-23-s0004.xlsx.

Metabolic model in Excel format.

DOI: 10.1128/msystems.00835-23.SuF4
File S5. msystems.00835-23-s0005.xlsx.

Predictions of essential reactions and genes.

DOI: 10.1128/msystems.00835-23.SuF5

Articles from mSystems are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES