ABSTRACT
New classes of antibiotics are needed to fight bacterial infections, and repurposing existing drugs as antibiotics may enable rapid deployment of new treatments. Screens for antibacterials have been traditionally performed in standard laboratory media, but bacterial pathogens experience very different environmental conditions during infection, including nutrient limitation. To introduce the next generation of researchers to modern drug discovery methods, we developed a course-based undergraduate research experience (CURE) in which undergraduate students screened a library of FDA-approved drugs for their ability, in a nutrient-poor medium, to prevent the growth of the human Gram-negative bacterial pathogen Salmonella enterica serovar Typhimurium. The nine drugs identified all disrupt DNA metabolism in bacteria and eukaryotes. One of the hit compounds, capecitabine, is a well-tolerated oncology drug that is administered orally, a preferred treatment route. We demonstrated that capecitabine is more effective at inhibiting S. Typhimurium growth in nutrient-limited than in standard rich microbiological broth, an explanation for why the antibiotic activity of this compound has not been previously recognized. Capecitabine is enzymatically converted to the active pyrimidine analogue, fluorouracil (5-FU), and Gram-positive bacteria, including Staphylococcus aureus, are significantly more sensitive to 5-FU than Gram-negative bacteria. We therefore tested capecitabine for efficacy in a murine model of S. aureus peritonitis. Oral capecitabine administration reduced the colonization of tissues and increased animal survival in a dose-responsive manner. Since capecitabine is inexpensive, orally available, and relatively safe, it may have utility for treatment of intractable Gram-positive bacterial infections.
IMPORTANCE As bacterial infections become increasingly insensitive to antibiotics, whether established, off-patent drugs could treat infections becomes an important question. At the same time, basic research has revealed that during infection, mammals starve pathogens for nutrients and, in response, bacteria dramatically alter their biology. Therefore, it may be fruitful to search for drugs that could be repurposed as antibiotics using bacteria grown with limited nutrients. This approach, executed with undergraduate student researchers, identified nine drugs known to interfere with the production and/or function of DNA. We further explored one of these drugs, capecitabine, a well-tolerated human oncology drug. Oral administration of capecitabine reduced infection with the human pathogen Staphylococcus aureus and increased survival in mice. These data suggest that capecitabine has potential as a therapy for patients with otherwise untreatable bacterial infections.
KEYWORDS: antibiotics, capecitabine, course-based undergraduate research experience (CURE), 5-fluorouracil (5-FU), nucleoside analog, repurposing, Salmonella enterica, Staphylococcus aureus
INTRODUCTION
Antimicrobial-resistant pathogens present a growing threat to human health. The rate of development of new antibiotics has not kept pace with the increasing incidence of resistance due to the difficulty of identifying new chemical classes of antimicrobials (1). Uncertainty as to whether new antibiotics could be priced in a manner that would recover research costs and earn profits also hinders development (2). A rapid, cost-effective alternative to new therapies for untreatable infections is to identify off-patent, well-known drugs with the potential to be repurposed as antibiotics (3, 4).
Historically, antibiotics have been identified and evaluated by screening for chemicals that prevent bacterial growth in microbiological media that are nutrient rich and support many rounds of replication. This approach identified most of the clinical antibiotic classes currently in use (5, 6). It has the advantage of a large window of detection and is seemingly logical because bacteria replicate rapidly during the acute stages of infection. However, prior to rapid replication, bacterial pathogens need to overcome nutritional immunity, a host defense mechanism that restricts microbial access to metabolic building blocks (7). For example, growth of the human Gram-negative bacterial pathogen S. enterica serovar Typhimurium in mammalian cells is limited by access to amino acids (8, 9) and to carbohydrates or lipids (10, 11). Bacteria respond to nutritional restrictions by reprograming gene expression and activating diverse metabolic and catabolic pathways that enable virulence (12–14). Researchers have applied this information by screening for compounds that have antimicrobial activity in nutrient-poor media (15, 16).
We aimed to combine the scientific advances in antibiotic discovery described above with recent developments in the science, technology, engineering, and math (STEM) education field. Student involvement in research during undergraduate education promotes retention across STEM, reduces the time to graduation, and fosters identity as a scientist (17–19). To capture these benefits, course-based undergraduate research experiences (CUREs) engage students in authentic research (18). CUREs provide students with a meaningful connection to research faculty at their institution (20) and increase the participation of research faculty in teaching (18). For faculty, offering a CURE related to their research increases satisfaction with teaching and encourages sustained engagement (20). We worked with a CURE to identify off-patent drugs with potential utility for treating bacterial infections. The CURE screened a drug library for compounds that inhibit S. Typhimurium growth under nutrient-limiting conditions and identified nine drugs that prevent S. Typhimurium replication and are known to interfere with bacterial DNA metabolism. Two of the drugs, capecitabine and floxuridine, are prodrugs for fluorouracil (5-FU) that are used in patients to treat cancers (21, 22).
5-FU is a pyrimidine analog with multiple effects on eukaryotic and bacterial cells. It is enzymatically converted into a series of active metabolites that incorporate into DNA (dUTP) and cause DNA damage (23). 5-FU derivatives are also incorporated into mRNA (5-FUTP), causing miscoding and defective protein production (24). The combined effects of 5-FU inhibit bacterial growth but are not bactericidal (25). As with many drugs, 5-FU is considerably more potent against Gram-positive than Gram-negative bacteria (25–29). For example, a comparison of Escherichia coli and Staphylococcus aureus clinical isolates reported 5-FU minimum inhibitory concentrations that differ by 100-fold: 250 μg/ml versus 2 to 4 μg/ml, respectively (25). The heightened sensitivity of Gram-positive bacteria to 5-FU and other drugs (30, 31) is consistent with their lack of a protective outer membrane barrier (32, 33).
Floxuridine and one other 5-FU prodrug, 5-fluoro-2′-deoxyuridine (FdUrd), mitigate murine infection with S. aureus, a major human Gram-positive bacterial pathogen that causes skin and soft-tissue infections and forms biofilms on implants (34). Mice inoculated intraperitoneally (i.p.) with S. aureus have improved survival upon i.p. treatment with floxuridine starting 1 h after infection and continuing daily for 7 days (35). Similarly, neutropenic mice inoculated i.p. with S. aureus have improved survival upon i.p. treatment with FdUrd starting 1 h after infection and continuing daily for 3 days (36). Since floxuridine and FdUrd have antibacterial activity in vivo against S. aureus but are not, like capecitabine, orally available (37), we studied capecitabine further.
Capecitabine was developed in the late 1990s and is used to treat colon, breast, and gastric cancers (21, 37, 38). It is not by itself considered antibacterial and requires three metabolic steps for conversion to 5-FU (29, 39). To validate the identification of capecitabine as an inhibitor of S. Typhimurium growth, we compared growth in the presence of capecitabine in the minimal medium versus a rich medium. Since S. aureus is more sensitive to capecitabine and other 5-FU prodrugs than S. Typhimurium (25–28, 35, 36, 40), we determined whether capecitabine delivered orally mitigates S. aureus systemic infection in mice.
RESULTS
Screen of drug library for inhibition of bacterial replication in a nutrient-poor medium.
To identify drugs with antibacterial activity under nutrient-limiting conditions, we worked with students in a CURE to screen a library of 129 drugs approved by the United States Food and Drug Administration (FDA), the Oncology Drug Set VII from the National Cancer Institute. We used a virulent strain of S. Typhimurium (SL1344) grown overnight in a rich medium, lysogeny broth (LB) (41, 42). Cultures were diluted in M9 minimal medium supplemented with 2% glucose. Students added bacteria into each well of a 96-well plate, followed by the addition of library compounds to a final concentration of 10 μM. Control samples included vehicle and ampicillin. Plates were incubated at 37°C without shaking for 24 h, and the optical density at 620 nm (OD620) was measured to estimate bacterial growth. Vehicle-treated bacteria reached an average OD620 of 0.192, whereas ampicillin-treated bacteria achieved an OD620 of only 0.05 (Fig. 1). As expected, most library compounds had little or inconsistent effects on growth (see Table S1 in the supplemental material). Nine compounds consistently prevented bacterial growth, as defined by the cutoff of two standard deviations from the mean absorbance of the vehicle control. All nine hit compounds are drugs that interfere with DNA metabolism and have been used or explored as human cancer therapies (Table 1). Five are pyrimidines or pyrimidine prodrugs that inhibit DNA synthesis (azacytidine, capecitabine, decitabine, floxuridine, and gemcitabine). Plicamycin cross-links DNA, and carboplatin is a prodrug for a DNA cross-linker. Mitomycin alkylates DNA and also inhibits cellular thioredoxin reductase (43). Bleomycin is a mixture of glycopeptides that cleave DNA (44–46). Only one hit compound, capecitabine, is routinely administered to humans via an oral route (37). We therefore pursued this compound further.
FIG 1.
Screen of a library of FDA-approved drugs by CURE students for the ability to inhibit S. Typhimurium growth in a nutrient-poor medium identified nine drugs. Shown are mean and SEM values at an OD620 of 2 to 5 after 24 h of compound exposure in M9 minimal medium with 2% glucose at 37°C in static cultures. Vehicle (DMSO at 5%; blue circle) and ampicillin (50 μg/ml, 143 μM; green triangle) were used. Compounds are arranged alphabetically. The nine hits (red circles) are azacytidine, bleomycin, capecitabine, carboplatin, decitabine, floxuridine, gemcitabine, mitomycin, and plicamycin. Blue horizontal dashed lines represent two standard deviations from the means of the vehicle control, the cutoff for identifying a hit. The hits, DMSO, ampicillin, and several nonhit compounds have error bars that are too small to be visualized on the graph.
TABLE 1.
Hit compounds identified as inhibitors of S. Typhimurium growth in M9 medium
| Compound | Mean OD600 | SEM | Mechanism of action; target, if known | Delivery routea |
|---|---|---|---|---|
| Vehicle (DMSO) | 0.192 | 0.003 | NA | NA |
| Ampicillin (control) | 0.040 | 0.000 | Inhibits cell wall synthesis | Oral |
| 13b Azacytidine | 0.036 | 0.000 | Inhibits DNA synthesis; pyrimidine, incorporates into DNA | i.v., s.c. |
| 16 Bleomycin | 0.037 | 0.001 | Inhibits DNA synthesis; unclear | i.m., i.p., i.v., s.c. |
| 22 Capecitabine | 0.046 | 0.010 | Inhibits DNA synthesis; prodrug for pyrimidine 5-FU, inhibitor of thymidine synthase | Oral, i.v. |
| 23 Carboplatin | 0.036 | 0.001 | Inhibits DNA synthesis; prodrug for cross-linker of DNA | i.v. |
| 40 Decitabine | 0.040 | 0.002 | Inhibits DNA synthesis; pyrimidine, incorporates into DNA | i.v. |
| 51 Floxuridine | 0.036 | 0.001 | Inhibits DNA synthesis; prodrug for pyrimidine 5-FU, inhibitor of thymidine synthase | i.v. |
| 56 Gemcitabine | 0.052 | 0.014 | Inhibits DNA synthesis; pyrimidine, incorporates into DNA | i.v. |
| 78 Mitomycin | 0.042 | 0.000 | Inhibits DNA synthesis; alkylates DNA, also other cellular targets | i.v. |
| 95 Plicamycin | 0.044 | 0.001 | Inhibits DNA synthesis; complexes with DNA and RNA | i.v. |
i.m., intramuscular; i.p., intraperitoneal; i.v., intravenous; s.c., subcutaneous; NA, not applicable.
Numbers correspond to the x axis in Fig. 1 and column A in Table S1.
Nutrient limitation sensitizes S. Typhimurium to growth inhibition by capecitabine.
To establish whether we identified capecitabine because we screened in a nutrient-poor medium, we compared S. Typhimurium growth in the presence of capecitabine in M9 with 2% glucose to LB. S. Typhimurium was considerably more sensitive to capecitabine at 10 and 100 μM in M9 than in LB (Fig. 2A and B). We also compared the effect of a dose range of capecitabine on growth in M9 versus LB under conditions that as closely as possible mimicked the screen: resuspension of capecitabine in dimethyl sulfoxide (DMSO) instead of water, growth at 37°C without shaking, and reading of the plate on the spectrophotometer used for the screen, which has a 620-nm filter (Fig. 2C). Capecitabine was not effective at inhibiting S. Typhimurium growth in LB until concentrations of 100 μM were reached, whereas 12.5 μM capecitabine inhibited growth in M9. These observations suggest that experiments performed in rich media are unlikely to reveal the antibacterial activity of capecitabine. We conclude that under nutrient-limited conditions, S. Typhimurium is sensitive to growth inhibition by capecitabine and that the use of a nutrient-poor medium in the screen facilitated the identification of this drug.
FIG 2.
Capecitabine is more effective at inhibiting the growth of S. Typhimurium in nutrient-poor than in rich media. (A and B) S. Typhimurium was grown in M9 minimal medium with 2% glucose (A) and LB (B) at 37°C with aeration in the presence of the capecitabine concentrations indicated. OD600 was monitored hourly on a plate reader. Error bars represent SEM from four independent clones tested in triplicate. Vehicle (10% water) and ampicillin (50 μg/ml) were used. (C) Endpoint experiment performed under conditions mimicking the screen. S. Typhimurium was grown at 37°C standing, and OD620 was monitored after 24 h. Vehicle (5% DMSO) and ampicillin (50 μg/ml) were used. Means with SEM from at least 6 biological replicates tested in triplicate are shown. ***, P < 0.0001; **, P < 0.005; *, P < 0.05, as established by analysis of variance with a Tukey-Kramer honestly significant difference.
Capecitabine reduces S. aureus colonization of tissues in mice.
5-FU is more potent against Gram-positive than Gram-negative bacteria (25–29), and two 5-FU prodrugs improve survival in S. aureus-infected mice (35, 36). Therefore, we tested capecitabine against S. aureus infection of mice. We used a clinical S. aureus human wound isolate (ATCC 6538) that is virulent in mice (47–49). S. aureus was inoculated intravenously into BALB/c mice at 5 × 107 CFU, as verified by plating on a selective medium (50–52). Capecitabine was delivered orally every 12 h over 5 days at 100 or 200 mg/kg of body weight. The dose of 400 mg/kg/day is the maximum tolerated by mice (53). We scored mice daily for health on the basis of coat, posture, and movement (54) (Fig. 3A). On day four, one mouse in the control group died, and its tissues were collected, homogenized, and plated to confirm colonization with S. aureus. After 5 days, remaining mice were euthanized. Tissues were harvested, weighed, homogenized, and plated for CFU enumeration. Mice treated with the higher dose of capecitabine had significantly better health scores and reduced bacterial tissue colonization compared to mock-treated mice (Fig. 3B). These findings indicate that capecitabine ameliorates acute S. aureus infection of mice.
FIG 3.
Capecitabine improved health and reduced tissue colonization of S. aureus-infected mice. BALB/c female mice (8 to 9 weeks old) were infected intravenously with 5 × 107 CFU of S. aureus. Mice were treated orally with capecitabine at the dosage indicated or with sterile water (200 μl) every 12 h for 5 days. (A) Health was monitored every 24 h. Scores were summed to a maximum of six, and higher scores represent an increased incidence of morbid signs: movement (0 to 3), posturing (0 to 2), and coat condition (0 to 1). Means with standard deviations from 4 to 5 mice are shown. *, P < 0.01, relative to the corresponding mock-treated sample, as established by analysis of variance with a Tukey-Kramer honestly significant difference. (B) Tissue colonization. Each circle represents a tissue from a mouse, and 10 CFU/g was the limit of detection. Open circles show the number of CFU from the mock-treated mouse that died on day 4, although this mouse was excluded from the geometric mean (gray bars) and statistical analysis. *, P < 0.05, Mann-Whitney U test, two-tailed.
DISCUSSION
Here, we describe working with a CURE to screen for drugs that inhibit the growth of a Gram-negative bacterial pathogen, S. Typhimurium. We performed the screen in a nutrient-poor medium, whereas screens for new antibiotics are more typically performed in a richer medium, such as MHB, which was developed to optimize the growth of Neisseria species (6, 55). We identified multiple drugs known to interfere with DNA metabolism. Capecitabine was the only drug identified that is orally available and, therefore, of practical use for repurposing. We demonstrated that capecitabine is more potent against S. Typhimurium in minimal than in a standard rich medium, indicating that screening in nutrient-poor media facilitated the identification of capecitabine. These observations highlight that screens for new antibacterials performed under conditions that mimic infection identify different compounds than screens performed in traditional media (15, 56–58). They also underscore the value of working with students in a CURE format on screens of biological interest (59, 60).
Capecitabine is a 5-FU prodrug (21). Metabolites of 5-FU slow bacterial growth by interfering with DNA and RNA structure (23, 24). Two other 5-FU prodrugs, floxuridine and FdUrd, have been demonstrated to mitigate infection in mice inoculated with the Gram-positive pathogen S. aureus (35, 36). 5-FU is approximately 100-fold more potent at inhibiting the growth of Gram-positive than Gram-negative bacteria (25–29). For these reasons, we tested capecitabine in mice inoculated with S. aureus, which demonstrated that capecitabine treatment reduced tissue colonization and increased murine survival.
The potential for S. aureus to evolve resistance to capecitabine is not practical to test in broth culture, because S. aureus is not growth inhibited by capecitabine at concentrations up to 200 μM (39). However, S. aureus 5-FU-resistant isolates evolved in MHB harbor mutations in genes consistent with the roles of 5-FU in disrupting DNA and RNA metabolism. The corresponding mutated proteins convert 5-FU into toxic pyrimidine analogs or increase concentrations of uracil, which competes with 5-FU analogs and thereby relieves toxicity (29). Resistant mutants grew similarly to the wild-type parent strain in broth, suggesting similar in vitro fitness levels (29). Whether the mutants are still able to cause infection in whole animals and remain resistant to 5-FU and/or capecitabine is unknown but seems unlikely given the centrality of the mutated genes to nucleotide metabolism and, therefore, bacterial replication.
Selection for bacterial mutants resistant to capecitabine in mice is similarly not feasible due to differences between capecitabine metabolism in primates and mice. Capecitabine is well absorbed in the murine and primate small intestine (61, 62). However, in murine intestinal epithelial cells, capecitabine is converted to 5-FU at rates at least 20 times higher than those in monkey or human intestinal cells (63). As a result, oral capecitabine causes severe nuclear degradation of intestinal crypt cells in mice at plasma 5′-DFUR concentrations that are benign in cynomolgus monkeys (63, 64). In primates, capecitabine travels from the intestine to hepatocytes, where the first two steps of catabolism occur. The third metabolic step, production of 5-FU, is catalyzed in many cell types (21, 37). Thus, the cytotoxicity of 5-FU is, in effect, distributed across the human body, mostly affecting rapidly dividing cells, such as in tumors. Mice are more than 6-fold less tolerant to oral capecitabine than humans: oncology patients typically receive 2,500 mg/kg/day of oral capecitabine, whereas mice tolerate only 400 mg/kg/day (37, 53, 65, 66). Thus, it would not be possible to select for resistant S. aureus mutants in mice at capecitabine dosages equivalent to those received by patients.
In humans, capecitabine oral administration is well tolerated, and toxic effects do not generally become limiting for months (67). The most common toxicities caused by sustained treatment with capecitabine or 5-FU are cumulative and due to damage to rapidly dividing cells. Dose-limiting toxicities include diarrhea, cytopenias, hyperbilirubinemia, and cutaneous damage, such as hand-foot syndrome (palmar-plantar erythrodysesthesia) (67). Some patients experience central neurotoxicity and/or acute cardiotoxicity (68). However, over the short time frames consistent with antibiotic treatment, days to weeks, toxicity is rare (67).
Capecitabine tolerability and efficacy against cancer in humans is influenced by genetic polymorphisms in pyrimidine metabolic and degradative enzymes (37). Infection therapy would likely be subject to the same polymorphisms, but limitations on time and resources will likely make it difficult to a priori identify patients with reduced tolerability. However, there is an approved treatment for capecitabine overdose, uridine triacetate, which competes with the 5-FU derivative fluorouridine triphosphate for incorporation into RNA (69, 70). We conclude that due to its off-patent status, oral formulation, high tolerability, and evidence for efficacy against S. aureus infection in mice, capecitabine merits exploration as a potential antibiotic therapy or cotherapy for difficult-to-treat Gram-positive bacterial infections.
MATERIALS AND METHODS
Bacterial strains and media.
S. Typhimurium strain (SL1344) is a virulent calf isolate (71). SL1344 was grown in LB (41, 42) with 30 μg/ml streptomycin or in M9 medium (26.11 mM Na2HPO4·7H2O, 22.04 mM KH2PO4, 18.70 mM NH4Cl, 8.56 mM NaCl, 0.1 mM CaCl2, 63 μM MgSO4) with 2% glucose and 0.04% histidine (SL1344 is a histidine auxotroph). S. aureus strain FDA209 (ATCC 6538) is a human wound isolate that was established as a standard strain for testing antiseptics by the Insecticide and Fungicide Division of the U.S. Food and Drug Administration in 1938 (72, 73). Mannitol salt-selective agar (MSA) plates with phenol red were used to identify and enumerate S. aureus (74).
Screen.
The primary screen was performed by undergraduate researchers within a CURE class (75). The library was the Approved Oncology Drug Set VII from the National Cancer Institute of the United States/National Institutes of Health Developmental Therapeutics Program (http://dtp.cancer.gov). We screened barcoded plates 4845 and 4846 (https://dtp.cancer.gov/organization/dscb/obtaining/available_plates.htm). The library was solubilized in DMSO (Sigma-Aldrich) at 10 mM and stored at −20°C.
S. Typhimurium cultures were grown overnight at 37°C in LB with aeration. Cultures were diluted 100-fold to approximately 5 × 106 CFU/ml in M9. To each 96-well round-bottomed test plate, 90 μl of diluted culture was added. Library compounds (10 mM) were diluted 1:100 into 50% DMSO and 10 μl was added to the plate, for final concentrations of 10 μM compound and 5% DMSO. Controls within each plate included DMSO and ampicillin (50 μg/ml; 143 μM). The concentration of ampicillin was chosen based on the need for a robust positive control given varied student pipetting techniques (59); we used the concentration of ampicillin routinely used to prevent the growth of S. Typhimurium in the laboratory (76). Plates were grown statically for 24 h at 37°C, and the OD620 was the measured with an accuSkan FC microplate photometer (ThermoFisher Scientific, Waltham, MA). Biological replicates were independently performed by 2 to 5 student researchers. Outliers for individual compounds were determined using a Grubs test and eliminated. Compounds with values more than two standard deviations below the DMSO control were deemed hits.
Growth curves and endpoint analyses.
S. Typhimurium cultures were grown overnight in LB at 37°C with aeration. Cultures were diluted to ∼5 × 106 bacteria/ml into M9 or LB. For the growth curve experiments, culture volumes of 180 μl were added to a flat-bottomed 96-well plate. Capecitabine was freshly diluted in water, and 20 μl volumes were added to reach the final concentrations indicated. Plates were grown for 24 h at 37°C with shaking, and the OD600 was measured hourly in a Synergy 2 multi-mode or Eon microplate reader (BioTek, Winooski, VT). Mean OD600 and SEM from four biological replicates tested in triplicate were calculated. For the endpoint analysis experiments, S. Typhimurium cultures were grown overnight at 37°C in LB with aeration. Cultures were diluted 100-fold to approximately 5 × 106 CFU/ml in M9 or in LB. Diluted culture (90 μl) was added to a 96-well round-bottomed plate. Capecitabine was freshly diluted in 50% DMSO and 10 μl was added to culture wells to reach the final concentrations indicated with 5% DMSO. Plates were grown statically for 24 h at 37°C, and the OD620 was measured with an accuSkan FC microplate photometer (ThermoFisher Scientific, Waltham, MA).
Ethics statement.
Animal work was carried out in accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health (77). Euthanasia was carried out by carbon dioxide asphyxiation followed by cervical dislocation. All protocols were approved by Institutional Committee for Animal Care and Use (protocol number 2445) at the University of Colorado, Boulder, an Association for Assessment and Accreditation of Laboratory Animal Care (001743) accredited institution.
Infection of mice.
Female BALB/c mice were purchased from Taconic Biosciences (Rensselaer, NY) and acclimated for 1 week. S. aureus were grown for 18 h overnight at 37°C in LB medium with aeration and diluted to ∼5 × 107 CFU/ml in phosphate-buffered saline (PBS) prior to inoculation. Mice that were 8 to 9 weeks old were weighed and then infected intravenously with ∼5 × 107 CFU of S. aureus, as determined by plating onto MSA and enumeration of CFU. Mice were treated orally by gavage with 200 mg/kg or 100 mg/kg capecitabine in 200 μl of water every 12 h for 5 days. Capecitabine was prepared 1 h prior to delivery by suspension in sterile water. Control animals were mock treated with water. Prior to dosing, visual health assessments were performed on the basis of movement (0 for normal, 1 for slow movement, 2 for movement after prodding, 3 for no movement with prodding), posture (0 for normal, 1 for hunched, 2 for prostrate), and coat health (0 for normal, 1 for a ruffled coat) (54). Scores were summed to a maximum of 6. Mice that succumbed to infection were scored as 6. Tissues were harvested, weighed, and homogenized in 3 ml of PBS. Diluted tissues were plated onto MSA and grown overnight at 37°C to enumerate CFU/ml and/or establish the presence of S. aureus in all tissues harvested. After 5 days of treatment, remaining mice were euthanized and tissues were similarly harvested and processed.
Statistical analysis.
Statistical analysis was performed with GraphPad Prism 8 and with JMP version 15.
Supplementary Material
ACKNOWLEDGMENTS
We thank T. Su, L. Niswander, and M. Winey for support of the CURE laboratory, T. Mufford for expert assistance with animal infection experiments, and J. Dombach and C. Ewing for feedback on the manuscript.
The CURE was supported by a grant to Deborah Wuttke from the Howard Hughes Medical Institute and by the University of Colorado, Boulder, Department of Molecular, Cellular and Developmental Biology and the College of Arts and Sciences. Additional support was provided by NIH grant AI121365 (C.S.D.).
We declare no conflict of interest.
Footnotes
Supplemental material is available online only.
Contributor Information
Pamela A. Harvey, Email: pamela.harvey@colorado.edu.
Corrella S. Detweiler, Email: detweile@colorado.edu.
S. Wesley Long, Houston Methodist Hospital.
REFERENCES
- 1.Theuretzbacher U, Gottwalt S, Beyer P, Butler M, Czaplewski L, Lienhardt C, Moja L, Paul M, Paulin S, Rex JH, Silver LL, Spigelman M, Thwaites GE, Paccaud J-P, Harbarth S. 2019. Analysis of the clinical antibacterial and antituberculosis pipeline. Lancet Infect Dis 19:e40–e50. doi: 10.1016/S1473-3099(18)30513-9. [DOI] [PubMed] [Google Scholar]
- 2.Årdal C, Balasegaram M, Laxminarayan R, McAdams D, Outterson K, Rex JH, Sumpradit N. 2020. Antibiotic development–economic, regulatory and societal challenges. Nat Rev Microbiol 18:267–274. doi: 10.1038/s41579-019-0293-3. [DOI] [PubMed] [Google Scholar]
- 3.Kinnings SL, Liu N, Buchmeier N, Tonge PJ, Xie L, Bourne PE. 2009. Drug discovery using chemical systems biology: repositioning the safe medicine Comtan to treat multi-drug and extensively drug resistant tuberculosis. PLoS Comput Biol 5:e1000423. doi: 10.1371/journal.pcbi.1000423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chopra S, Torres-Ortiz M, Hokama L, Madrid P, Tanga M, Mortelmans K, Kodukula K, Galande AK. 2010. Repurposing FDA-approved drugs to combat drug-resistant Acinetobacter baumannii. J Antimicrob Chemother 65:2598–2601. doi: 10.1093/jac/dkq353. [DOI] [PubMed] [Google Scholar]
- 5.Mohr KI. 2016. History of antibiotics research. Curr Top Microbiol Immunol 398:237–272. doi: 10.1007/82_2016_499. [DOI] [PubMed] [Google Scholar]
- 6.Nizet V. 2017. The accidental orthodoxy of Drs. Mueller and Hinton. EBioMedicine 22:26–27. doi: 10.1016/j.ebiom.2017.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hood MI, Skaar EP. 2012. Nutritional immunity: transition metals at the pathogen-host interface. Nat Rev Microbiol 10:525–537. doi: 10.1038/nrmicro2836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hamilton S, Bongaerts RJM, Mulholland F, Cochrane B, Porter J, Lucchini S, Lappin-Scott HM, Hinton JCD. 2009. The transcriptional programme of Salmonella enterica serovar Typhimurium reveals a key role for tryptophan metabolism in biofilms. BMC Genomics 10:599. doi: 10.1186/1471-2164-10-599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Popp J, Noster J, Busch K, Kehl A, Zur Hellen G, Hensel M. 2015. Role of host cell-derived amino acids in nutrition of intracellular Salmonella enterica. Infect Immun 83:4466–4475. doi: 10.1128/IAI.00624-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Eisele NA, Ruby T, Jacobson A, Manzanillo PS, Cox JS, Lam L, Mukundan L, Chawla A, Monack DM. 2013. Salmonella require the fatty acid regulator PPARδ for the establishment of a metabolic environment essential for long-term persistence. Cell Host Microbe 14:171–182. doi: 10.1016/j.chom.2013.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Reens AL, Nagy TA, Detweiler CS. 2019. Salmonella enterica requires lipid metabolism genes to replicate in proinflammatory macrophages and mice. Infect Immun 88:e00776-19. doi: 10.1128/IAI.00776-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kröger C, Colgan A, Srikumar S, Händler K, Sivasankaran SK, Hammarlöf DL, Canals R, Grissom JE, Conway T, Hokamp K, Hinton JCD. 2013. An infection-relevant transcriptomic compendium for Salmonella enterica Serovar Typhimurium. Cell Host Microbe 14:683–695. doi: 10.1016/j.chom.2013.11.010. [DOI] [PubMed] [Google Scholar]
- 13.Dastgheyb SS, Otto M. 2015. Staphylococcal adaptation to diverse physiologic niches: an overview of transcriptomic and phenotypic changes in different biological environments. Future Microbiol 10:1981–1995. doi: 10.2217/fmb.15.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Richardson AR. 2019. Virulence and metabolism. Microbiol Spectr 7. doi: 10.1128/microbiolspec.GPP3-0011-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Miller CH, Nisa S, Dempsey S, Jack C, O'Toole R. 2009. Modifying culture conditions in chemical library screening identifies alternative inhibitors of mycobacteria. Antimicrob Agents Chemother 53:5279–5283. doi: 10.1128/AAC.00803-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.El Zahed SS, Kumar G, Tong M, Brown ED. 2018. Nutrient stress small-molecule screening platform for Escherichia coli. Methods Mol Biol 1787:1–18. doi: 10.1007/978-1-4939-7847-2_1. [DOI] [PubMed] [Google Scholar]
- 17.Russell SH, Hancock MP, McCullough J. 2007. The pipeline. Benefits of undergraduate research experiences. Science 316:548–549. doi: 10.1126/science.1140384. [DOI] [PubMed] [Google Scholar]
- 18.Auchincloss LC, Laursen SL, Branchaw JL, Eagan K, Graham M, Hanauer DI, Lawrie G, McLinn CM, Pelaez N, Rowland S, Towns M, Trautmann NM, Varma-Nelson P, Weston TJ, Dolan EL. 2014. Assessment of course-based undergraduate research experiences: a meeting report. CBE Life Sci Educ 13:29–40. doi: 10.1187/cbe.14-01-0004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Eagan MK, Hurtado S, Chang MJ, Garcia GA, Herrera FA, Garibay JC. 2013. Making a difference in science education: the impact of undergraduate research programs. Am Educ Res J 50:683–713. doi: 10.3102/0002831213482038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shortlidge EE, Bangera G, Brownell SE. 2017. Each to their own CURE: faculty who teach course-based undergraduate research experiences report why you too should teach a CURE. J Microbiol Biol Educ 18:18.2.29. doi: 10.1128/jmbe.v18i2.1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Miwa M, Ura M, Nishida M, Sawada N, Ishikawa T, Mori K, Shimma N, Umeda I, Ishitsuka H. 1998. Design of a novel oral fluoropyrimidine carbamate, capecitabine, which generates 5-fluorouracil selectively in tumours by enzymes concentrated in human liver and cancer tissue. Eur J Cancer 34:1274–1281. doi: 10.1016/S0959-8049(98)00058-6. [DOI] [PubMed] [Google Scholar]
- 22.Power DG, Kemeny NE. 2009. The role of floxuridine in metastatic liver disease. Mol Cancer Ther 8:1015–1025. doi: 10.1158/1535-7163.MCT-08-0709. [DOI] [PubMed] [Google Scholar]
- 23.Nakada D, Magasanik B. 1964. The roles of inducer and catabolite repressor in the synthesis of beta-galactosidase by Escherichia coli. J Mol Biol 8:105–127. doi: 10.1016/s0022-2836(64)80153-4. [DOI] [PubMed] [Google Scholar]
- 24.Rosen B, Rothman F, Weigert MG. 1969. Miscoding caused by 5-fluorouracil. J Mol Biol 44:363–375. doi: 10.1016/0022-2836(69)90181-8. [DOI] [PubMed] [Google Scholar]
- 25.Nyhlén A, Ljungberg B, Nilsson-Ehle I, Odenholt I. 2002. Bactericidal effect of combinations of antibiotic and antineoplastic agents against Staphylococcus aureus and Escherichia coli. Chemotherapy 48:71–77. doi: 10.1159/000057665. [DOI] [PubMed] [Google Scholar]
- 26.Hamilton-Miller JM. 1984. Antimicrobial activity of 21 anti-neoplastic agents. Br J Cancer 49:367–369. doi: 10.1038/bjc.1984.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bodet CA, Jorgensen JH, Drutz DJ. 1985. Antibacterial activities of antineoplastic agents. Antimicrob Agents Chemother 28:437–439. doi: 10.1128/aac.28.3.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gieringer JH, Wenz AF, Just HM, Daschner FD. 1986. Effect of 5-fluorouracil, mitoxantrone, methotrexate, and vincristine on the antibacterial activity of ceftriaxone, ceftazidime, cefotiam, piperacillin, and netilmicin. Chemotherapy 32:418–424. doi: 10.1159/000238445. [DOI] [PubMed] [Google Scholar]
- 29.Oe C, Hayashi H, Hirata K, Kawaji K, Hashima F, Sasano M, Furuichi M, Usui E, Katsumi M, Suzuki Y, Nakajima C, Kaku M, Kodama EN. 2019. Pyrimidine analogues as a new class of gram-positive antibiotics, mainly targeting thymineless-death related proteins. ACS Infect Dis 6:1490–1500. doi: 10.1021/acsinfecdis.9b00305. [DOI] [PubMed] [Google Scholar]
- 30.Flórez AB, Sierra M, Ruas-Madiedo P, Mayo B. 2016. Susceptibility of lactic acid bacteria, bifidobacteria and other bacteria of intestinal origin to chemotherapeutic agents. Int J Antimicrob Agents 48:547–550. doi: 10.1016/j.ijantimicag.2016.07.011. [DOI] [PubMed] [Google Scholar]
- 31.Hind CK, Dowson CG, Sutton JM, Jackson T, Clifford M, Garner RC, Czaplewski L. 2019. Evaluation of a library of FDA-approved drugs for their ability to potentiate antibiotics against multidrug-resistant gram-negative pathogens. Antimicrob Agents Chemother 63:e00769-19. doi: 10.1128/AAC.00769-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zgurskaya HI, Rybenkov VV. 2020. Permeability barriers of Gram-negative pathogens. Ann N Y Acad Sci 1459:5–18. doi: 10.1111/nyas.14134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Singh SB, Young K, Silver LL. 2017. What is an “ideal” antibiotic? Discovery challenges and path forward. Biochem Pharmacol 133:63–73. doi: 10.1016/j.bcp.2017.01.003. [DOI] [PubMed] [Google Scholar]
- 34.Schilcher K, Horswill AR. 2020. Staphylococcal biofilm development: structure, regulation, and treatment strategies. Microbiol Mol Biol Rev 84:e00026-19. doi: 10.1128/MMBR.00026-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yeo W-S, Arya R, Kim KK, Jeong H, Cho KH, Bae T. 2018. The FDA-approved anti-cancer drugs, streptozotocin and floxuridine, reduce the virulence of Staphylococcus aureus. Sci Rep 8:2521. doi: 10.1038/s41598-018-20617-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Younis W, Thangamani S, Seleem MN. 2015. Repurposing non-antimicrobial drugs and clinical molecules to treat bacterial infections. Curr Pharm Des 21:4106–4111. doi: 10.2174/1381612821666150506154434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lam SW, Guchelaar HJ, Boven E. 2016. The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50:9–22. doi: 10.1016/j.ctrv.2016.08.001. [DOI] [PubMed] [Google Scholar]
- 38.Bajetta E, Carnaghi C, Somma L, Stampino CG. 1996. A pilot safety study of capecitabine, a new oral fluoropyrimidine, in patients with advanced neoplastic disease. Tumori 82:450–452. doi: 10.1177/030089169608200507. [DOI] [PubMed] [Google Scholar]
- 39.Ayon NJ, Gutheil WG. 2019. Dimensionally enhanced antibacterial library screening. ACS Chem Biol 14:2887–2894. doi: 10.1021/acschembio.9b00745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Oe C, Hayashi H, Hirata K, Kawaji K, Hashima F, Sasano M, Furuichi M, Usui E, Katsumi M, Suzuki Y, Nakajima C, Kaku M, Kodama EN. 2020. Pyrimidine analogues as a new class of gram-positive antibiotics, mainly targeting thymineless-death related proteins. ACS Infect Dis 6:1490–1500. doi: 10.1021/acsinfecdis.9b00305. [DOI] [PubMed] [Google Scholar]
- 41.Bertani G. 1951. Studies on lysogenesis. I. The mode of phage liberation by lysogenic Escherichia coli. J Bacteriol 62:293–300. doi: 10.1128/JB.62.3.293-300.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bertani G. 2004. Lysogeny at mid-twentieth century: P1, P2, and other experimental systems. J Bacteriol 186:595–600. doi: 10.1128/jb.186.3.595-600.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Paz MM, Zhang X, Lu J, Holmgren A. 2012. A new mechanism of action for the anticancer drug mitomycin C: mechanism-based inhibition of thioredoxin reductase. Chem Res Toxicol 25:1502–1511. doi: 10.1021/tx3002065. [DOI] [PubMed] [Google Scholar]
- 44.Umezawa H, Maeda K, Takeuchi T, Okami Y. 1966. New antibiotics, bleomycin A and B. J Antibiot 19:200–209. [PubMed] [Google Scholar]
- 45.Umezawa H, Suhara Y, Takita T, Maeda K. 1966. Purification of bleomycins. J Antibiot 19:210–215. [PubMed] [Google Scholar]
- 46.Suzuki H, Nagai K, Yamaki H, Tanaka N, Umezawa H. 1969. On the mechanism of action of bleomycin: scission of DNA strands in vitro and in vivo. J Antibiot 22:446–448. doi: 10.7164/antibiotics.22.446. [DOI] [PubMed] [Google Scholar]
- 47.Pittillo RF, Hunt DE. 1967. Broad-spectrum antimicrobial activity of a new triazenoimidazole. Appl Microbiol 15:531–532. doi: 10.1128/am.15.3.531-532.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bhimani RS, Vendrov Y, Furmanski P. 1999. Influence of lactoferrin feeding and injection against systemic staphylococcal infections in mice. J Appl Microbiol 86:135–144. doi: 10.1046/j.1365-2672.1999.00644.x. [DOI] [PubMed] [Google Scholar]
- 49.Jiang X, Wang Y, Qin Y, He W, Benlahrech A, Zhang Q, Jiang X, Lu Z, Ji G, Zheng Y. 2017. Micheliolide provides protection of mice against Staphylococcus aureus and MRSA infection by down-regulating inflammatory response. Sci Rep 7:41964. doi: 10.1038/srep41964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cheng AG, Kim HK, Burts ML, Krausz T, Schneewind O, Missiakas DM. 2009. Genetic requirements for Staphylococcus aureus abscess formation and persistence in host tissues. FASEB J 23:3393–3404. doi: 10.1096/fj.09-135467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cheng AG, McAdow M, Kim HK, Bae T, Missiakas DM, Schneewind O. 2010. Contribution of coagulases towards Staphylococcus aureus disease and protective immunity. PLoS Pathog 6:e1001036. doi: 10.1371/journal.ppat.1001036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kim HK, Missiakas D, Schneewind O. 2014. Mouse models for infectious diseases caused by Staphylococcus aureus. J Immunol Methods 410:88–99. doi: 10.1016/j.jim.2014.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shaked Y, Pham E, Hariharan S, Magidey K, Beyar-Katz O, Xu P, Man S, Wu FTH, Miller V, Andrews D, Kerbel RS. 2016. Evidence implicating immunological host effects in the efficacy of metronomic low-dose chemotherapy. Cancer Res 76:5983–5993. doi: 10.1158/0008-5472.CAN-16-0136. [DOI] [PubMed] [Google Scholar]
- 54.Krarup A, Chattopadhyay P, Bhattacharjee AK, Burge JR, Ruble GR. 1999. Evaluation of surrogate markers of impending death in the galactosamine-sensitized murine model of bacterial endotoxemia. Lab Anim Sci 49:545–550. [PubMed] [Google Scholar]
- 55.Mueller JH, Hinton J. 1941. A protein-free medium for primary isolation of the Gonococcus and Meningococcus. Proc Soc Exp Biol Med 48:330–333. doi: 10.3181/00379727-48-13311. [DOI] [Google Scholar]
- 56.Ersoy SC, Heithoff DM, Barnes L, Tripp GK, House JK, Marth JD, Smith JW, Mahan MJ. 2017. Correcting a fundamental flaw in the paradigm for antimicrobial susceptibility testing. EBioMedicine 20:173–181. doi: 10.1016/j.ebiom.2017.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nagy TA, Crooks AL, Quintana JLJ, Detweiler CS. 2020. Clofazimine reduces the survival of Salmonella enterica in macrophages and mice. ACS Infect Dis 6:1238–1249. doi: 10.1021/acsinfecdis.0c00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Dombach JL, Quintana JLJ, Nagy TA, Wan C, Crooks AL, Yu H, Su C-C, Yu EW, Shen J, Detweiler CS. 2020. A small molecule that mitigates bacterial infection disrupts Gram-negative cell membranes and is inhibited by cholesterol and neutral lipids. PLoS Pathog 16:e1009119. doi: 10.1371/journal.ppat.1009119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.McDonough J, Goudsouzian LK, Papaj A, Maceli AR, Klepac-Ceraj V, Peterson CN. 2017. Stressing Escherichia coli to educate students about research: a CURE to investigate multiple levels of gene regulation. Biochem Mol Biol Educ 45:449–458. doi: 10.1002/bmb.21055. [DOI] [PubMed] [Google Scholar]
- 60.Hall DJ, Martin CG, Welford M, Debbert SL. 2018. An inquiry-based exercise in medicinal chemistry: synthesis of a molecular library and screening for potential antimalarial and anti-inflammatory compounds. Biochem Mol Biol Educ 46:424–434. doi: 10.1002/bmb.21138. [DOI] [PubMed] [Google Scholar]
- 61.Shimma N, Umeda I, Arasaki M, Murasaki C, Masubuchi K, Kohchi Y, Miwa M, Ura M, Sawada N, Tahara H, Kuruma I, Horii I, Ishitsuka H. 2000. The design and synthesis of a new tumor-selective fluoropyrimidine carbamate, capecitabine. Bioorg Med Chem 8:1697–1706. doi: 10.1016/s0968-0896(00)00087-0. [DOI] [PubMed] [Google Scholar]
- 62.Reigner B, Blesch K, Weidekamm E. 2001. Clinical pharmacokinetics of capecitabine. Clin Pharmacokinet 40:85–104. doi: 10.2165/00003088-200140020-00002. [DOI] [PubMed] [Google Scholar]
- 63.Shindoh H, Nakano K, Yoshida T, Ishigai M. 2011. Comparison of in vitro metabolic conversion of capecitabine to 5-FU in rats, mice, monkeys and humans–toxicological implications. J Toxicol Sci 36:411–422. doi: 10.2131/jts.36.411. [DOI] [PubMed] [Google Scholar]
- 64.Shindoh H, Kawashima A, Shishido N, Nakano K, Kobayashi K, Horii I. 2006. Relationship between AUC of 5’-DFUR and toxicity of capecitabine, fluoropyrimidine carbamate analogs, and 5’-DFUR in monkeys, mice, and rats. J Toxicol Sci 31:265–285. doi: 10.2131/jts.31.265. [DOI] [PubMed] [Google Scholar]
- 65.Kolinsky K, Shen B-Q, Zhang Y-E, Kohles J, Dugan U, Zioncheck TF, Heimbrook D, Packman K, Higgins B. 2009. In vivo activity of novel capecitabine regimens alone and with bevacizumab and oxaliplatin in colorectal cancer xenograft models. Mol Cancer Ther 8:75–82. doi: 10.1158/1535-7163.MCT-08-0596. [DOI] [PubMed] [Google Scholar]
- 66.Venturini M. 2002. Rational development of capecitabine. Eur J Cancer 38(Suppl 2):3–9. doi: 10.1016/S0959-8049(01)00414-2. [DOI] [PubMed] [Google Scholar]
- 67.Twelves C, Wong A, Nowacki MP, Abt M, Burris H, Carrato A, Cassidy J, Cervantes A, Fagerberg J, Georgoulias V, Husseini F, Jodrell D, Koralewski P, Kröning H, Maroun J, Marschner N, McKendrick J, Pawlicki M, Rosso R, Schüller J, Seitz J-F, Stabuc B, Tujakowski J, Van Hazel G, Zaluski J, Scheithauer W. 2005. Capecitabine as adjuvant treatment for stage III colon cancer. N Engl J Med 352:2696–2704. doi: 10.1056/NEJMoa043116. [DOI] [PubMed] [Google Scholar]
- 68.Petrelli F, Cabiddu M, Barni S. 2012. 5-Fluorouracil or capecitabine in the treatment of advanced colorectal cancer: a pooled-analysis of randomized trials. Med Oncol 29:1020–1029. doi: 10.1007/s12032-011-9958-0. [DOI] [PubMed] [Google Scholar]
- 69.Ma WW, Saif MW, El-Rayes BF, Fakih MG, Cartwright TH, Posey JA, King TR, von Borstel RW, Bamat MK. 2017. Emergency use of uridine triacetate for the prevention and treatment of life-threatening 5-fluorouracil and capecitabine toxicity. Cancer 123:345–356. doi: 10.1002/cncr.30321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Peters GJ, van Dijk J, Laurensse E, van Groeningen CJ, Lankelma J, Leyva A, Nadal JC, Pinedo HM. 1988. In vitro biochemical and in vivo biological studies of the uridine “rescue” of 5-fluorouracil. Br J Cancer 57:259–265. doi: 10.1038/bjc.1988.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hoiseth SK, Stocker D. 1981. Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines. Nature 291:238–239. doi: 10.1038/291238a0. [DOI] [PubMed] [Google Scholar]
- 72.Makarova O, Johnston P, Walther B, Rolff J, Roesler U. 2017. Complete genome sequence of the disinfectant susceptibility testing reference strain Staphylococcus aureus subsp. aureus ATCC 6538. Genome Announc 5:e00293-17. doi: 10.1128/genomeA.00293-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Myers GE, McCready RGL. 1963. Adverse effect of ammonium salts on the antibacterial activity of paraformaldehyde solutions. Appl Microbiol 1:357–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chapman GH. 1945. The significance of sodium chloride in studies of staphylococci. J Bacteriol 50:201–203. doi: 10.1128/JB.50.2.201-203.1945. [DOI] [PubMed] [Google Scholar]
- 75.Dolan EL. 2017. Undergraduate research as curriculum. Biochem Mol Biol Educ 45:293–298. doi: 10.1002/bmb.21070. [DOI] [PubMed] [Google Scholar]
- 76.Reens AL, Crooks AL, Su C-C, Nagy TA, Reens DL, Podoll JD, Edwards ME, Yu EW, Detweiler CS. 2018. A cell-based infection assay identifies efflux pump modulators that reduce bacterial intracellular load. PLoS Pathog 14:e1007115. doi: 10.1371/journal.ppat.1007115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.National Research Council. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental file 1. Download SPECTRUM00275-21_Supp_1_seq5.xlsx, XLSX file, 0.9 MB (972.4KB, xlsx)



