Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are still difficult to treat, despite the availability of many FDA-approved antibiotics. Thus, new compound scaffolds are still needed to treat MRSA. The oxadiazole-containing compound, HSGN-94, has been shown to reduce lipoteichoic acid (LTA) in S. aureus, but the mechanism that accounts for LTA biosynthesis inhibition remains uncharacterized. Here, we report the elucidation of the mechanism by which HSGN-94 inhibits LTA biosynthesis via utilization of global proteomics, activity-based protein profiling, and lipid analysis via multiple reaction monitoring (MRM). Our data suggest that HSGN-94 inhibits LTA biosynthesis via direct binding to PgcA and downregulation of PgsA. We further show that HSGN-94 reduces the MRSA load in skin infection (mouse) and decreases pro-inflammatory cytokines in MRSA infected wounds. Collectively, HSGN-94 merits further consideration as a potential drug for staphylococcal infections.
Keywords: oxadiazole, antibiotic, drug-resistant bacteria, MRSA, lipoteichoic acid
Introduction:
Staphylococcus aureus is one of the major causative agents of community- and hospital-acquired bacteremia, surgical site infections, osteomyelitis, pneumonia, and skin infections1. Methicillin-resistant S. aureus (MRSA) bacteremia causes higher death rates than infections caused by non-MRSA strains2,3. Although there are several antibiotics used to treat MRSA such as clindamycin, minocycline, daptomycin or vancomycin4, drug-resistance has been documented, attributing to the fact that 14% of patients who get serious MRSA infections die4–8. Additionally, newer antibacterial agents developed to treat drug-resistant bacterial pathogens like MRSA are only derivatives of existing drugs. Thus it is likely that resistance mechanisms affecting the older drugs would alter the activity of the newer ones9. The need for novel chemical entities with new mechanisms of action cannot be overstated.
The membrane of many Gram-positive bacteria, such as Streptococcus pneumonia and S. aureus, is decorated with an anionic 1,3-glycerolphosphate-containing polymer called LTA (lipoteichoic acid)11. In S. aureus, LTA is important for bacterial growth10, cell wall physiology12, membrane homeostasis13, virulence14–15, and biofilm formation16. LTA synthesis begins in the cytoplasm where the α-phosphoglucomutase (PgcA) synthesizes glucose-1-phosphate from glucose-6-phosphate. Next, uridyltransferase GtaB produces uridyl diphosphate-glucose (UDP-glucose) via the activaton of uridyl triphosphate (UTP). Then, YpfP transports glucose from UDP-Glucose to diacylglycerol (DAG) to give diglucosyl-diacylglycerol (Glc2-DAG). Glc2-DAG is flipped to membrane by the flippase LtaA where it then acts as a starting unit for LTA. Lastly, the synthase LtaS catalyzes the addition of glycerol phosphate to Glc2-DAG, thereby generating LTA10, 17 (Figure 1A). The mechanism by which LtaS generates LTA involves the use of catalytic threonine (T300)18–20. Phosphatidyl glycerol (PG) transfers phosphoglycerol units to T300, realeasing DAG to produce a covalent intermediate. Phosphoglycerol is then transferred to Glc2-DAG via reaction with the covalent intermediate to give glycerol-phosphate- diglucosyl-diacylglycerol (GroP-Glc2DAG). Repeat units are then added to the glycerol tip, giving rise to LTA (Figure 1B). Since LTA is essential to S. aureus, it has been deemed a potential antimicrobial target21.
Figure 1.

LTA biosynthesis in S. aureus involves both Glc2DAG and PG. (A) PgcA synthesizes glucose-1-phosphate from glucose-6-phosphate. Next, UDP-glc is produced from GtaB’s activation of UTP. YpfP then transfers glucose from UDP-Glc to DAG in order to make Glc2-DAG. Then, LtaA flips Glc2-DAG to the outer membrane where LtaS catalyzes the addition of glycerol phosphate to Glc2-DAG, generating LTA10. (B) LtaS synthesizes LTA via utilization of catalytic T300. PG transfers phosphoglycerol units to T300 formig a covalent intermediate. Then, phosphoglycerol units are transferred to Glc2DAG through a reaction with the covalent intermediate, giving rise to GroP-Glc2DAG. Repeat units are then added to the polymer, producing LTA. (C) HSGN-94 is a highly potent antimicrobial agent discovered to inhibit LTA biosynthesis.
Our lab has demonstrated that N-(1,3,4-oxadiazol-2-yl)benzamides are effective antimicrobial agents22–27. We previously demonstrated that the N-(1,3,4-oxadiazol-2-yl)benzamide HSGN-94 was an antimicrobial agent against drug-resistant Gram-positive bacteria27. The MIC (minimum inhibitory concentrations) of HSGN-94 against bacteria ranged from 0.25 μg/mL to 2 μg/mL27 (Figure 1C). Additionally, HSGN-94 was found to be an inhibitor of LTA biosynthesis in S. aureus27. We also demonstrated HSGN-94’s ability to inhibit MRSA and vancomycin-resistant Enterococci biofilm formation26.
Building upon our previous studies, this study investigated the antibacterial profile of HSGN-94 including structure-activity relationship studies, MICs against various multidrug-resistant Gram-positive bacteria, cytotoxicity assessment, and the resistance development assay. We also elucidated the mechanism by which HSGN-94 inhibits LTA biosynthesis in S. aureus, utilizing several mechanistic studies, including global proteomics, activity-based protein, and transcriptional profiling, lipidomics, and macromolecular synthesis inhibition. Finally, we evaluated HSGN-94’s in vivo efficacy in a MRSA murine wound infection model as well as its ability to inhibit the manifestation of pro-inflammatory cytokines and the effect of HSGN-94’s treatment on the histopathological features of the mice skin.
Results and Discussion:
Structure-activity relationship studies
We established structure-activity-relationship (SAR) studies of HSGN-94, which has indicated important structural features of the molecule that account for its potent antibacterial activity. The synthetic schemes of these new analogs can be found in the supporting information (SI; Schemes S1-S8). The results of the initial screening of the synthesized analogs against S. aureus are included in Table S1. We discovered that the size of the sulfonamide ring A (Figure 2, Series 1) and nature of substitution were all critical for antibacterial activity. For instance, while HSGN-94 (3,5-dimethylpiperidine) had an MIC of 0.25 μg/mL (0.5 μM), the analog 2-azabicyclo [2.2.1]heptane (5) had an MIC value of 2 μg/mL (4.1 μM). Furthermore, monomethyl substituted analogs 6, 7, and 8, also had less antibacterial activity with MICs of 1 μg/mL (2 μM), 2 μg/mL (4 μM), and 2 μg/mL (4 μM) respectively. Analogs containing smaller ring systems (1, 2, 3, and 4) showed less activity than HSGN-94 as well, with MIC values from 2 μg/mL (4.2 μM) to 16 μg/mL (36.3 μM) (Table S1).
Figure 2.

Summary of structure-activity-relationship study of sulfonamide-containing N-(1,3,4-oxadiazol-2-yl)benzamides
Moreover, we determined that an unsubstituted phenyl moiety of the benzamide (ring B) was optimal for the antibacterial activity, as replacing ring B with other heterocycles (Figure 2, Series 2) or substituting ring B with electron-donating or electron-withdrawing groups (Figure 2, Series 3) showed a decrease in the antibacterial activity by 4 to >256 times (Table S1). Likewise, both the amide bond and trifluoromethylphenyl group (ring C) were imperative for antibacterial activity. Conversion of the amide to a tertiary amide (17) or reversion of the amide (18) completely abrogated the anti-S. aureus activity (MICs of >64 μg/mL). Substitution of ring C with alkyl-containing groups (19 and 20) showed a reduction in antibacterial activity (MICs = 64 - >64 μg/mL) (Table S1). Altogether, our SAR investigations demonstrated that HSGN-94 displayed the most potent activity against staphylococci. Therefore, it was selected for further characterization.
Profiling the antibacterial activity of HSGN-94
We explored the antibacterial activity of HSGN-94 versus a panel of multidrug-resistant bacterial strains, such as methicillin-sensitive, methicillin-, and vancomycin-resistant S. aureus, S. epidermidis, Streptococcus pneumonia, S. pyogenes, vancomycin-resistant Enterococcus faecium and E. faecalis, and Listeria monocytogenes. As reported in our study before27, HSGN-94 showed potent activity against staphylococcal strains with MIC values ranging from 0.25 μg/mL (0.5 μM) to 1 μg/mL (2 μM) (Table S2). HSGN-94 was also found to be highly potent to drug-resistant S. pneumoniae and S. pyogenes as well (MICs ranging from 0.06 μg/mL (0.1 μM) to 0.25 μg/mL (0.5 μM), surpassing the activity of linezolid and vancomycin (Table S3). S. pneumoniae is a pathogen that produces a variety of infections such as ear and sinus infections as well as pneumonia, bloodstream infections, and meningitis28–29. Additionally, S. pneumoniae causes around 2 million infection in the United States annually, resulting in more than 6,000 deaths as well as $4 billion in health-care costs. Additionally, greater than 30% of pneumococcal infections are resistant to FDA approved antibiotics30–31. Thus, the CDC classifies S. pneumoniae as a serious threat and new agents against this pathogen are needed31. Additionally, the compound maintained the same potency against vancomycin-resistance enterococci (MIC = 0.25 μg/mL (0.5 μM)), a serious threat pathogen that greatly demands new therapeutics32–34. HSGN-94 was also superior to linezolid and vancomycin against L. monocytogenes with an MIC of 0.06 μg/mL (0.1 μM) (Table S3).
The cytotoxicity profile of HSGN-94 was assessed against human keratinocyte (HaCaT) cells (Figure S1). The compound was non-toxic to HaCaT cells at a concentration up to 64 μg/mL, which represents 64 to 256 times its MIC values against staphylococcal strains.
Moreover, we assessed MRSA USA300’s ability to form resistance to HSGN-94, using the multi-step resistance selection assay. Excitingly, the compound displayed low propensity to develop resistance to MRSA USA300, where its MIC remained unchanged over 65 passages (Figure S2). In contrast, MRSA formed rapid resistance to ciprofloxacin where its MIC progressively increased after following passages reaching 128-fold increase in the MIC after 65 passages (Figure S2).
Effects of HSGN-94 on global proteomics in S. aureus:
After recognizing that obtainment of HSGN-94 resistant mutants via serial passaging was not possible (Figure S2), we went ahead and utilized global proteomics to ascertain the proteins that are impacted after S. aureus treatment with HSGN-94. S. aureus was treated with HSGN-94 for 2 hours and then the total protein content was isolated for investigation via liquid chromatography-tandem mass spectrometry (LC-MS/MS). Samples treated with HSGN-94 were related to samples treated only with DMSO (control). Proteomics data was sorted using label-free quantitation (LFQ) and analysis was pursued for proteins showing LFQ response in all 3 samples tested. During the evaluation of the proteomics data, we discovered a total number of 1475 proteins. Yet, out of these proteins, 1431 proteins (97.0%) were found to be common to both DMSO- and HSGN-94-treated samples, while 30 proteins (3.0%) were only detected in DMSO-treated sample, and 14 proteins (1.0%) were only found in HSGN-94-treated sample (Figure 3A). Stringent filtration using statistical analysis of the 1475 total proteins (where p <0.05) gave 423 proteins, which demonstrated that the treatment and control samples gathered into two differentially expressed groups (Figure 3B). From those 423 significant proteins, 198 were downregulated, while 225 were upregulated. We then filtered this data set using p-value (p) and fold change (Log2FC). Based on this analysis, 18 significant proteins were downregulated by Log2FC ≤ −2, whereas 7 proteins were upregulated by Log2FC ≥ 2 after HSGN-94 treatment (Figure 3B & 3C).
Figure 3.

Global proteomics assessment of S. aureus cells after HSGN-94 treatment. (A) A Venn diagram was constrcuted to compare proteins found in DMSO-treated cells individually, HSGN-94-treated cells individually and in both groups. (B) Heatmap analysis of significant proteins (p <0.05) demonstrating differential expression between both DMSO- and HSGN-94-treated S. aureus. (C) Volcano plot of significant proteins displaying the Log2 fold change (Log2FC, x-axis) vs. p-value (y-axis). Most down and upregulated proteins have been labled. Differentially expressed proteins were identified as p ≤0 .05 and Log2FC ≤ −2 for downregulated proteins or p ≤ 0.05 and Log2FC ≥ 2 for upregulated ones. The Perseus software was used to analyze data35. OriginPro 2017 Software (OriginLab, Massachusetts, USA) was utilized to construct the volcano plot.
Evaluation of the 18 downregulated proteins (those with Log2FC ≤ −2) demonstrated that S. aureus treatment with HSGN-94 impacted several bacterial processes such as transcription, translation, nucleotide metabolism, amino acid biosynthesis, and carbohydrate biosynthesis (Table 1). Importantly, HSGN-94 seemed to have the most significant impact on virulence (Figure S3 and Table 1). For instance, thermonuclease (Nuc) which is an important virulence factor in S. aureus as it is vital for DNA and RNA degradation, a crucial part of the organism’s denfense mechanism36–37, was the most downregulated protein (Log2FC = −5.1) (Figure S3 and Table 1). Likewise, other virulence factors were also downregulated such as gamma-hemolysin subunit B, LukS-PV (Log2FC = −3.7), leucotoxin LukD (Log2FC = −2), Nitrate reductase subunit alpha NarG (Log2FC = −2), Lipoyl synthase LipA (Log2FC = −2), and hydrolase SdrD (Log2FC = −2) (Figure S3 and Table 1). Additionally, HSGN-94 appears to substantially affect the type VII secretion system (T7SS) of S. aureus, which is an essential pathway for bacterial virulence38. The T7SS, activated by host’s lipids, consists of four membrane-associated proteins (EsaA, EssA, EssB, and EssC), three cytosolic proteins (EsaB, EsaE, and EsaG), and five virulence factors that are produced (EsxA, EsxC, EsxB, EsxD, and EsaD)39,40. S. aureus treatment with HSGN-94 showed downregulation of several proteins in the T7SS pathway like EsaA (Log2FC = −2.0), EsxA (Log2FC = −2.0), and EssB (Log2FC = −3.3) (Figure S3 and Table 1). Similarly, both EssC and EsaB were also downregulated by HSGN-94 treatment as they were only identified in the DMSO-treated group (Table 2).
Table 1.
Proteins that were downregulated in S. aureus after HSGN-94 treatment.
| ID | Protein | Classification | Log2FC | p-value |
|---|---|---|---|---|
| Virulence | ||||
| gi|685631952 | Nuc | Thermonuclease | −5.1 | 0.0010 |
| gi|685633550 | LukS-PV | Gamma-hemolysin subunit B | −3.7 | 0.000026 |
| gi|685631441 | EssB | Type VII secretion protein EssB | −3.3 | 0.024 |
| gi|685631437 | EsxA | Type VII secretion protein EsxA | −2.0 | 0.00063 |
| gi|685631438 | EsaA | Type VII secretion protein EsaA | −2.0 | 0.00011 |
| gi|685631717 | SdrD | Hydrolase | −2.0 | 0.00010 |
| gi|685632604 | LukD | Leucotoxin LukD | −2.0 | 0.013 |
| gi|685633523 | NarG | Nitrate reductase subunit alpha | −2.0 | 0.0021 |
| gi|685631988 | LipA | Lipoyl synthase | −2.0 | 0.000029 |
| Nucleotide metabolism | ||||
| gi|685632728 | GloB | Hydroxyacylglutathione hydrolase | −2.0 | 0.000081 |
| gi|685632295 | PyrF | Orotidine 5-phosphate decarboxylase | −2.2 | 0.00010 |
| gi|685632294 | CarB | Carbamoyl-phosphate synthase large chain | −2.0 | 0.000046 |
| Transcription | ||||
| gi|685633415 | SarR | MarR family transcriptional regulator | −2.0 | 0.000014 |
| Translation | ||||
| gi|685632759 | MtaB | 30S ribosomal protein S12 methylthiotransferase | −2.0 | 0.0067 |
| Amino acid biosynthesis | ||||
| gi|685633333 | Als | Acetolactate synthase | −2.7 | 0.0031 |
| gi|685633124 | DapE | Succinyl-diaminopimelate desuccinylase | −3.3 | 0.000044 |
| gi|685632550 | IlvA | Threonine dehydratase | −2.0 | 0.0030 |
| Carbohydrate metabolism | ||||
| gi|685631305 | AdhE | Acetaldehyde dehydrogenase | −2.5 | 0.0039 |
Table 2.
Proteins that were upregulated in S. aureus after treatment with HSGN-94.
| ID | Protein | Classification | Log2FC | p-value |
|---|---|---|---|---|
| Metabolism | ||||
| gi|685633108 | Bsh | choloylglycine hydrolase | 3.6 | 0.0011 |
| gi|685631431 | peptidase M23 | 2.0 | 0.000037 | |
| gi|685631436 | TraG | CHAP domain-containing protein | 2.0 | 0.000010 |
| gi|685632306 | DefA | peptide deformylase | 2.0 | 0.0075 |
| Iron Regulation | ||||
| gi|685631322 | IsdI | Heme oxygenase (staphylobilin-producing) 2 | 3.20 | 0.00014 |
| Glycolysis | ||||
| gi|685633543 | GpmA | 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase | 2.2 | 0.00000055 |
| Proteolysis | ||||
| gi|685632066 | MecA | adaptor protein MecA | 2.0 | 0.000077 |
Furthermore, we also analyzed the 30 proteins that were only detected in the DMSO-treated group (see figure Figure 3A and Table 3). Detection of these proteins in the DMSO group only indicates that they were massively downregulated by HSGN-94. In this group of proteins, we utilized the report by Charles et al40 that provided a thorough listing of S. aureus essential genes to identify essential proteins that were only found in the DMSO-treated group. We discovered that HSGN-94 downregulated CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase (PgsA). PgsA is an essential protein in S. aureus because of its important role in phospholipid synthesis in the bacteria41. Additionally, PgsA shares 29% sequence identity with the human cardiolipid synthase which is responsible for the conversion of cytosine diphosphate diacylglycerol (CDP-DAG) and PG to cardiolipin and cytosine monophosphate (CMP) in mitochondria42.
Table 3.
Proteins found only in DMSO-treatment group and considered downregulated by HSGN-94 treatment
| Protein | Biological process |
|---|---|
| DNA recombination protein RecF | Homologous recombination |
| PTS mannose transporter subunit IIABC | Mannose uptake |
| glycerophosphodiester phosphodiesterase | lipid metabolism |
| copper-translocating P-type ATPase | copper transport |
| oxidoreductase | |
| DNA-binding response regulator | Transcription |
| molybdenum ABC transporter ATP-binding protein | Molybdate transport |
| threonine synthase | threonine biosynthesis |
| nucleoside permease | nucleoside intramembrane transporter |
| sodium ABC transporter permease | |
| CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase | Phosphatidylglycerol (PG) synthesis |
| aureolysin | Virulence factor |
| carboxylesterase | Metabolism |
| type VII secretion protein EsaB | Virulence factor |
| hydrolase | Metabolism |
| signal peptidase II | lipoprotein biosynthesis |
| peptidase | Metabolism |
| sodium ABC transporter ATP-binding protein | sodium transport |
| MAP domain-containing protein | Virulence factor |
| protein EssC | Virulence factor |
| nitrite reductase (NAD(P)H) small subunit | Nitrate assimilation |
| glutamyl endopeptidase | Virulence factor |
| 50S rRNA methyltransferase | Translation |
| multifunctional 2,3-cyclic-nucleotide 2-phosphodiesterase/5-nucleotidase/3-nucleotidase | Nucleotide catabolism |
| site-specific integrase | |
| holo-ACP synthase | Pantothenate and CoA biosynthesis |
| Membrane protein | |
| uroporphyrinogen-III C-methyltransferase | Porphyrin and chlorophyll metabolism |
| peptide ABC transporter permease | |
| orthopoxovirus protein, PF05708 family | hydolase |
In S. aureus, PgsA synthesizes PG-P from CDP-DAG by exhanging glycerol-3-phosphate (Gro-3-P) for CMP. Then, phosphatidylglycerol phosphate phosphatase (PgpP) rapdily dephosphorylates PG-P to give PG. Next, cardiolipin synthase (Cls) synthesizes cardiolipin (CL) via the condensation of two PG molecules43. Additionally, as mentioned above, LtaS also uses PG to synthesize LTA. PgsA is essential for the synthesis of PG and PG is important for LTA biosynthesis (Figure 4A). Next we sought to confirm that HSGN-94 downregulates PgsA via utilization of RT-qPCR. These results showed that after HSGN-94 treatment, the tested S. aureus cells exhibited a reduction in pgsA gene expression (Figure 4B), which confirmed our global proteomics analysis.
Figure 4.

HSGN-94 downregulates PgsA. (A) PgsA synthesizes phosphatidylglycerol phosphate (PG-P) from cytosine diphosphate diacylglycerol (CDP-DAG) by exhanging glycerol-3-phosphate (Gro-3-P) for cytosine monophosphate (CMP). Phosphatidylglycerol phosphate phosphatase (PgpP) rapdily dephosphorylates PG-P to give PG. Cardiolipin synthase (Cls) synthesizes cardiolipin (CL) via the condensation of two PG molecules. LtaS also uses PG in order to synthesize LTA. (B) The effect of S. aureus treated with HSGN-94 (0.25 μg/mL) pgsA expression. Experiments were performed in triplicate and normalized with 16S RNA. Error bars represent standard-deviation. Statistical significance amongst both DMSO- and HSGN-94-treated groups were established by Student’s t-test assessment (unpaired, two-tailed) and is characterized as *p ≤ 0.05.
While evaluating proteins that upregulated by HSGN-94 treatment, we discovered that the compound played a significant role in iron acquisition in S. aureus. For example, S. aureus treated with HSGN-94 resulted in upregulation of IsdI, IsdE, HarA, SbnA, SbnC, SbnF, and SbnE, proteins associated with iron obtainment (Table 2 and Table 4). HarA, IsdI, and IsdE are all part of the Isd system in S. aureus found within the cell wall, which facilitates iron acquisition44. Specifically, HarA binds heme within the cell wall and travels along the near iron transporter (NEAT) domains of IsdC and IsdA to IsdE resulting in the introduction of heme in the cytoplasm. Then, heme is degraded by IsdI which releases available iron to fulfill nutrient needs44. HrtA is a protein that regulates heme transportation into the cell and is a component of the heme-regulated transport (Hrt) system45. SbnA, SbnC, SbnF, and SbnE are all part of the sbn operon which are important for the synthesis of the siderophore staphyloferrin B44.
Table 4.
Proteins found only in HSGN-94-treatment group and considered upregulated
| Protein | Biological process |
|---|---|
| zinc ABC transporter ATP-binding protein | Zinc transport |
| siderophore biosynthesis protein SbnE | iron acquistion |
| DUF4930 domain-containing protein | metabolism |
| glycine cleavage system protein H | glycine metabolism |
| thiol reductase thioredoxin | cell redox/ glycerol ether metabolic process |
| Cro/Cl family transcriptional regulator | Transcription |
| 23S rRNA (pseudouridine(1915)-N(3))-methyltransferase RlmH | Translation |
| L-serine dehydratase, iron-sulfur-dependent subunit alpha | Amino acid metabolism/gluconeogenesis |
| CHAP domain-containing protein | Pentaglycine cleavage |
| acylphosphatase | Pyruvate metabolism |
| heme uptake system protein IsdE | iron acquisition |
| siderophore biosynthesis protein SbnC | iron acquisition |
| dihydroxyacetone kinase subunit L | glycerol metabolism (Glycerone phoshorlyation) |
| haptoglobin-binding heme uptake protein HarA | iron acquisition |
| ribonuclease HIII | DNA replication, cleave RNA-DNA hybrids |
| siderophore biosynthesis protein SbnA | iron acquisition |
| siderophore biosynthesis protein SbnF | iron acquisition |
| 30S ribosomal protein S20 | translation |
HSGN-94 Inhibits Glc2-DAG in S. aureus:
The global proteomics analysis demonstrated that HSGN-94 had an effect on numerous biological processes in S. aureus (Figure S3 and Table 1) and the levels of several proteins (~80) were impacted by HSGN-94. However global proteomics does not reveal the actual binding protein(s) to the compound so we further performed activity-based protein profiling to identify the putative targets of HSGN-9446–49. An affinity probe (HSGN-probe) was synthesized in 3 steps via click chemistry between the biotin azide and an alkynyl oxadiazole benzamide (I11) (Figure 5B). Then, a pull-down assay was performed. To ensure that proteins being captured with the HSGN-Probe also bind the unlabeled compound HSGN-94, we used a competition strategy, vide infra, to identify proteins that putatively bind to HSGN-94. Briefly, S. aureus ATCC 25923 cells (at the exponential phase) were incubated with HSGN-Probe plus 50 μM HSGN-94 (sample A) or with HSGN-Probe only (sample B) for 4 hours at 25°C with gentle agitation (Figure 5B). Then, cells were lysed and the biotinylated probes were captured with steptavidin beads, and enriched proteins were evalauted via SDS-PAGE. The bands that show on sample A lane but not on sample B lane (i.e. unique to the HSGN-probe without HSGN-94 competition) would be the proteins that bind to HSGN-94.
Figure 5.

Probe Synthesis and Pull-Down Assay. (A) Synthesis of HSGN-Probea. aReagents and Conditions: (a) MeOH, rt, 12 h 53% (b) BOP Reagent, DIPEA, DMF, rt, 12 h, 24% (c) CuSO4·5H2O, Na Ascorbate, DMF:H2O (10:1), 60°C, 12 h, 77%. (B) Schematic of pull-down assay experiment to identify target(s) of HSGN-94.
Phosphoglucomutase (PgcA) was one of the proteins unique to HSGN-Probe group (Table S4). PgcA is found in both bacteria as well as mammalian cells. However, it has been shown that the structure of bacterial PgcA shares just 25% amino acid identity with mammalian PgcA50–51. In S. aureus, PgcA synthesizes glucose-1-phosphate from glucose-6-phosphate (the first step in the LTA biosynthesis pathway, Figure 1A) which later gives rise to Glc2-DAG. Thus, since HSGN-94 inhibits PgcA, it would also inhibit Glc2-DAG formation. To test this hypothesis, we treated S. aureus ATCC 25923 with HSGN-94 (0.25×, 1×, and 8×MIC) for 5 hours and then extracted total membrane lipids using a similar procedure outlined by Scheewind et al52. The lipids were then separated by thin layer chromatography (TLC), and imaged by α-naphthol/sulfuric acid staining. The TLC indicated that HSGN-94 treatment reduced the amount of the α-naphthol-reactive species, which we tentatively assigned as Glc2-DAG and further characterized (Figure 6A). To identify the stained species, lipids were isolated from TLC plates and examined by MALDI-TOF mass spectrometry. The mass-to-charge (m/z) ratio of the major ion signal of the stained species agreed with the expected mass of Glc2-DAG [M + H]+ containing C15 to C18 fatty acids chain lengths (Figure 6B). For instance, the expected m/z for the [M + H]+ of Glc2-DAG bearing C18 and C15 fatty acid chains was 907.63 and the observed m/z for DMSO and HSGN-94 treated samples were the same as that of the projected m/z (907.62–907.63) (Figure 6B and SI for mass spectra). In summary, our results reveal that, compared to DMSO-treated S. aureus, HSGN-94 (0.25×, 1×, and 8×MIC) inhibits Glc2-DAG which we hypothesize is a result of HSGN-94’s direct binding to PgcA.
Figure 6.

HSGN-94 inhibits Glc2-DAG. (A) Evaluation of S. aureus glycolipids via TLC. Membrane lipids were isolated from S. aureus ATCC 25923, treated with either DMSO (control) or HSGN-94 (0.25×, 1×, and 8×MIC). (B) Glycolipids were separated from TLC and assessed via MALDI mass spectoscopy.
Multiple reaction monitoring profiling (MRM-Profiling) of lipids demonstrates HSGN-94 selectively affects phosphaditlyglycerol (PG) in S. aureus
It has been demonstrated that even in the absence of Glc2-DAG, LTA can still be formed as LtaS uses PG as an alternative starter unit12. Polymers formed on PG to make the alternative PG-LTA are much longer than polymers formed on Glc2-DAG12, 52. Additionally, it was reported that S. aureus cells that make these longer polymers have cell division defects12, 53, are less virulent15, 52, and are more sensitive to β-lactam antibiotics as well as other cell envelope stresses12. However, since the inhibition of Glc2-DAG would not completely deplete LTA, and our results indicte that HSGN-94 potently depletes LTA from S. aureus,27 we hypothesized there must be another mechanism by which HSGN-94 inhibits LTA biosynthesis. Since global proteomics and RT-qPCR show that HSGN-94 downregulates pgsA, an essential protein in S. aureus for PG synthesis, we wondered if HSGN-94 also had an effect on PG. To determine this, we proceeded to perform multiple reaction monitoring profiling (MRM-Profiling)54 to differentiate lipid profile distinctions among S. aureus ATCC 25923 treated with either DMSO (control), or HSGN-94 (1×, and 8× MIC) for 5 hours. Lipids were extracted and analyzed using using electrospray ionization mass spectroscopy (ESI-MS). MRMs were refined by ion counts and false discovery rate (FDR) modified p-value in ANOVA, and then investigated using principal component analysis (PCA). Our experiment focused on analyzing differences in glycerophospholipids and total membrane lipids in S. aureus treated cells. After obtaining this data, PCA scores plots were generated to evaluate HSGN-94’s effects on PG (Figure 7A), total membrane lipids (Figure 7B), phosphotitdylcholine (PC) (Figure 7C), and phosphatidylethanolamine (PE) (Figure 7D). Each point in the PCA plot signifies a single sample of lipid extract from the staphyloccocci and the shaded oval region is characterized as the calculated 95% confidence region for every group. Interestingly, we discovered that HSGN-94 seemed to affect PG (Figure 7A). However, HSGN-94 did not affect total lipids, PC or PE (Figures 7B–D).
Figure 7.

PCA scores plots of S. aureus treated with either DMSO (control) or HSGN-94 ((1×, and 8× MIC). (A) Effects on PG synthesis. (B) Effects on total membrane lipids. (C) Effects on PC synthesis. (D) Effects on PE synthesis. *Note: Blue = DMSO, Red = 1× MIC HSGN-94, and Green = 8× MIC HSGN-94.
Proposed mechanism for HSGN-94’s inhibition of LTA biosynthesis
Since our data demonstrates that HSGN-94 directly binds to PgcA as well as downregulates pgsA, we hypothesized that HSGN-94 inhibits LTA biosynthesis in two distinct manners (Figure 8). First, HSGN-94 directly binds to PgcA thereby inhibiting the synthesis of Glc2-DAG. Secondly, HSGN-94 downregulates PgsA expression, which causes an effect on PG synthesis. We currently do not know if the inhibition of Glc2-DAG synthesis has an indirect effect on PgsA expression or whether the reduction of PgsA expression by HSGN-94 is via a separate mechanism. This will be the focus of a future study.
Figure 8.

Proposed mechanism for HSGN-94 inhibition of LTA biosynthesis. We hypothesize that HSGN-94 inhibits LTA biosynthesis in a dual-mechanistic way.
In addition to LTA inhibition, the global proteomics data (Tables 1 and 3) also showed that treatment of S. aureus with HSGN-94 downregulated proteins involved in translation (mtaB), transcription (SarR and GntR), and nucleotide metabolism (GloB, PyrF, and CarB). Furthermore, we performed GO function analysis using Metascape™ software of the 423 significant proteins (p < 0.05) and identified that nucleotide metabolic and small molecules catabolic processes, as well as precursor metabolites and energy were the most significantly regulated biological processes identified (−log10(p) >10, see Figure S4). Likewise, the proteomics data using affinity HSGN-probe (Table S4) indicated that HSGN-94 interacted with proteins involved in protein synthesis (miaB, rplD, PheT, rplJ, and trmFO), DNA/RNA synthesis (SigA, rpiA, Xpt, RecA, adk, and guaA), as well as cell-wall synthesis (IsaA). Therefore, to further evaluate this, we performed a macromolecular biosynthesis inhibition assay to analyze the integration of radiolabeled precursors into the biosynthesis of macromolecules (DNA, RNA, cell wall, and proteins). As expected from the data outlined in Tables 1, 3, and S4, HSGN-94 exhibited similar or superior inhibition of the biosynthesis of each macromolecule tested as compared to FDA approved antibiotics (Figure S5A-D). It thus appears that the potent antimicrobial activity of HSGN-94 is derived from LTA biosynthesis inhibition and the inhibition of other essential processes in bacteria. This multi-pronged inhibition of essential processes in bacteria explains why attempts to generate resistant clones towards HSGN-94 failed. It has emerged that successful antibiotics used in the clinic, while developed against single targets, are successful because they indeed target other pathways55. For example, daptomycin, which has long been categorized as mainly acting via depolarization of bacterial membrane has now been found to also target cell wall biosynthesis via a mechanism that involves complex formation with undecaprenyl-coupled intermediates and membrane lipids56. Recently, it was also reported that tetracyclines, long thought of as mainly acting via ribosome inhibition, also act via bacterial membrane targeting57.
In vivo efficacy of HSGN-94 in a MRSA murine skin infection:
Based on its potent antibacterial activity, interesting mechanism of action, and the fact that HSGN-94 was not toxic to HaCat cells at concentrations up to 64 μg/mL, the compound was evaluated for its in vivo efficacy in a MRSA murine skin infection model. MRSA-infected wounds were treated as either clindamycin I.P. (25 mg/kg once daily), 2% HSGN-94, 2% mupirocin, or the vehicle alone (petroleum jelly) for five days. twice daily with either 2% HSGN-94, 2% mupirocin, or the vehicle alone (petroleum jelly). Excitingly, HSGN-94 (93.8% reduction) performed similarly to FDA-approved antibiotics clindamycin (93.4% reduction) and mupirocin (98.8% reduction) in lessening the quantity of MRSA in the infected wounds of mice after 5 days (Figure 9).
Figure 9: Reduction of MRSA USA300 in infected wounds of mice after 5 days.

The data are presented as average percent reduction of MRSA CFU/mL in murine skin wounds. A one-way ANOVA with post-hoc Dunnet’s multiple assessments discovered no statistical variation amongst mice treated with mupirocin or clindamycin and mice treated with HSGN-94 (2%).
HSGN-94 reduces pro-inflammatory cytokines:
The severity of S. aureus skin infections is propelled by the overabundance of host pro-inflammatory cytokines58. Several reports have shown that in S. aureus infected wounds, LTA contributes to the increased development of inflammation and skin barrier defects59–62. Since HSGN-94 is a potent LTA inhibitor and effectively reduced MRSA USA300 in infected wounds, we proceeded to examine its effect on pro-inflammatory cytokine expression in the treated wounds. . As depicted from Figure 10A–C, HSGN-94 reduced the levels of the pro-inflammatory cytokines, monocyte chemo attractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), and interleukin-1 beta (IL-1β) in MRSA USA300 skin lesions. Additionally, the reduction of the levels of IL-1β and MCP-1 in response to HSGN-94 was was higher than that generated by mupirocin and clindamycin treatment (Figure 10A and 10B). It was reported that persistent inflammation because of inflammatory cytokines like MCP-1 and TNF-α, significantly impedes recovery in chronic wounds63. It is exciting that HSGN-94 treatment resulted in the reduction of pro-inflammator cytokines, which could result in acceleration of wound healing.
Figure 10.

HSGN-94 Reduces Pro-Inflammatory Cytokines in MRSA Infected Wounds. For A to C, samples were pooled and average values are shown. (A) Level (pg/mL) of IL-1β in MRSA infected wound treated with vehicle, mupirocin, clidamycin, or HSGN-94. (B) Level (pg/mL) of MCP-1 in MRSA infected wound treated with vehicle, mupirocin, clidamycin, or HSGN-94. (C) Level (pg/mL) of TNF-α in MRSA infected wound treated with vehicle, mupirocin, clidamycin, or HSGN-94. (D) MRSA infected wound treated with vehicle with diffuse necrosis, chronic inflammation, edema and bacterial colonization present. (E) MRSA infected wound treated with mupirocin with moderate necrosis, chronic inflammation, and hemorrahge. (F) MRSA infected wound treated with clidamycin demonstrated epithelial hyperplasia on wound periphery, with moderate chronic inflammation and necrosis. (G) Histopathology of MRSA infected wound treated with HSGN-94 demonstrated resolution of inflammation within the deep dermis and subcutis.
To further evaluate HSGN-94’s effect on pro-inflammatory cytokines, we performed histopathology (Figure 10D–G). It is evident that MRSA infected wounds treated with mupirocin (Figure 10E), clidamycin (Figure 10F), or HSGN-94 (Figure 10G) demonstrate evidence of necrosis, hemorrhage, and a chronic inflammatory response. However, the response is less pronounced than the than vehicle treated group (Figure 10D). However, HSGN-94 showed resolution of inflammation and necrosis which was similar to clidamycin treatment but was much superior than mupriocin treatment (see Figure 10E–G). MRSA infected wounds treated with HSGN-94 show reduced levels of pro-inflammatory cytokines, which is superior to that of wounds treated with clindamycin or mupirocin and may be due to HSGN-94’s ability to inhibit LTA biosynthesis.
Conclusion:
In conclusion, using a panoply of techniques, including global proteomics, activity-based protein profiling, lipid analysis, and MRM profiling experiments, we have been able to propose how HSGN-94 inhibits LTA biosynthesis. We propose that HSGN-94 inhibits LTA in two ways: direct binding to PgcA which causes inhibition of Glc2-DAG; and downregulation of PgsA, which leads to a reduction in PG synthesis. Excitingly, HSGN-94 showed high efficacy in diminishing the burden of MRSA in a murine skin infection model and also reduced the pro-inflammatory cytokines in MRSA infected wounds. Antimicrobial resistance is a growing threat and many groups have disclosed new chemical scaffolds that inhibit bacterial growth64–74. Detailed mechanistic work to uncover how each of these unique compounds killing bacteria would likely reveal many novel tactics to tackle this global health challenge.
Experiemental Section:
Chemistry:
All reagents and solvents were purchased from commercial sources and utilized without purification. The 1H, 13C, and 19F NMR spectra were obtained in DMSO-d6, chloroform-d, or methanol-d4 as solvent using a 500 MHz or 800 MHz spectrometer with tetramethylsilane as the internal standard. 1H NMR spectra data are reported as chemical shift (δ ppm) (multiplicity, coupling constant (Hz), integration). High resolution mass spectra (HRMS) were taken using electron spray ionization (ESI) and a TOF mass analyzer. Characterization of compounds was done using 1H NMR, 13C NMR, 19F NMR, and HRMS data. The purity of compounds was determined to be ≥95% by measuring the absorbance at 280 nm with high performance liquid chromatography (HPLC). HPLC spectra were recorded on an Agilent 1260 Infinity system using a ZORBAX SB-C18 column.
Synthetic Procedures:
General Procedure I: Synthesis of Sulfonamide-Containing Intermediates S1-S10, E1-E2, I5, I6, I8, and I10
To a solution of chlorosulfonyl-containing intermediate (1 eq) in methanol (25 mL) was added amine (3 eq). This mixture was stirred overnight at room temperature. Next, the mixture was concentrated under reduced pressure, diluted with ethyl acetate (20 mL), washed twice with water (10 mL), once with brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure. The crude mixture was then purified via column chromatography using hexanes: ethyl acetate (80:20) as solvent system to give pure product.
General Procedure II: Synthesis of Aromatic 1,3,4-oxadiazol-2-amines A1:
The synthesis of A1 was performed using a literature-reported procedure75. Obtained 1H, 13C, and 19F spectra were in agreement with literature-reported data.
General Procedure III: Sandmeyer Reaction for the Synthesis of I1, I3, and I4
Thionyl chloride (5.5 eq) was added dropwise to water (15 mL) at 0°C and then stirred for 18 hours at room temperature. CuCl2 (0.05 eq) was added at 0°C and the mixture was stirred for 15 minutes. In a separate flask, a solution of NaNO2 (1.5 eq) in water (5 mL) was added to a stirred solution of 4-amino-3-nitrobenzonitrile (1 eq), 4-amino3-methoxybenzonitrile (1 eq), or 5-aminonicotinic acid (1 eq) in concentrated HCl (5 mL) at 0°C, over 15 minutes. The diazonium salt solution was added dropwise to the thionyl chloride/CuCl2 solution at 0°C and stirred for 1 hour. Over this time, a precipitate formed which was collected via vacuum filtration and washed with water (10 mL) which gave the sulfonylchloride intermediates I1, I3, or I4. These intermediates were used in the next step without purification or characterization.
General Procedure IV: Hydrolysis of Benzonitriles I5-I7 to Benzoic Acids S13, S14, and S15
To a mixture of benzonitriles I5-I7 in ethanol (10 mL) was added sodium hydroxide (2M in H2O) (16.6 mL) and the solution was refluxed at 100°C overnight. After, the reaction was cooled to room temperature and concentrated under reduced pressure. The crude material was dissolved in H2O and acidified to pH = 1 with 6 M HCl to give an off-white precipitate which was collected via vacuum filtration to give desired product which was continued without further purification or characterization.
General Procedure V: Hydrolysis of Esters E1 and E2 to Carboxylic Acids S11 and S12
To a solution of methyl ester E1 or E2 (1 eq) in MeOH: H2O (2:1) was added LiOH (22 eq) and the solution was stirred at room temperature for 12 hours. After, the mixture was concentrated under reduced pressure and the crude product was dissolved in H2O and acidified to pH = 1 with 6 M HCl to afford the carboxylic acid intermediate as an off-white solid which was collected via vacuum filtration.
General Procedure VI: Reduction of Nitro for the Synthesis of 15 and I9:
To a 50 mL round-bottom flask charged with nitro containing compound 13 (1 eq) or intermediate I8 (1 eq) was dissolved in DMF: H2O (9:1). Na2S2O4 (3.5 eq) was added, and the reaction was run at 90°C for 12 hours. After, the reaction mixture was concentrated under reduced pressure and diluted with ethyl acetate (20 mL). The organic layer was washed twice with H2O (10 mL) and once with brine (10 mL). The organic layer was separated, dried over Na2SO4, and concentrated under reduced pressure to give a crude mixture which was purified via column chromatography (hexanes/ethyl acetate 70:30).
General Procedure VII: Synthesis of Alkyl containing 1,3,4-oxadiazol-2-amines A2 and A3:
The synthesis of A2 and A3 was performed using a literature-reported procedure76. Obtained 1H, and 13C spectra were in agreement with literature-reported data.
General Procedure VIII: Amide Coupling for the Synthesis of Compounds 1–14, 16, 18, and I11:
To a round-bottomed flask was added benzoic acid (1 eq), amine (1 eq), BOP reagent (2.7 eq), and DIPEA (1.5 mL) in DMF solvent (5 mL) was stirred at room temperature for 24 h. After completion, the reaction mixture was concentrated under reduced pressure. The crude reaction mixture was purified by flash column chromatography (hexanes/ethyl acetate 90:10 to 70:30) to give the desired product.
General Procedure IX: Synthesis of Compounds 19 and 20:
To a round-bottom flask with acyl chloride (1.4 eq) and 1,3,4-oxadiazol-2-amine (1 eq) in 1,4, -dioxane (5 mL) was added N-methylimidazole (5.5 eq). The mixture was stirred at 90°C for 2 hours and then concentrated under reduced pressure. The crude mixture was diluted in ethyl acetate (20 mL), washed twice with water (10 mL), once with brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure. Column chromatography (hexanes/ethyl acetate 70:30) gave desired product.
Synthesis of Intermediate I2:
To a round-bottomed flask containing thiophene-2-carboxylic acid was slowly added chlorosulfonic acid (10 mL) at 0°C. After addition, the reaction was stirred at room temperature overnight. Then, the mixture was slowly poured over ice and extracted with ethyl acetate (3 × 30 mL). The combined organic layer was dried over Na2SO4 and concentrated under reduced pressure to give chlorosulfonyl containing intermediate I2 which was proceeded to the next step without further purification or characterization.
Synthesis of Intermediate I7:
To a round-bottom flask containing I6 (1 eq) in anhydrous DCM (10 mL) under argon gas was slowly added BBr3 (5 eq) at −78°C. The mixture was stirred at −78°C for 1 hour and then warmed to room temperature to continue stirring for 12 hours. After, the reaction was cooled to 0°C and quenched slowly with iced water (10 mL). Next, the two layers were separated, and the water layer extracted twice with DCM (10 mL). The organic layers were combined and washed once with NaHCO3 (10 mL), once with brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure to give a crude mixture which was purified via column chromatography (hexanes: ethyl acetate (95:5) to give intermediate I7 as an off-white solid.
Synthesis of Analog 17:
To a solution of HSGN-94 (1 eq) in dry DMF (5 mL), sodium hydride, NaH (3 eq), was added, and the reaction mixture was allowed to stir for 30 min at room temperature. Then, methyl iodide (2 eq) was added dropwise, and the mixture was stirred for 2 h at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was treated with aqueous NH4OH (30% in water, 10 mL). The crude product was extracted with Et2O (3 × 15 mL) and purified by silica gel column chromatography using hexanes: ethyl acetate (70:30) to give desired product.
Synthesis of HSGN-Probe:
To a solution of I11 (14 mg, 31.1 μmol) and biotin-PEG3-N3 (20 mg, 45 μmol, 1.5 eq) in DMF (2 mL) was added a mixture of CuSO4·5H2O (3 mg) and sodium ascorbate (3 mg) in water (0.2 mL). The solution was allowed to stir at 60°C for 12 hours. The solvents were evaporated, and the product was purified by column chromatography using DCM/MeOH (90/10) as eluent to obtain HSGN-Probe (77% yield) as a clear oil.
Characterization Data:
4-(Piperidin-1-ylsulfonyl)benzoic acid (S1):
Off-white solid (190 mg, 71% yield). 1H NMR (500 MHz, DMSO-d6) δ 8.14 (d, J = 8.3 Hz, 2H), 7.83 (d, J = 8.2 Hz, 2H), 2.90 (d, J = 5.4 Hz, 4H), 1.52 (t, J = 5.7 Hz, 4H), 1.35 (d, J = 3.7 Hz, 2H). 13C NMR (126 MHz, DMSO-d6) δ 166.7, 139.8, 135.1, 130.6, 128.1, 47.0, 25.1, 23.2. HRMS (ESI) m/z calcd for C12H16NO4S [M + H]+ 270.0794, found 270.0793.
4-(Pyrrolidin-1-ylsulfonyl)benzoic acid (S2):
Off-white solid (154 mg, 44% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.15 – 8.14 (m, 2H), 7.91 – 7.90 (m, 2H), 3.19 – 3.18 (m, 4H), 1.68 (q, J = 3.7 Hz, 4H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 140.8, 135.1, 130.5, 127.8, 48.2, 25.1. HRMS (ESI) m/z calcd for C11H14NO4S [M + H]+ 256.0644, found 256.0646.
4-(N-Cyclopropylsulfamoyl)benzoic acid (S3):
Off-white solid (166 mg, 51% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.14 (d, J = 8.0 Hz, 2H), 7.93 – 7.92 (m, 3H), 2.19 – 2.18 (m, 1H), 0.50 (dd, J = 7.0, 2.1 Hz, 2H), 0.40 (t, J = 3.1 Hz, 2H). 13C NMR (201 MHz, DMSO-d6) δ 166.6, 144.6, 134.7, 130.3, 127.4, 24.5, 5.5. HRMS (ESI) m/z calcd for C10H12NO4S [M + H]+ 242.0487, found 242.0484.
4-(N,N-Dimethylsulfamoyl)benzoic acid (S4):
Off-white solid (177 mg, 57% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.16 (dd, J = 8.4, 1.8 Hz, 2H), 7.86 (dd, J = 8.4, 1.7 Hz, 2H), 2.66 (s, 6H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 139.4, 135.2, 130.5, 128.0, 37.8. HRMS (ESI) m/z calcd for C9H12NO4S [M + H]+ 230.0487, found 230.0488.
4-((2-Azabicyclo[2.2.1]heptan-2-yl)sulfonyl)benzoic acid (S5):
Off-white solid (210 mg, 55% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.14 – 8.13 (m, 2H), 7.92 – 7.91 (m, 2H), 4.14 (s, 1H), 3.06 – 3.04 (m, 1H), 2.99 (d, J = 9.0 Hz, 1H), 2.45 (s, 1H), 1.56 – 1.54 (m, 3H), 1.32 – 1.30 (m, 1H), 1.20 (d, J = 9.9 Hz, 1H), 0.84 (d, J = 10.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 142.5, 134.9, 130.5, 127.7, 60.3, 54.7, 37.4, 36.6, 31.2, 27.1. HRMS (ESI) m/z calcd for C13H16NO4S [M + H]+ 282.0800, found 282.0801.
4-((4-Methylpiperidin-1-yl)sulfonyl)benzoic acid (S6):
Off-white solid (235 mg, 61% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.15 – 8.14 (m, 2H), 7.85 – 7.84 (m, 2H), 3.64 (d, J = 12.1 Hz, 2H), 2.35 (td, J = 12.1, 2.7 Hz, 2H), 1.66 – 1.64 (m, 2H), 1.36 – 1.32 (m, 1H), 1.15 (qd, J = 12.2, 4.0 Hz, 2H), 0.86 (d, J = 6.5 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 140.4, 135.2, 130.5, 127.9, 46.3, 33.3, 29.6, 21.5. HRMS (ESI) m/z calcd for C13H18NO4S [M + H]+ 284.0957, found 284.0956.
4-((3-Methylpiperidin-1-yl)sulfonyl)benzoic acid (S7):
Off-white solid (227 mg, 59% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.15 – 8.14 (m, 2H), 7.85 – 7.84 (m, 2H), 3.52 (ddd, J = 16.2, 11.9, 6.1 Hz, 2H), 2.36 (td, J = 11.5, 3.2 Hz, 1H), 2.05 – 2.03 (m, 1H), 1.68 – 1.60 (m, 3H), 1.47 (dt, J = 12.9, 3.9 Hz, 1H), 0.90 – 0.87 (m, 1H), 0.84 (d, J = 6.4 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 140.4, 135.1, 130.5, 127.9, 52.9, 46.4, 31.6, 30.5, 24.5, 18.9. HRMS (ESI) m/z calcd for C13H18NO4S [M + H]+ 284.0957, found 284.0959.
4-((2-Methylpiperidin-1-yl)sulfonyl)benzoic acid (S8):
Off-white solid (247 mg, 64% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.12 – 8.11 (m, 2H), 7.91 – 7.90 (m, 2H), 4.13 (d, J = 4.8 Hz, 1H), 3.64 (dt, J = 13.2, 3.2 Hz, 1H), 3.02 (td, J = 13.1, 2.7 Hz, 1H), 1.55 – 1.49 (m, 2H), 1.43 – 1.41 (m, 3H), 1.22 (qt, J = 12.5, 4.2 Hz, 1H), 1.03 (d, J = 7.0 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 166.5, 145.1, 134.7, 130.6, 127.2, 48.8, 40.5, 30.2, 25.1, 18.0, 15.8. HRMS (ESI) m/z calcd for C13H18NO4S [M + H]+ 284.0957, found 284.0957.
5-(3,5-Dimethylpiperidin-1-yl)sulfonyl)nicotinic acid (S9):
Off-white solid (173 mg, 53% yield). 1H NMR (800 MHz, DMSO-d6) δ 9.29 (s, 1H), 9.11 (s, 1H), 8.42 (d, J = 1.8 Hz, 1H), 3.69 – 3.67 (m, 2H), 1.93 (t, J = 11.3 Hz, 2H), 1.66 (tdd, J = 17.8, 15.2, 7.5, 3.8 Hz, 3H), 0.84 (d, J = 6.5 Hz, 6H), 0.57 (q, J = 12.1 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 165.3, 153.9, 151.1, 135.7, 133.8, 127.9, 52.3, 40.9, 31.0, 18.9. HRMS (ESI) m/z calcd for C13H19N2O4S [M + H]+ 299.1066, found 299.1067.
4-(3,5-Dimethylpiperidin-1-yl)sulfonyl)thiophene-2-carboxylic acid (S10):
Off-white solid (220 mg, 66% yield). 1H NMR (500 MHz, DMSO-d6) δ = 8.45 (d, J = 1.6, 1H), 7.72 (d, J = 1.6, 1H), 3.60 – 3.57 (m, 2H), 1.78 (t, J = 11.2, 2H), 1.64 – 1.61 (m, 3H), 0.82 (d, J = 6.4, 6H), 0.57 – 0.49 (m, 1H). 13C NMR (126 MHz, DMSO-d6) δ = 162.4, 137.7, 137.6, 136.7, 130.7, 52.5, 40.9, 31.0, 19.2. HRMS (ESI) m/z calcd for C12H18NO4S2 [M + H]+ 304.0677, found 304.0676.
3-((3,5-Dimethylpiperidin-1-yl)sulfonyl)thiophene-2-carboxylic acid (S11):
Off-white solid (121 mg, 59% yield). 1H NMR (800 MHz, DMSO-d6) δ 7.85 (d, J = 5.1 Hz, 1H), 7.38 (d, J = 5.2 Hz, 1H), 3.77 – 3.75 (m, 2H), 2.14 (t, J = 11.8 Hz, 2H), 1.69 (d, J = 12.9 Hz, 1H), 1.58 – 1.55 (m, 2H), 0.82 (d, J = 6.7 Hz, 6H), 0.64 (q, J = 12.2 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 161.4, 139.7, 136.3, 130.3, 130.2, 130.1, 130.0, 52.5, 41.3, 31.0, 19.0. HRMS (ESI) m/z calcd for C12H18NO4S2 [M + H]+ 304.0677, found 304.0678.
5-((3,5-Dimethylpiperidin-1-yl)sulfonyl)furan-2-carboxylic acid (S12):
Off-white solid (171 mg, 60% yield). 1H NMR (800 MHz, DMSO-d6) δ 7.32 – 7.31 (m, 1H), 7.23 (d, J = 3.6 Hz, 1H), 3.66 – 3.64 (m, 2H), 2.16 (t, J = 11.7 Hz, 2H), 1.70 – 1.68 (m, 1H), 1.61 – 1.59 (m, 2H), 0.85 (d, J = 6.8 Hz, 6H), 0.65 (q, J = 12.3 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 158.8, 149.8, 148.1, 118.0, 117.6, 52.2, 40.9, 30.9, 18.9. HRMS (ESI) m/z calcd for C12H18NO5S [M + H]+ 288.0906, found 288.0907.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-3-nitrobenzonitrile (I5):
Yellow solid (280 mg, 61% yield). 1H NMR (500 MHz, DMSO-d6) δ 8.26 (d, J = 2.1 Hz, 1H), 7.82 (dd, J = 8.9, 2.2 Hz, 1H), 7.35 (d, J = 8.9 Hz, 1H), 3.25 – 3.21 (m, 2H), 2.54 – 2.49 (m, 2H), 1.77 (d, J = 12.1 Hz, 1H), 1.69 – 1.64 (m, 2H), 0.83 (d, J = 6.5 Hz, 6H), 0.78 (q, J = 12.2 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ 147.8, 138.7, 136.7, 131.8, 121.6, 118.4, 99.6, 57.3, 41.4, 31.1, 19.1. HRMS (ESI) m/z calcd for C14H18N3O4S [M + H]+ 324.1018, found 324.1015.
4-((3,5-dimethylpiperidin-1-yl)sulfonyl)-3-methoxybenzonitrile (I6):
Off-white solid (273 mg, 51% yield). 1H NMR (500 MHz, DMSO-d6) δ 7.87 (d, J = 8.1 Hz, 1H), 7.77 (d, J = 1.5 Hz, 1H), 7.55 (dd, J = 8.1, 1.4 Hz, 1H), 3.93 (s, 3H), 3.61 (dd, J = 12.1, 4.0 Hz, 2H), 2.13 – 2.08 (m, 2H), 1.67 (ddt, J = 13.2, 4.1, 2.2 Hz, 1H), 1.55 (dq, J = 11.0, 3.6 Hz, 2H), 0.80 (d, J = 6.6 Hz, 6H), 0.62 (q, J = 12.0 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ 157.0, 131.8, 131.4, 124.7, 118.1, 117.4, 117.0, 57.2, 52.3, 41.3, 31.4, 19.1. HRMS (ESI) m/z calcd for C15H21N2O3S [M + H]+ 309.1273, found 309.1270.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-3-hydroxybenzonitrile (I7):
Off-white solid (158 mg, 72% yield). 1H NMR (500 MHz, Chloroform-d) δ 9.07 (s, 1H), 7.61 (d, J = 8.2 Hz, 1H), 7.32 (d, J = 1.6 Hz, 1H), 7.26 (dd, J = 8.2, 1.6 Hz, 1H), 3.71 – 3.68 (m, 2H), 1.90 – 1.85 (m, 2H), 1.75 – 1.72 (m, 3H), 0.86 (d, J = 6.5 Hz, 6H), 0.58 – 0.50 (m, 1H). 13C NMR (126 MHz, Chloroform-d) δ 155.4, 129.4, 129.4, 124.3, 123.2, 123.2, 122.8, 122.7, 118.2, 117.0, 52.4, 41.0, 30.9, 18.9. HRMS (ESI) m/z calcd for C14H19N2O3S [M + H]+ 295.1116, found 295.1116.
3,5-Dimethyl-1-((4-nitrophenyl)sulfonyl)piperidine (I8):
Yellow solid (214 mg, 53% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.42 (d, J = 8.4 Hz, 2H), 8.02 (d, J = 8.4 Hz, 2H), 3.67 – 3.65 (m, 2H), 1.90 (t, J = 11.4 Hz, 2H), 1.68 – 1.61 (m, 3H), 0.83 (d, J = 6.6 Hz, 6H), 0.57 (q, J = 12.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 150.4, 142.6, 129.2, 129.1, 125.0, 124.9, 52.4, 41.0, 30.9, 18.9. HRMS (ESI) m/z calcd for C13H19N2O4S [M + H]+ 299.1066, found 299.1065.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)aniline (I9):
Off-white solid (115 mg, 64% yield). 1H NMR (800 MHz, DMSO-d6) δ 7.37 (d, J = 9.1 Hz, 2H), 6.67 (d, J = 8.3 Hz, 2H), 5.85 (s, 2H), 3.54 (dd, J = 11.1, 3.3 Hz, 2H), 1.67 – 1.60 (m, 5H), 0.81 (d, J = 6.1 Hz, 6H), 0.49 (q, J = 11.9 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 153.3, 129.6, 129.5, 121.6, 113.3, 113.2, 52.8, 41.3, 30.8, 19.2. HRMS (ESI) m/z calcd for C13H21N2O2S [M + H]+ 269.1324, found 269.1326.
4-(N-(Prop-2-yn-1-yl)sulfamoyl)benzoic acid (I10):
Off-white solid (173 mg, 63% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.19 (t, J = 6.0 Hz, 1H), 8.11 – 8.10 (m, 2H), 7.93 – 7.92 (m, 2H), 3.76 (d, J = 3.6 Hz, 2H), 2.94 (s, 1H). 13C NMR (201 MHz, DMSO-d6) δ 166.6, 144.8, 134.7, 130.3, 127.3, 79.6, 75.0, 32.3. HRMS (ESI) m/z calcd for C10H10NO4S [M + H]+ 240.0331, found 240.0328.
4-(N-(Prop-2-yn-1-yl)sulfamoyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (I11):
Off-white solid (45 mg, 24% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.37 (t, J = 5.9 Hz, 1H), 8.21 – 8.16 (m, 4H), 7.99 – 7.96 (m, 4H), 3.77 (dd, J = 5.9, 2.5 Hz, 2H), 3.03 (t, J = 2.5 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 164.5, 160.3, 158.7, 144.8, 136.1, 132.0 (q, J = 34.2 Hz), 129.5, 127.5, 127.4, 127.3, 127.1, 126.9, 126.8, 126.6, 126.2, 124.8 (q, J = 273.4 Hz), 79.6, 75.0, 32.4. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C19H14F3N4O4S [M + H]+ 451.0688, found 451.0690.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (HSGN-94):
The synthesis and characterization of HSGN-94 was demonstrated in our previous report27. For in vivo analysis, the compound was scaled up. Purity by HPLC was found to be 98%.
4-(Piperidin-1-ylsulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (1):
Off-white solid (53 mg, 30% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.26 – 8.25 (m, 2H), 8.19 – 8.18 (m, 2H), 7.98 – 7.97 (m, 2H), 7.91 – 7.90 (m, 2H), 3.01 (t, J = 5.5 Hz, 4H), 1.57 (q, J = 5.8 Hz, 4H), 1.41 (t, J = 5.9 Hz, 2H). 13C NMR (201 MHz, DMSO-d6) δ 165.3, 159.9, 159.0, 140.3, 137.0, 132.0 (q, J = 32.2 Hz), 129.7, 127.9, 127.5, 127.3, 126.7, 124.8 (q, J = 271.4 Hz), 46.9, 25.1, 23.3. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C21H20F3N4O4S [M + H]+ 481.1157, found 481.1155. Purity by HPLC was found to be 98%.
4-(Pyrrolidin-1-ylsulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (2):
Off-white solid (32 mg, 18% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.33 – 8.32 (m, 2H), 8.14 – 8.12 (m, 2H), 7.92 – 7.88 (m, 4H), 3.20 – 3.18 (m, 4H), 1.68 (d, J = 6.5 Hz, 4H). 13C NMR (201 MHz, DMSO-d6) δ 171.4, 168.7, 156.8, 141.8, 139.4, 131.4 (q, J = 32.2 Hz), 129.8, 128.2, 127.5, 126.9, 126.6, 124.9 (q, J = 271.4 Hz), 48.2, 25.1. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C20H18F3N4O4S [M + H]+ 467.1001, found 467.1000. Purity by HPLC was found to be 99%.
4-(N-cyclopropylsulfamoyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (3):
Off-white solid (41 mg, 22% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.24 – 8.23 (m, 2H), 8.19 – 8.18 (m, 2H), 7.99 – 7.97 (m, 4H), 2.23 (tq, J = 6.8, 3.3 Hz, 1H), 0.53 (dd, J = 7.1, 2.2 Hz, 2H), 0.42 – 0.41 (m, 2H). 13C NMR (201 MHz, DMSO-d6) δ 165.2, 160.0, 158.9, 144.6, 136.5, 132.0 (q, J = 32.2 Hz), 129.6, 127.5, 127.3, 126.8, 126.2, 124.8 (q, J = 273.4 Hz), 24.6, 5.5. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C19H16F3N4O4S [M + H]+ 453.0844, found 453.0844. Purity by HPLC was found to be 95%
4-(N,N-Dimethylsulfamoyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (4):
Off-white solid (49 mg, 26% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.27 – 8.26 (m, 2H), 8.19 – 8.18 (m, 2H), 7.98 – 7.97 (m, 2H), 7.94 – 7.93 (m, 2H), 2.70 (s, 6H). 13C NMR (201 MHz, DMSO-d6) δ 165.3, 160.0, 158.9, 139.4, 137.0, 132.0 (q, J = 32.2 Hz), 129.7, 128.0, 127.5, 127.3, 126.8, 124.8 (q, J = 271.4 Hz), 37.9. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C18H16F3N4O4S [M + H]+ 441.0844, found 441.0847. Purity by HPLC was found to be 99%.
4-((2-Azabicyclo[2.2.1]heptan-2-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (5):
Off-white solid (26 mg, 15% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.24 – 8.23 (m, 2H), 8.19 – 8.18 (m, 2H), 7.99 – 7.97 (m, 4H), 4.19 (s, 1H), 3.10 (dd, J = 9.0, 2.8 Hz, 1H), 3.03 (d, J = 9.1 Hz, 1H), 2.49 (s, 1H), 1.61 – 1.57 (m, 3H), 1.35 – 1.34 (m, 1H), 1.23 (d, J = 9.9 Hz, 1H), 0.89 (dt, J = 9.9, 2.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 165.3, 160.0, 159.0, 142.4, 136.8, 132.0 (q, J = 32.2 Hz), 129.7, 127.7, 127.5, 127.3, 126.8, 124.8 (q, J = 271.4 Hz), 60.3, 54.7, 37.4, 36.6, 31.3, 27.1. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C22H20F3N4O4S [M + H]+ 493.1157, found 493.1156. Purity by HPLC was found to be 98%.
4-((4-Methylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (6):
Off-white solid (40 mg, 23% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.26 – 8.25 (m, 2H), 8.18 – 8.17 (m, 2H), 7.98 – 7.97 (m, 2H), 7.92 – 7.91 (m, 2H), 3.67 (d, J = 11.9 Hz, 2H), 2.38 – 2.35 (m, 2H), 1.67 – 1.65 (m, 2H), 1.36 – 1.34 (m, 1H), 1.16 (qd, J = 12.1, 3.9 Hz, 2H), 0.87 (d, J = 6.6 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 165.2, 159.9, 159.0, 140.4, 137.0, 132.0 (q, J = 32.2 Hz), 129.7, 127.9, 127.5, 127.3, 126.8, 124.8 (q, J = 273.4 Hz), 46.3, 33.3, 29.6, 21.5. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C22H22F3N4O4S [M+H]+ 495.1310, found 495.1306. Purity by HPLC was found to be 97%.
4-((3-Methylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (7):
Off-white solid (36 mg, 21% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.26 – 8.25 (m, 2H), 8.19 – 8.18 (m, 2H), 7.98 – 7.97 (m, 2H), 7.92 – 7.91 (m, 2H), 3.55 (ddd, J = 16.2, 11.9, 5.7 Hz, 2H), 2.40 (td, J = 11.6, 2.8 Hz, 1H), 2.09 – 2.06 (m, 1H), 1.70 (dt, J = 13.5, 3.6 Hz, 1H), 1.67–1.62 (m, 2H), 1.50 – 1.45 (m, 1H), 0.93 (dtd, J = 14.8, 11.5, 3.7 Hz, 1H), 0.88 (d, J = 6.4 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 165.2, 159.9, 159.1, 140.4, 137.1, 131.9 (q, J = 32.2 Hz), 129.7, 127.8, 127.6, 127.3, 126.8, 124.8 (q, J = 271.4 Hz), 53.0, 46.5, 31.7, 30.5, 24.5, 18.9. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C22H22F3N4O4S [M+H]+ 495.1310, found 495.1312. Purity by HPLC was found to be 98%
4-((2-Methylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (8):
Off-white solid (24 mg, 14% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.22 – 8.21 (m, 2H), 8.19 – 8.18 (m, 2H), 7.98 – 7.97 (m, 4H), 4.18 – 4.16 (m, 1H), 3.68 – 3.65 (m, 1H), 3.05 (td, J = 13.1, 2.7 Hz, 1H), 1.58 – 1.51 (m, 2H), 1.44 (dq, J = 9.2, 5.6, 5.0 Hz, 3H), 1.25 (dt, J = 12.8, 4.3 Hz, 1H), 1.06 (d, J = 6.9 Hz, 3H). 13C NMR (201 MHz, DMSO-d6) δ 165.2, 160.0, 159.0, 145.0, 136.6, 132.0 (q, J = 32.2 Hz), 129.8, 127.5, 127.3, 127.2, 126.8, 126.8, 124.8 (q, J = 273.4 Hz), 48.8, 30.3, 25.1, 18.0, 15.9. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C22H22F3N4O4S [M+H]+ 495.1310, found 495.1311. Purity by HPLC was found to be 99%.
5-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)nicotinamide (9):
Off-white solid (37 mg, 22% yield). 1H NMR (800 MHz, DMSO-d6) δ = 9.40 (d, J=2.1, 1H), 9.12 (d, J=2.1, 1H), 8.67 (s, 1H), 8.17 (d, J=8.1, 2H), 7.99 (d, J=8.2, 2H), 3.69 (dd, J=11.4, 3.8, 2H), 1.89 (t, J=11.3, 2H), 1.66 (d, J=10.8, 3H), 0.82 (d, J=6.4, 6H), 0.57 – 0.50 (m, 1H). 13C NMR (201 MHz, DMSO-d6) δ 164.1, 159.6, 159.0, 153.3, 150.9, 135.1, 133.7, 132.0 (q, J = 32.2 Hz), 129.8, 127.4, 127.3, 126.8, 124.8 (q, J = 273.4 Hz), 52.3, 41.0, 31.0, 19.0. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C22H23F3N5O4S [M+H]+ 510.1417, found 510.1415. Purity by HPLC was found to be 99%
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)thiophene-2-carboxamide (10):
Off-white solid (49 mg, 29% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.54 (s, 1H), 8.35 (s, 1H), 8.15 (d, J = 8.0 Hz, 2H), 7.98 (d, J = 8.0 Hz, 2H), 3.59 (dd, J = 11.2, 3.8 Hz, 2H), 1.85 (t, J = 11.2 Hz, 2H), 1.68 – 1.61 (m, 3H), 0.83 (d, J = 6.4 Hz, 6H), 0.64 – 0.44 (m, 1H). 13C NMR (201 MHz, DMSO-d6) δ 160.0, 159.6, 159.0, 141.0, 137.5, 131.9 (q, J = 32.2 Hz), 129.4, 127.6, 127.3, 126.8, 126.2, 124.8 (q, J = 271.4 Hz), 52.5, 41.1, 31.0, 19.1. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C21H22F3N4O4S2 [M+H]+ 515.1029, found 515.1029. Purity by HPLC was found to be 96%.
3-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)thiophene-2-carboxamide (11):
Off-white solid (30 mg, 18% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.14 – 8.13 (m, 2H), 7.97 – 7.96 (m, 2H), 7.91 (d, J = 5.2 Hz, 1H), 7.38 (d, J = 5.2 Hz, 1H), 3.72 (dd, J = 12.2, 4.0 Hz, 2H), 2.08 (t, J = 11.7 Hz, 2H), 1.68 (d, J = 13.1 Hz, 1H), 1.61 – 1.58 (m, 2H), 0.84 (d, J = 6.6 Hz, 6H), 0.63 (q, J = 12.1 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 159.6, 158.3, 139.5, 136.7, 131.9 (q, J = 32.2 Hz), 129.5, 127.9, 127.5, 127.3, 126.8, 126.7, 124.8 (q, J = 273.4 Hz), 52.4, 41.2, 30.9, 19.0. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C21H22F3N4O4S2 [M+H]+ 515.1029, found 515.1026. Purity by HPLC was found to be 96%.
5-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)furan-2-carboxamide (12):
Off-white solid (38 mg, 22% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.17 – 8.16 (m, 2H), 7.98 – 7.97 (m, 2H), 7.62 (d, J = 3.8 Hz, 1H), 7.32 (d, J = 3.6 Hz, 1H), 3.70 (dd, J = 12.0, 4.1 Hz, 2H), 2.22 (t, J = 11.7 Hz, 2H), 1.71 (d, J = 13.4 Hz, 1H), 1.64 – 1.60 (m, 2H), 0.86 (d, J = 6.6 Hz, 6H), 0.66 (q, J = 12.3 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 159.5, 158.9, 156.3, 150.0, 149.5, 132.0 (q, J = 30.2 Hz), 127.5, 127.3, 126.8, 124.8 (q, J = 273.4 Hz), 117.9, 117.7, 117.3, 52.3, 40.9, 30.9, 18.9. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C21H22F3N4O5S [M+H]+ 499.1263, found 499.1262. Purity by HPLC was found to be 97%.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-3-nitro-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (13):
Yellow solid (42 mg, 26% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.53 (s, 1H), 8.17 – 8.16 (m, 2H), 8.14 – 8.12 (m, 1H), 7.96 – 7.95 (m, 2H), 7.36 (d, J = 8.8 Hz, 1H), 3.29 – 3.27 (m, 2H), 2.54 (t, J = 12.0 Hz, 2H), 1.81 (d, J = 13.0 Hz, 1H), 1.75 – 1.74 (m, 1H), 0.88 (d, J = 6.6 Hz, 6H), 0.78 (q, J = 12.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 163.4, 160.1, 159.0, 148.2, 139.1, 133.6, 131.9 (q, J = 32.2 Hz), 127.6, 127.4, 127.2, 126.8, 126.7, 124.8 (q, J = 271.4 Hz), 120.7, 57.7, 41.5, 31.0, 19.0. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C23H23F3N5O6S [M+H]+ 554.1321, found 554.1322. Purity by HPLC was found to be 95%.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-3-methoxy-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (14):
Off-white solid (31 mg, 19% yield). 1H NMR (500 MHz, DMSO-d6) δ 8.17 – 8.16 (m, 2H), 7.99 – 7.98 (m, 2H), 7.90 – 7.88 (m, 1H), 7.81 (d, J = 1.6 Hz, 1H), 7.71 – 7.69 (m, 1H), 3.97 (s, 3H), 3.64 (dd, J = 12.1, 4.0 Hz, 2H), 2.13 – 2.07 (m, 2H), 1.68 (d, J = 13.0 Hz, 1H), 1.56 (ddt, J = 14.5, 11.3, 4.6 Hz, 2H), 0.81 (d, J = 6.6 Hz, 6H), 0.62 (q, J = 12.2 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ 164.0, 158.7, 156.9, 137.9, 131.3 (q, J = 32.2 Hz), 127.4, 127.0, 125.3, 123.1 (q, J = 271.4 Hz), 120.7, 113.3, 56.8, 52.5, 41.4, 31.4, 19.1. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C24H26F3N4O5S [M+H]+ 539.1576, found 539.1577. Purity by HPLC was found to be 95%.
3-Amino-4-((3,5-dimethylpiperidin-1-yl)sulfonyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (15):
Off-white solid (34 mg, 73% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.18 – 8.17 (m, 2H), 7.96 – 7.95 (m, 2H), 7.37 (d, J = 2.2 Hz, 1H), 7.34 – 7.32 (m, 1H), 6.97 (d, J = 8.1 Hz, 1H), 3.13 (dd, J = 11.3, 3.5 Hz, 2H), 2.09 (t, J = 11.0 Hz, 2H), 1.84 – 1.78 (m, 3H), 0.89 (d, J = 6.8 Hz, 6H), 0.68 (q, J = 12.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 165.8, 160.3, 159.2, 143.6, 142.4, 131.8 (q, J = 32.2 Hz), 127.7, 127.5, 127.2, 126.7, 124.8 (q, J = 271.4 Hz), 119.1, 117.9, 114.6, 58.3, 42.3, 31.5, 19.5. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C23H25F3N5O4S [M+H]+ 524.1579, found 524.1579. Purity by HPLC was found to be 95%.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-3-hydroxy-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (16):
Off-white solid (38 mg, 23% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.16 – 8.15 (m, 2H), 7.98 (d, J = 8.4 Hz, 2H), 7.89 (d, J = 8.1 Hz, 1H), 7.81 – 7.69 (m, 1H), 7.57 – 7.54 (m, 2H), 3.67 – 3.63 (m, 2H), 2.12 – 2.06 (m, 2H), 1.67 (d, J = 13.0 Hz, 1H), 1.57 (q, J = 8.8, 7.7 Hz, 2H), 0.80 (d, J = 6.6 Hz, 6H), 0.62 – 0.54 (m, 1H). 13C NMR (201 MHz, DMSO-d6) δ 165.3, 160.2, 158.7, 155.8, 138.3, 132.1 (q, J = 32.2 Hz), 131.1, 128.5, 127.4, 126.7, 123.3 (q, J = 271.4 Hz), 118.6, 117.7, 52.5, 41.3, 31.1, 18.9. 19F NMR (471 MHz, DMSO-d6) δ −62.7 (s, 3F). HRMS (ESI) m/z calcd for C23H24F3N4O5S [M+H]+ 525.1420, found 525.1419. Purity by HPLC was found to be 95%.
4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)-N-methyl-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (17):
Off-white solid (29 mg, 57% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.38 – 8.37 (m, 2H), 8.12 – 8.11 (m, 2H), 8.00 – 7.99 (m, 2H), 7.86 – 7.84 (m, 2H), 3.75 (s, 3H), 3.66 (dd, J = 11.3, 3.8 Hz, 2H), 1.82 (t, J = 11.4 Hz, 3H), 1.67 – 1.62 (m, 3H), 0.83 (d, J = 6.6 Hz, 6H), 0.55 (q, J = 12.1, 11.5 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 170.1, 156.0, 154.3, 140.9, 132.3 (q, J = 32.2 Hz), 130.3, 128.8, 128.0, 127.1, 126.9, 126.8, 124.7 (q, J = 271.4 Hz), 52.6, 41.1, 34.9, 30.9, 19.0. 19F NMR (471 MHz, DMSO-d6) δ −62.8 (s, 3F). HRMS (ESI) m/z calcd for C24H26F3N4O4S [M+H]+ 523.1627, found 523.1629. Purity by HPLC was found to be 95%.
N-(4-((3,5-Dimethylpiperidin-1-yl)sulfonyl)phenyl)-5-phenyl-1,3,4-oxadiazole-2-carboxamide (18):
Off-white solid (90 mg, 39% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.14 – 8.13 (m, 2H), 8.10 – 8.08 (m, 2H), 7.79 – 7.78 (m, 2H), 7.72 – 7.70 (m, 1H), 7.70 – 7.66 (m, 2H), 3.64 – 3.62 (m, 2H), 1.78 (t, J = 11.4 Hz, 2H), 1.66 – 1.62 (m, 3H), 0.84 (d, J = 6.5 Hz, 6H), 0.55 – 0.51 (m, 1H). 13C NMR (201 MHz, DMSO-d6) δ 165.9, 158.8, 152.3, 141.9, 133.2, 132.4, 129.9, 128.8, 127.6, 123.1, 121.3, 52.7, 41.1, 30.9, 19.1. HRMS (ESI) m/z calcd for C22H25N4O4S [M+H]+ 441.1591, found 441.1592. Purity by HPLC was found to be 96%.
N-(5-cyclohexyl-1,3,4-oxadiazol-2-yl)-4-((3,5-dimethylpiperidin-1-yl)sulfonyl)benzamide (19):
Off-white solid (45 mg, 32% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.21 – 8.20 (m, 2H), 7.89 – 7.88 (m, 2H), 3.67 (dd, J = 11.6, 3.9 Hz, 2H), 2.95 (ddd, J = 10.8, 7.0, 3.8 Hz, 1H), 2.03 (dd, J = 13.2, 4.0 Hz, 2H), 1.83 (t, J = 11.4 Hz, 2H), 1.77 (dt, J = 13.3, 4.1 Hz, 2H), 1.74 – 1.64 (m, 4H), 1.63 (qd, J = 11.4, 3.5 Hz, 2H), 1.44 (tdd, J = 15.4, 11.8, 3.6 Hz, 2H), 1.32 – 1.27 (m, 1H), 0.83 (d, J = 6.5 Hz, 6H), 0.55 (q, J = 12.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 166.3, 165.7, 158.2, 140.3, 137.5, 129.7, 127.8, 52.6, 41.1, 34.6, 30.9, 29.7, 25.6, 24.9, 19.0. HRMS (ESI) m/z calcd for C22H31N4O4S [M+H]+ 447.2066, found 447.2062. Purity by HPLC was found to be 95%.
N-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)-4-((3,5-dimethylpiperidin-1-yl)sulfonyl)benzamide (20):
Off-white solid (30 mg, 24% yield). 1H NMR (800 MHz, DMSO-d6) δ 8.20 – 8.19 (m, 2H), 7.89 – 7.88 (m, 2H), 3.67 – 3.65 (m, 2H), 2.21 – 2.18 (m, 1H), 1.84 (t, J = 11.3 Hz, 2H), 1.67 – 1.62 (m, 3H), 1.14 – 1.13 (m, 2H), 1.02 – 1.01 (m, 2H), 0.83 (dd, J = 6.6, 1.3 Hz, 6H), 0.54 (q, J = 12.0 Hz, 1H). 13C NMR (201 MHz, DMSO-d6) δ 164.8, 157.8, 140.3, 137.2, 129.6, 128.7, 127.8, 52.6, 41.1, 30.9, 19.0, 7.8, 6.2. HRMS (ESI) m/z calcd for C19H25N4O4S [M+H]+ 405.1597, found 405.1596. Purity by HPLC was found to be 97%.
4-(N-((1-(13-Oxo-17-(2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-3,6,9-trioxa-12-azaheptadecyl)-1H-1,2,3-triazol-4-yl)methyl)sulfamoyl)-N-(5-(4-(trifluoromethyl)phenyl)-1,3,4-oxadiazol-2-yl)benzamide (HSGN-Probe):
Clear oil (21 mg, 77% yield). 1H NMR (800 MHz, Methanol-d4) δ 8.23 – 8.19 (m, 4H), 8.00 – 7.95 (m, 2H), 7.90 – 7.88 (m, 2H), 7.82 (s, 1H), 4.49 (d, J = 4.2 Hz, 2H), 4.30 (dd, J = 7.8, 4.3 Hz, 1H), 4.25 (s, 2H), 3.82 (t, J = 4.7 Hz, 2H), 3.60 – 3.59 (m, 8H), 3.53 (t, J = 5.4 Hz, 2H), 3.20 (dt, J = 9.5, 4.9 Hz, 1H), 2.92 – 2.89 (m, 1H), 2.70 – 2.67 (m, 1H), 2.20 – 2.18 (m, 2H), 1.73 – 1.57 (m, 5H), 1.55 – 1.38 (m, 3H), 1.30 – 1.25 (m, 2H). 13C NMR (201 MHz, Methanol-d4) δ 174.7, 165.1, 164.6, 159.4, 158.8, 144.4, 143.6, 136.4, 132.8 (q, J = 34.2 Hz), 128.8, 126.9, 126.0, 124.4 (q, J = 271.4 Hz), 123.9, 70.1, 70.0, 69.9, 69.8, 69.1, 68.8, 61.9, 60.1, 55.5, 49.8, 39.6, 38.8, 37.7, 35.3, 28.3, 28.0, 25.4. 19F NMR (471 MHz, Methanol-d4) δ −65.7 (s, 3F). HRMS (ESI) m/z calcd for C37H46F3N10O9S2 [M+H]+ 895.2843, found 895.2846. Purity by HPLC was found to be 91%.
Biological analysis
Bacterial strains, media, cell lines and reagents
Clinical isolates used in this study (Table S5) were obtained from the Biodefense and Emerging Infections Research Resources Repository (BEI Resources) and the American Type Culture Collection (ATCC). Cation-adjusted Mueller Hinton broth (CAMHB), tryptic soy broth (TSB) and tryptic soy agar (TSA) were purchased from Becton, Dickinson and Company (Cockeysville, MD, USA). Human keratinocyte cell line (HaCaT) was obtained from AddexBio (San Diego, CA, USA). Dulbecco’s Modified Eagle Medium (DMEM) was obtained from Sigma-Aldrich (St. Louis, MO, USA). Fetal bovine serum (FBS) and phosphate-buffered saline (PBS) were purchased from Corning (Manassas, VA, USA). Linezolid and vancomycin were purchase from Chem-Impex International (Wood Dale, IL, USA). Compounds were synthesized from commercial sources in our laboratory and prepared in stock solutions in DMSO.
Determination of the MICs against clinically-important Gram-positive bacteria
MICs of 1,3,4-oxadiazol-2-yl benzamides were determined using the broth microdilution method as outlined previously77. Briefly, a bacterial solution equivalent to 0.5 McFarland standard was prepared and diluted in CAMHB to achieve a bacterial concentration of about 5 × 105 CFU/mL and seeded in 96-well plates. Streptococci, enterococci and Listeria were diluted in TSB. Serial dilutions of tested agents were incubated with the bacteria aerobically at 37° C for 18–20 hours (except for S. pneumoniae which was incubated in presence of 5% CO2). MICs were determined as the lowest concentration of the each test agent that completely inhibited the bacterial growth as determined visually.
In vitro cytotoxicity analysis of HSGN-94 against human keratinocytes (HaCat) cells
Cytotoxicity assessment for HSGN-94 was determined as previously described78–79. HSGN-94 was assayed (at concentrations of 16, 32, 64 and 128 μg/mL) against human keratinocyte cells (HaCat) to determine its potential toxic effect to mammalian skin cells in vitro. Briefly, cells were incubated with the compound (in sextuplicate) at 37 °C with 5% CO2 for 2 hours. DMSO was included as a control to determine the baseline measure of the cytotoxic impact of the compound. MTS 3-(4, 5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (Promega, Madison, WI, USA) was subsequently added and the plate was incubated for three hours before the absorbance readings (at OD490) were recorded.
Multi-step Resistance Selection:
To assess if MRSA USA300 could form resistance to HSGN-94, a multi-step serial passaging experiment was conducted for 65 days, as described previously25, 80–81. Resistance was considered as a greater than four-fold increase in the MIC as compared to the initial MIC.
Proteomics Analysis:
HSGN-94 (0.25 μg/mL) or a corresponding quantity of DMSO was added to a culture of S. aureus ATCC 25923 at OD600 0.6. The samples were treated for 2 hours at 37°C and then normalized to OD600 1.1 using sterile saline. Next, samples were centrifuged and then washed with PBS (2×). Both LC/MS/MS preparation and acquisition of data were executed as previously described82. Protein extraction was peformed using a Barocycler NEP2320 (Pressure Biosciences, South Easton, MA) system in which samples were lysed in 100 mM ammonium bicarbonate at 5 °C for 90 cycles (each cycle with 50 seconds at 35 kpsi and 10 seconds at atmospheric pressure). Protein concentration was measured by Bicinchoninic Acid (BCA) assay, and 50 μg (equivalent volume) of lysate was precipitated using four volume of cold (−20°C) 100% acetone overnight. Protein pellets were obtained via centrifugation of samples and then re-suspended in 8M urea with 10 mM DTT followed by incubation for 45 minutes. Next, cysteine alkylation was performed in the dark at room tempeature for 45 minutes with 20 mM iodoacetamide followed by the addition of 5 mM DTT and incubation at 37 °C for 20 minutes. Dilution of alkylated and reduced samples was executed to lessen the urea concentrataion to 1.5 M followed by overnight digestion at 37°C via 1:25 (w/w) enzyme-protein ratio using Trypsin/Lys-C Mix (Promega, Madison, WI, USA). Peptide desaltation was then performed by passing the digested peptides through C18 silica micro spin columns (The Nest Group Inc., Southborough, MA, USA). Then, the BCA assay was used to determine concentration of peptides as described above. The peptide concentration was altered to 0.2 μg/μL, and ~1 μg (5 μL) was used as injection volume for LC-MS/MS. A reverse-phase HPLC-ESI-MS/MS system containing an UltiMate™ 3000 RSLCnano system joined to a Q-Exactive (QE) High Field (HF) Hybrid Quadrupole-Orbitrap™ mass spectrometer (Thermo Fisher Scientific, Waltham, MA) and a Nano-spray Flex™ ion source (Thermo Fisher Scientific) was used to aquire the data. A resolution of 15,000 at m/z 200 was used for MS/MS scan acquisition and a 30 second dyanmic exclusion time was set to circumvent indistinguishable peptides. Both the MaxQuant83–85 and Perseus35 softwares were utilized for data and bioinformatics analysis respectively, as previously described82. GO function analysis was performed using Metascape™ software.
Total RNA isolation and RT-PCR:
HSGN-94 (0.25 μg/mL) or DMSO was added to a culture of S. aureus ATCC 25923 at OD600 0.6 in triplicates. The samples were incubated for 2 hours at 37°C and then centrifuged to obtain sample pellets. RT-PCR was executed using a BioRad CFX96™ Touch Real-Time PCR detection system follwing a procedure that has been previously described82. The PCR primers used were either contructed with Primer-BLAST or acquied from literature (Table S6). 16S rRNA was used as reference for data normalization. Statistical significance was sought by means of student’s t-test.
Pull-Down Assay:
The pull down assay and proteomic analysis was executed using a procedure outlined previously86. Breifly, 50 mL of an overnight culture of S. aureus ATCC 25923 was grown to OD600 = 1. This culture was pelleted by centrifugation and resuspended in 1 mL 10 mM Tris·HCl (pH 7.5), 50 mM NaCl buffer containing EDTA-free complete protease inhibitor (Roche). These cells were combined with 0.1-mm glass beads and lysed twice for 45 seconds using a Fast-Prep machine (MP Biomedicals). Samples were centrifuged at 17,000 × g for 5 minutes followed by centrifugation at 100,000 × g for 1 hour in order to obtain protein extracts. 40 μL streptavidin dynabeads (Invitrogen) coupled with either 2.4 μM HSGN-Probe or 2.4 μM HSGN-Probe plus 50 μM HSGN-94 were incubated at room temperature for 30 minutes with 1.2 mg cytoplasmic proteins in sample buffer prepared with 1.5 mL 10% glycerol, 1 mM MgCl2, 230 mM NaCl, 0.5 mM DTT, 5 mM Tris (pH 7.5), and 4 mM EDTA containing 50 μg/mL BSA. After incubation, samples were washed 4 × with the sample buffer lacking BSA and suspended in 50 μL protein sample buffer. Next, samples were heated to boiling for 5 min and then the beads were removed. 18 μL of sample were run on 12% (wt/vol) SDS/PAGE gels which were then stained using the SilverQuest kit (Invitrogen). This solution then underwent LC/MS/MS analysis as described above. Data aqusition was performed using MaxQuant and bioinformatic analysis was done using Perseus as mentioned above.
Membrane Lipid Extraction and TLC Analysis:
Exponentially growing S. aureus ATCC 25923 was treated with either DMSO or HSGN-94 (0.25 ×, 1× and 8× MIC) for 5 hours at 37°C. The samples were normalized to OD600 0.9 and then centrifuged at 10,000 rpm for 10 minutes. The cells were collected and washed twice with 0.1 M sodium acetate (pH 4.7). Next, lipids were extracted following the Bligh-Dyer method87 and lipids were isolated and dried over nitrogen stream. Detection of glycolipids was performed via TLC using a previously reported procedure52. MALDI-TOF analysis was used to identify glycolipids on TLC. See SI for MALDI mass spectra.
Multiple reaction monitoring profiling (MRM-Profiling) of Lipids
S. aureus ATCC 25923 (at OD600 0.6) was treated with DMSO, 0.25 μg/mL (1× MIC) HSGN-94, or 2 μg/mL (8× MIC) HSGN-94 for 5 hours at 37°C. The samples were normalized to OD600 0.9 and then centrifuged at 10,000 rpm for 10 minutes. The cells were collected and washed twice with 0.1 M sodium acetate (pH 4.7). Next, lipids were extracted following the Bligh-Dyer method87 and lipids were isolated and dried over nitrogen stream. Experiments were carried out following a previously reported procedure54. Briefly, data was acquired using a triple quadrupole mass spectrometer, Agilent QQQ 6410 (Santa Clara, CA), equipped with Agilent G1367A 1100 series autosampler (Santa Clara, CA). Samples were introduced into the mass spectrometer by direct flow injection ESI at a flow rate of 7 μL/min. PCA plots were constructed using MetaboAnalyst 5.0.
Macromolecular biosynthesis assay:
The inhibition of macromolecules by HSGN-94 at (0.125–4 × MIC) were assayed in triplicate via scintillation counting using the previously reported procedure88.
MRSA murine skin infection model:
The study was approved by the Purdue Animal Care and Use Committee and carried out following the guide of the National Institutes of Health for the Care and Use of Laboratory Animals. female Balb/c mice 8-weeks old (Jackson laboratories, ME, USA), weighing on average 20 g, were used for this study. MRSA murine skin infection model was performed as described in previous reports89–91. Brielfy, the dorsal region of each mosue mice was shaved and disinfected with ethanol and two excisional 6-mm diameter wounds were formed in each mouse using a 6 mm-diameter sterile biopsy punch (Sklar instruments, PA, USA) under isoflurane anesthesia. A subcutaneous injection of buprenorphine (0.03 mg/kg) was administered immediately before the application of biopsy punches to reduce pain. Excisional wounds were infected with 20 μL (per each wound) of 4.2×109 CFU/mL of MRSA USA300. The wounds was covered with Tegaderm film (3M, MN, USA) that is fixed with Uro-Bond V silicone adhesive (Urocare, CA, USA). Two days after infection, mice were randomly divided into groups (n=5) to receive treatments. One group was injected clindamycin I.P. (25 mg/kg once daily). The formed wounds for the remaining groups received topical treatment (twice daily) with either 2% HSGN-94, petroleum jelly (vehicle), or 2% mupirocin,. After each treatment, Tegaderm was applied over the wounds. After five days of treatment, mice were humanely euthanized via CO2 asphyxiation. The skin tissues of the wounds were harvested aseptically. For each mouse, the left wound was fixed in 10% formalin for histopathological analysis and the right wound was homogenized in sterile PBS, and plated selective MRSA plates (mannitol salt agar). The plates were incubated at 37°C before bacterial colonies were enumerated.
Expression of Proinflammatory Cytokines:
Skin homogenates obtained from the right wound of each mouse were centrifuged (10,000 rpm for 10 minutes) and the supernatant was transferred to a separate tube. For each treatment group, 100 μL aliquots from each sample (from all five mice) were pooled together. The total protein content for each sample was measured via the bicinchoninic acid assay, standardized, and expression of cytokines and growth factors was subsequently determined via the Quantibody Mouse cytokine array 4000 kit (RayBiotech Life, Norcross, GA). The concentrations of cytokines (pg/mL) in each treatment group was calculated and presented as a bar graph using GraphPad Prism 8.0 (La Jolla, CA).
Evlauation of Inflammation via Histopathology:
The left wound of infected mice in all the four groups (vehicle, HSGN-94, mupirocin and clindamycin) were evaluated histologically. Tissues were processed and sections (4-μm thick) were prepared on slides and stained with H & E. Slides were scanned using Aperio® VERSA as a whole glass slide scanner and analyzed by a board certified veterinary pathologist.
In Silico PAINS Analysis
All synthesized compounds were assesed for PAINS using SwissADME program which demonstrated no evidence of PAINS92.
Supplementary Material
Acknowledgements:
Proteomics experiments including sample preparation, data collection and analysis was done at the Purdue Proteomics Facility. The Q Exactive Orbitrap HF mass spectrometer used for LC-MS/MS analysis was purchased through funding from the Purdue Executive Vice President for Research and Partnership (EVPRP).
Sources of Funding
Research reported in this publication was supported by Purdue University, and the National Institute of Allergy And Infectious Diseases of the National Institutes of Health under Award Numbers T32AI148103 (G.A.N.) and R01AI130186 (M.N.S.).
Abbreviations
- ATCC
American type culture collection
- BHIS
brain heart infusion supplemented
- BOP
benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate
- CLSI
clinical and laboratory standards institute
- DIPEA
diisopropylethylamine
- DMEM
dulbecco’s modified eagle medium
- DMF
dimethylformamide
- DMSO
dimethyl sulfoxide
- FBS
fetal bovine serum
- MIC
minimum inhibitory concentration
- MRSA
methicillin-resistant S. aureus
- MTS
3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)
- NaOAc
sodium acetate
- PAINS
pan assay interference compounds
- PBS
phosphate buffered saline
- RT
room temperature
- TSA
tryptic soy agar
- TSB
tryptic soy broth
Footnotes
Supporting Information
- Synthetic Schemes
- Tables of MICs of HSGN-94 against various bacterial strains
- Cytotoxicity of HSGN-94 against mammalian cells
- Table of proteins identified from pull-down assay
- Bacterial strains used in study
- Sequence of primers used in RT-PCR
- MALDI mass spectra of glycolipid TLC samples
- 1H NMR, 13C NMR, and 19F NMR spectra of analogs
- HPLC traces of analogs
- Molecular SMILES strings and MIC values
The authors declare no competing financial interest.
Data Availability:
All raw LC-MS/MS data can be found in the Mass Spectrometry Interactive Virtual Environment (http://massive.ucsd.edu) with the ID: MSV000088342.
References:
- 1.Tong SYC; Davis JS; Eichenberger E; Holland TL; Fowler VG, Staphylococcus aureus Infections: Epidemiology, Pathophysiology, Clinical Manifestations, and Management. Clin. Microbiol. Rev 2015, 28 (3), 603. DOI: 10.1128/CMR.00134-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chambers HF; Deleo FR, Waves of Resistance: Staphylococcus aureus in the Antibiotic Era. Nat. Rev. Microbiol 2009, 7 (9), 629–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cosgrove SE; Sakoulas G; Perencevich EN; Schwaber MJ; Karchmer AW; Carmeli Y, Comparison of Mortality Associated with Methicillin-Resistant and Methicillin-Susceptible Staphylococcus aureus Bacteremia: A Meta-analysis. Clin. Infect. Dis 2003, 36 (1), 53–59. [DOI] [PubMed] [Google Scholar]
- 4.Moellering RC Jr., Current Treatment Options for Community-Acquired Methicillin-resistant Staphylococcus aureus Infection. Clin. Infect. Dis 2008, 46 (7), 1032–1037. [DOI] [PubMed] [Google Scholar]
- 5.Rasmussen RV; Fowler VG Jr.; Skov R; Bruun NE, Future Challenges and Treatment of Staphylococcus aureus Bacteremia with Emphasis on MRSA. Future Microbiol. 2011, 6 (1), 43–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Huovinen P, Resistance to Trimethoprim-Sulfamethoxazole. Clin. Infect. Dis 2001, 32 (11), 1608–1614. [DOI] [PubMed] [Google Scholar]
- 7.Azhar A; Rasool S; Haque A; Shan S; Saeed M; Ehsan B; Haque A, Detection of High Levels of Resistance to Linezolid and Vancomycin in Staphylococcus aureus. J. Med. Microbiol 2017, 66 (9), 1328–1331. [DOI] [PubMed] [Google Scholar]
- 8.Roch M; Gagetti P; Davis J; Ceriana P; Errecalde L; Corso A; Rosato AE, Daptomycin Resistance in Clinical MRSA Strains Is Associated with a High Biological Fitness Cost. Front. Microbiol 2017, 8, 2303. DOI: 10.3389/fmicb.2017.02303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Naclerio GA; Sintim HO, Multiple Ways to Kill Bacteria via Inhibiting Novel Cell Wall or Membrane Targets. Future Med. Chem 2020, 12 (13), 1253–1279. [DOI] [PubMed] [Google Scholar]
- 10.Gründling A; Schneewind O, Synthesis of Glycerol Phosphate Lipoteichoic Acid in Staphylococcus aureus. Proc. Natl. Acad. Sci. U.S.A 2007, 104 (20), 8478–8483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Rajagopal M; Walker S, Envelope Structures of Gram-Positive Bacteria. Curr. Top. Microbiol. Immunol 2017, 404, 1–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hesser AR; Matano LM; Vickery CR; Wood BM; Santiago AG; Morris HG; Do T; Losick R; Walker S, The Length of Lipoteichoic Acid Polymers Controls Staphylococcus aureus Cell Size and Envelope Integrity. J. Bacteriol 2020. DOI: 10.1128/JB.00149-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Neuhaus FC; Baddiley J, A Continuum of Anionic Charge: Structures and Functions of D-Alanyl-Teichoic Acids in Gram-Positive Bacteria. Microbiol. Mol. Biol. Rev 2003, 67 (4), 686–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Collins LV; Kristian SA; Weidenmaier C; Faigle M; Van Kessel KP; Van Strijp JA; Gotz F; Neumeister B; Peschel A, Staphylococcus aureus Strains Lacking D-Alanine Modifications of Teichoic Acids are Highly Susceptible to Human Neutrophil Killing and are Virulence Attenuated in Mice. J. Infect. Dis 2002, 186 (2), 214–219. [DOI] [PubMed] [Google Scholar]
- 15.Sheen TR; Ebrahimi CM; Hiemstra IH; Barlow SB; Peschel A; Doran KS, Penetration of the Blood-Brain Barrier by Staphylococcus aureus: Contribution of Membrane-Anchored Lipoteichoic Acid. J. Mol. Med. (Berl) 2010, 88 (6), 633–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Percy MG; Gründling A, Lipoteichoic Acid Synthesis and Function in Gram-Positive Bacteria. Annu. Rev. Microbiol 2014, 68 (1), 81–100. [DOI] [PubMed] [Google Scholar]
- 17.Schirner K; Marles-Wright J; Lewis RJ; Errington J, Distinct and Essential Morphogenic Functions for Wall- and Lipo-teichoic Acids in Bacillus subtilis. EMBO J. 2009, 28 (7), 830–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cabacungan E; Pieringer RA, Mode of Elongation of the Glycerol Phosphate Polymer of Membrane Lipoteichoic Acid of Streptococcus faecium ATCC 9790. J. Bacteriol 1981, 147 (1), 75–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Taron DJ; Childs WC 3rd; Neuhaus FC, Biosynthesis of D-Alanyl-Lipoteichoic Acid: Role of Diglyceride Kinase in the Synthesis of Phosphatidylglycerol for Chain Elongation. J. Bacteriol 1983, 154 (3), 1110–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lu D; Wörmann ME; Zhang X; Schneewind O; Gründling A; Freemont PS, Structure-Based Mechanism of Lipoteichoic Acid Synthesis by Staphylococcus aureus LtaS. Proc. Natl. Acad. Sci. U.S.A 2009, 106 (5), 1584–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Canepari P; Boaretti M, Lipoteichoic Acid as a Target for Antimicrobial Action. Microb. Drug. Resist 1996, 2 (1), 85–89. [DOI] [PubMed] [Google Scholar]
- 22.Naclerio GA; Abutaleb NS; Li D; Seleem MN; Sintim HO, Ultrapotent Inhibitor of Clostridioides difficile Growth, Which Suppresses Recurrence In Vivo. J. Med. Chem 2020, 63 (20), 11934–11944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Naclerio GA; Abutaleb NS; Onyedibe KI; Seleem MN; Sintim HO, Potent Trifluoromethoxy, Trifluoromethylsulfonyl, Trifluoromethylthio and Pentafluorosulfanyl Containing (1,3,4-oxadiazol-2-yl)benzamides Against Drug-Resistant Gram-Positive Bacteria. RSC Med. Chem 2020, 11 (1), 102–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Naclerio GA; Abutaleb NS; Alhashimi M; Seleem MN; Sintim HO, N-(1,3,4-Oxadiazol-2-yl)Benzamides as Antibacterial Agents against Neisseria gonorrhoeae. Int. J. Mol. Sci 2021, 22 (5), 2427. DOI: 10.3390/ijms22052427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Opoku-Temeng C; Naclerio GA; Mohammad H; Dayal N; Abutaleb NS; Seleem MN; Sintim HO, N-(1,3,4-oxadiazol-2-yl)benzamide Analogs, Bacteriostatic Agents Against Methicillin- and Vancomycin-Resistant Bacteria. Eur. J. Med. Chem 2018, 155, 797–805. [DOI] [PubMed] [Google Scholar]
- 26.Naclerio GA; Onyedibe KI; Sintim HO, Lipoteichoic Acid Biosynthesis Inhibitors as Potent Inhibitors of S. aureus and E. faecalis Growth and Biofilm Formation. Molecules 2020, 25 (5), 2277. DOI: 10.3390/molecules25102277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Naclerio GA; Karanja CW; Opoku-Temeng C; Sintim HO, Antibacterial Small Molecules That Potently Inhibit Staphylococcus aureus Lipoteichoic Acid Biosynthesis. ChemMedChem 2019, 14 (10), 1000–1004. [DOI] [PubMed] [Google Scholar]
- 28.Bridy-Pappas AE; Margolis MB; Center KJ; Isaacman DJ, Streptococcus pneumoniae: Description of the Pathogen, Disease Epidemiology, Treatment, and Prevention. Pharmacotherapy 2005, 25 (9), 1193–1212. [DOI] [PubMed] [Google Scholar]
- 29.Brooks LRK; Mias GI, Streptococcus pneumoniae’s Virulence and Host Immunity: Aging, Diagnostics, and Prevention. Front. Immunol 2018, 9, 1366. DOI: 10.3389/fimmu.2018.01366. eCollection 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kourtis AP; Hatfield K; Baggs J; Mu Y; See I; Epson E; Nadle J; Kainer MA; Dumyati G; Petit S; Ray SM; Ham D; Capers C; Ewing H; Coffin N; McDonald LC; Jernigan J; Cardo D, Vital Signs: Epidemiology and Recent Trends in Methicillin-Resistant and in Methicillin-Susceptible Staphylococcus aureus Bloodstream Infections — United States. MWR Morb. Mortal. Wkly. Rep 2019, 68, 214–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.CDC. Antibiotic Resistance Threats in the United States. www.cdc.gov/DrugResistance/Biggest-Threats.html (accessed 07/26/2021).
- 32.Abutaleb NS; Elkashif A; Flaherty DP; Seleem MN, In Vivo Antibacterial Activity of Acetazolamide. Antimicrob. Agents Chemother 2021, 65 (4). DOI: 10.1128/AAC.01715-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Abutaleb NS; Seleem MN, Antivirulence Activity of Auranofin Against Vancomycin-Resistant Enterococci: In Vitro and In Vivo Studies. Int. J. Antimicrob. Agents 2020, 55 (3), 105828. DOI: 10.1016/j.ijantimicag.2019.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Abutaleb NS; Elhassanny AEM; Flaherty DP; Seleem MN, In Vitro and In Vivo Activities of the Carbonic Anhydrase Inhibitor, Dorzolamide, Against Vancomycin-Resistant Enterococci. PeerJ 2021, 9, e11059. DOI: 10.7717/peerj.11059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tyanova S; Temu T; Sinitcyn P; Carlson A; Hein MY; Geiger T; Mann M; Cox J, The Perseus Computational Platform for Comprehensive Analysis of (Prote)omics Data. Nat. Methods 2016, 13 (9), 731–740. [DOI] [PubMed] [Google Scholar]
- 36.Hu Y; Meng J; Shi C; Hervin K; Fratamico PM; Shi X, Characterization and Comparative Analysis of a Second Thermonuclease from Staphylococcus aureus. Microbiol. Res 2013, 168 (3), 174–182. [DOI] [PubMed] [Google Scholar]
- 37.Parrish JZ; Xue D, Cuts Can Kill: The Roles of Apoptotic Nucleases in Cell Death and Animal Development. Chromosoma 2006, 115 (2), 89–97. [DOI] [PubMed] [Google Scholar]
- 38.Kneuper H; Cao ZP; Twomey KB; Zoltner M; Jager F; Cargill JS; Chalmers J; van der Kooi-Pol MM; van Dijl JM; Ryan RP; Hunter WN; Palmer T, Heterogeneity in Ess Transcriptional Organization and Variable Contribution of the Ess/Type VII Protein Secretion System to Virulence Across Closely Related Staphylocccus aureus Strains. Mol. Microbiol 2014, 93 (5), 928–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hsia KC; Li CL; Yuan HS, Structural and Functional Insight Into Sugar-Nonspecific Nucleases in Host Defense. Curr. Opin. Struct. Biol 2005, 15 (1), 126–134. [DOI] [PubMed] [Google Scholar]
- 40.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, Comprehensive Identification of Essential Staphylococcus aureus Genes Using Transposon-Mediated Differential Hybridisation (TMDH). BMC Genomics 2009, 10, 291. DOI: 10.1186/1471-2164-10-291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li C; Tan BK; Zhao J; Guan Z, In Vivo and In Vitro Synthesis of Phosphatidylglycerol by an Escherichia coli Cardiolipin Synthase. J. Biol. Chem 2016, 291 (48), 25144–25153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yang B; Yao H; Li D; Liu Z, The Phosphatidylglycerol Phosphate Synthase PgsA Utilizes a Trifurcated Amphipathic Cavity for Catalysis at the Membrane-Cytosol Interface. Curr. Res. Struct. Biol 2021, 3, 312–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhang YM; Rock CO, Membrane Lipid Homeostasis in Bacteria. Nat. Rev. Microbiol 2008, 6 (3), 222–233. [DOI] [PubMed] [Google Scholar]
- 44.Hammer ND; Skaar EP, Molecular Mechanisms of Staphylococcus aureus Iron Acquisition. Annu. Rev. Microbiol 2011, 65, 129–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Stauff DL; Skaar EP, The Heme Sensor System of Staphylococcus aureus. Contrib. Microbiol 2009, 16, 120–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wright MH; Sieber SA, Chemical Proteomics Approaches for Identifying the Cellular Targets of Natural Products. Nat. Prod. Rep 2016, 33 (5), 681–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Schenone M; Dancik V; Wagner BK; Clemons PA, Target Identification and Mechanism of Action in Chemical Biology and Drug Discovery. Nat. Chem. Biol 2013, 9 (4), 232–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ong S; Schenone M; Margolin AA; Li XL; Do K; Doud MK; Mani DR; Kuai L; Wang X; Wood JL; Tolliday NJ; Koehler AN; Marcaurelle TR; Gould RJ; Schreiber S; Carr SA, Identifying the Proteins to Which Small-Molecule Probes and Drugs Bind in Cells. Proc. Natl. Acad. Sci. U.S.A 2009, 106 (12), 4617–4622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen X; Wang Y; Ma N; Tian J; Shao Y; Zhu B; Wong YK; Liang Z; Zou C; Wang J, Target Identification of Natural Medicine with Chemical Proteomics Approach: Probe Synthesis, Target Fishing and Protein Identification. Signal Transduct. Target Ther 2020, 5 (1), 72. DOI: 10.1038/s41392-020-0186-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lin Z; Konno M; Abad-Zapatero C; Wierenga R; Murthy MR; Ray WJ Jr.; Rossmann MG, The Structure of Rabbit Muscle Phosphoglucomutase at Intermediate Resolution. J. Biol. Chem 1986, 261 (1), 264–274. [PubMed] [Google Scholar]
- 51.Mehra-Chaudhary R; Mick J; Tanner JJ; Henzl MT; Beamer LJ, Crystal Structure of a Bacterial Phosphoglucomutase, An Enzyme Involved in the Virulence of Multiple Human Pathogens. Proteins 2011, 79 (4), 1215–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Grundling A; Schneewind O, Genes Required for Glycolipid Synthesis and Lipoteichoic Acid Anchoring in Staphylococcus aureus. J. Bacteriol 2007, 189 (6), 2521–2530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kiriukhin MY; Debabov DV; Shinabarger DL; Neuhaus FC, Biosynthesis of the Glycolipid Anchor in Lipoteichoic Acid of Staphylococcus aureus RN4220: Role of YpfP, the Diglucosyldiacylglycerol Synthase. J. Bacteriol 2001, 183 (11), 3506–3514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Xie Z; Gonzalez LE; Ferreira CR; Vorsilak A; Frabutt D; Sobreira TJP; Pugia M; Cooks RG, Multiple Reaction Monitoring Profiling (MRM-Profiling) of Lipids To Distinguish Strain-Level Differences in Microbial Resistance in Escherichia coli. Anal. Chem 2019, 91 (17), 11349–11354. [DOI] [PubMed] [Google Scholar]
- 55.Wenzel M, Do We Really Understand How Antibiotics Work? Future Microbiol. 2020, 15, 1307–1311. [DOI] [PubMed] [Google Scholar]
- 56.Grein F; Muller A; Scherer KM; Liu X; Ludwig KC; Klockner A; Strach M; Sahl HG; Kubitscheck U; Schneider T, Ca(2+)-Daptomycin Targets Cell Wall Biosynthesis by Forming a Tripartite Complex with Undecaprenyl-Coupled Intermediates and Membrane Lipids. Nat. Commun 2020, 11 (1), 1455. DOI: 10.1038/s41467-020-15257-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wenzel M; Dekker MP; Wang B; Burggraaf MJ; Bitter W; van Weering JR; Hamoen LW, A flat embedding method for transmission electron microscopy reveals an unknown mechanism of tetracycline. Commun. Biol 2021, 4. 1307–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Montgomery CP; Daniels MD; Zhao F; Spellberg B; Chong AS; Daum RS, Local Inflammation Exacerbates the Severity of Staphylococcus aureus Skin Infection. PLoS One 2013, 8 (7), e69508. DOI: 10.1371/journal.pone.0069508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kengatharan KM; De Kimpe S; Robson C; Foster SJ; Thiemermann C, Mechanism of Gram-Positive Shock: Identification of Peptidoglycan and Lipoteichoic Acid Moieties Essential in the Induction of Nitric Oxide Synthase, Shock, and Multiple Organ Failure. J. Exp. Med 1998, 188 (2), 305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kang SS; Sim JR; Yun CH; Han SH, Lipoteichoic Acids as a Major Virulence Factor Causing Inflammatory Responses via Toll-like Receptor 2. Arch. Pharm. Res 2016, 39 (11), 1519–1529. [DOI] [PubMed] [Google Scholar]
- 61.Huang BR; Tsai CF; Lin HY; Tseng WP; Huang SS; Wu CR; Lin C; Yeh WL; Lu DY, Interaction of Inflammatory and Anti-Inflammatory Responses in Microglia by Staphylococcus aureus-Derived Lipoteichoic Acid. Toxicol. Appl. Pharmacol 2013, 269 (1), 43–50. [DOI] [PubMed] [Google Scholar]
- 62.Brauweiler AM; Goleva E; Leung DYM, Staphylococcus aureus Lipoteichoic Acid Damages the Skin Barrier through an IL-1-Mediated Pathway. J. Invest. Dermatol 2019, 139 (8), 1753–1761 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fournier B; Philpott DJ, Recognition of Staphylococcus aureus by the Innate Immune System. Clin. Microbiol. Rev 2005, 18 (3), 521–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang H; Kundra S; Chojnacki M; Liu K; Fuse MA; Abouelhassan Y; Kallifidas D; Zhang P; Huang G; Jin S; Ding Y; Luesch H; Rohde KH; Dunman PM; Lemos JA; Huigens RW 3rd, A Modular Synthetic Route Involving N-Aryl-2-nitrosoaniline Intermediates Leads to a New Series of 3-Substituted Halogenated Phenazine Antibacterial Agents. J. Med. Chem 2021, 64 (11), 7275–7295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kaplan AR; Musaev DG; Wuest WM, Pyochelin Biosynthetic Metabolites Bind Iron and Promote Growth in Pseudomonads Demonstrating Siderophore-like Activity. ACS Infect. Dis 2021, 7 (3), 544–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mattingly AE; Cox KE; Smith R; Melander RJ; Ernst RK; Melander C, Screening an Established Natural Product Library Identifies Secondary Metabolites That Potentiate Conventional Antibiotics. ACS Infect. Dis 2020, 6 (10), 2629–2640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Weig AW; Barlock SL; O’Connor PM; Marciano OM; Smith R; Ernst RK; Melander RJ; Melander C, A Scaffold Hopping Strategy to Generate New Aryl-2-Amino Pyrimidine MRSA Biofilm Inhibitors. RSC Med. Chem 2021, 12 (2), 293–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Milly TA; Engler ER; Chichura KS; Buttner AR; Koirala B; Tal-Gan Y; Bertucci MA, Harnessing Multiple, Nonproteogenic Substitutions to Optimize CSP:ComD Hydrophobic Interactions in Group 1 Streptococcus pneumoniae. Chembiochem 2021, 22 (11), 1940–1947. [DOI] [PubMed] [Google Scholar]
- 69.Garcia Chavez M; Garcia A; Lee HY; Lau GW; Parker EN; Komnick KE; Hergenrother PJ, Synthesis of Fusidic Acid Derivatives Yields a Potent Antibiotic with an Improved Resistance Profile. ACS Infect. Dis 2021, 7 (2), 493–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.West KHJ; Shen W; Eisenbraun EL; Yang T; Vasquez JK; Horswill AR; Blackwell HE, Non-Native Peptides Capable of Pan-Activating the agr Quorum Sensing System across Multiple Specificity Groups of Staphylococcus epidermidis. ACS Chem. Biol 2021, 16 (6), 1070–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Boudreau MA; Ding D; Meisel JE; Janardhanan J; Spink E; Peng Z; Qian Y; Yamaguchi T; Testero SA; O’Daniel PI; Leemans E; Lastochkin E; Song W; Schroeder VA; Wolter WR; Suckow MA; Mobashery S; Chang M, Structure-Activity Relationship for the Oxadiazole Class of Antibacterials. ACS Med. Chem. Lett 2020, 11 (3), 322–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Peng Z; Nguyen TT; Song W; Anderson B; Wolter WR; Schroeder VA; Hesek D; Lee M; Mobashery S; Chang M, Selective MMP-9 Inhibitor (R)-ND-336 Alone or in Combination with Linezolid Accelerates Wound Healing in Infected Diabetic Mice. ACS Pharmacol. Transl. Sci 2021, 4 (1), 107–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lin Y; Ribaucourt A; Moazami Y; Pierce JG, Concise Synthesis and Antimicrobial Evaluation of the Guanidinium Alkaloid Batzelladine D: Development of a Stereodivergent Strategy. J. Am. Chem. Soc 2020, 142, 9850–9857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Yang H; Liu K; Jin S; Huigens RW, Design, Synthesis and Biological Evaluation of a Halogenated Phenazine-Erythromycin Conjugate Prodrug for Antibacterial Applications. Org. Biomol. Chem 2021, 19, 1483–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kaur J; Soto-Velasquez M; Ding Z; Ghanbarpour A; Lill MA; van Rijn RM; Watts VJ; Flaherty DP, Optimization of a 1,3,4-oxadiazole Series for Inhibition of Ca(2+)/Calmodulin-Stimulated Activity of Adenylyl Cyclases 1 and 8 for the Treatment of Chronic Pain. Eur. J. Med. Chem 2019, 162, 568–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Meo P; Khan N Arenoazolylamines as Antibiotic Compounds and Their Preparation. U.S. Patent WO 2018037223 March 1, 2018.
- 77.Clinical and Laboratory Standards Institute, C., Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically. 9 ed.; 2012. [Google Scholar]
- 78.Hosny Y; Abutaleb NS; Omara M; Alhashimi M; Elsebaei MM; Elzahabi HS; Seleem MN; Mayhoub AS, Modifying the Lipophilic Part of Phenylthiazole Antibiotics to Control Their Drug-Likeness. Eur. J. Med. Chem 2020, 185, 111830. DOI: 10.1016/j.ejmech.2019.111830. [DOI] [PubMed] [Google Scholar]
- 79.Elsebaei MM; Abutaleb NS; Mahgoub AA; Li D; Hagras M; Mohammad H; Seleem MN; Mayhoub AS, Phenylthiazoles With Nitrogenous Side Chain: An Approach to Overcome Molecular Obesity. Eur. J. Med. Chem 2019, 182, 111593. DOI: 10.1016/j.ejmech.2019.111593. [DOI] [PubMed] [Google Scholar]
- 80.Mohammad H; Abutaleb NS; Seleem MN, Auranofin Rapidly Eradicates Methicillin-resistant Staphylococcus aureus (MRSA) in an Infected Pressure Ulcer Mouse Model. Sci. Rep 2020, 10, 7251. DOI: 10.1038/s41598-020-64352-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mohammad H; Kyei-Baffour K; Abutaleb NS; Dai M; Seleem MN, An Aryl Isonitrile Compound with an Improved Physicochemical Profile That is Effective in Two Mouse Models of Multidrug-Resistant Staphylococcus aureus Infection. J. Glob. Antimicrob. Resist 2019, 19, 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Opoku-Temeng C; Onyedibe KI; Aryal UK; Sintim HO, Proteomic Analysis of Bacterial Response to a 4-Hydroxybenzylidene Indolinone Compound, Which Re-sensitizes Bacteria to Traditional Antibiotics. J. Proteomics 2019, 202, 103368. DOI: 10.1016/j.jprot.2019.04.018. [DOI] [PubMed] [Google Scholar]
- 83.Cox J; Mann M, MaxQuant Enables High Peptide Identification Rates, Individualized P.P.B.-Range Mass Accuracies and Proteome-Wide Protein Quantification. Nat. Biotechnol 2008, 26 (12), 1367–1372. [DOI] [PubMed] [Google Scholar]
- 84.Cox J; Hein MY; Luber CA; Paron I; Nagaraj N; Mann M, Accurate Proteome-Wide Label-Free Quantification by Delayed Normalization and Maximal Peptide Ratio Extraction, Termed MaxLFQ. Mol. Cell. Proteomics 2014, 13 (9), 2513–2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cox J; Neuhauser N; Michalski A; Scheltema RA; Olsen JV; Mann M, Andromeda: A Peptide Search Engine Integrated Into the MaxQuant Environment. J. Proteome Res 2011, 10 (4), 1794–1805. [DOI] [PubMed] [Google Scholar]
- 86.Corrigan RM; Campeotto I; Jeganathan T; Roelofs KG; Lee VT; Grundling A, Systematic Identification of Conserved Bacterial c-di-AMP Receptor Proteins. Proc. Natl. Acad. Sci. U.S.A 2013, 110 (22), 9084–9089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bligh EG; Dyer WJ, A Rapid Method of Total Lipid Extraction and Purification. Can. J. Biochem. Physiol 1959, 37 (8), 911–917. [DOI] [PubMed] [Google Scholar]
- 88.Dayal N; Opoku-Temeng C; Mohammad H; Abutaleb NS; Hernandez D; Onyedibe KI; Wang M; Zeller M; Seleem MN; Sintim HO, Inhibitors of Intracellular Gram-Positive Bacterial Growth Synthesized via Povarov-Doebner Reactions. ACS Infect. Dis 2019, 5 (11), 1820–1830. [DOI] [PubMed] [Google Scholar]
- 89.Brubaker AL; Carter SR; Kovacs EJ, Experimental Approaches to Tissue Injury and Repair in Advanced Age. Methods Mol. Biol 2015, 1343, 35–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hoffmann JP; Friedman JK; Wang Y; McLachlan JB; Sammarco MC; Morici LA; Roy CJ, In situ Treatment With Novel Microbiocide Inhibits Methicillin Resistant Staphylococcus aureus in a Murine Wound Infection Model. Front. Microbiol 2019, 10, 3106. DOI: 10.3389/fmicb.2019.03106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Vågesjö E; Öhnstedt E; Morier A; Lofton H; Huss F; Proost P; Roos S; Phillipson M, Accelerated Wound Healing in Mice by On-Site Production and Delivery of CXCL12 by Transformed Lactic Acid Bacteria. Proc. Natl. Acad. Sci. U.S.A 2018, 115 (8), 1895–1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Daina A; Michielin O; Zoete V, SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep 2017, 7, 42717. DOI: 10.1038/srep42717. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All raw LC-MS/MS data can be found in the Mass Spectrometry Interactive Virtual Environment (http://massive.ucsd.edu) with the ID: MSV000088342.
