Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Am J Pharm Educ ; : 101249, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39032698

ABSTRACT

OBJECTIVE: Professional pharmacy fraternal organizations are among the most common student organizations in schools and colleges of pharmacy, present on 98% of campuses. However, sparse literature explores the educational value these organizations offer. The purpose of this review is to explore the alignment of national requirements of two major pharmacy fraternities with co-curricular learning objectives. METHODS: All four fraternal pharmacy organizations recognized by the Professional Fraternal Association were invited to collaborate on this project. Ultimately, two fraternities participated by gathering national office requirements for reports and activities for collegiate chapters. Qualitative review of fraternity requirements was conducted via manifest content analysis by two independent reviewers, and items were coded to the relevant co-curricular domain(s) from the Accreditation Council for Pharmacy Education Standards 2016; disagreements were resolved by a third author. RESULTS: A total of 50 fraternity requirements were identified and mapped to one or more co-curricular domains, for a total of 63 codings. All six co-curricular domains were coded at least once. The most common codings were professionalism and leadership. Significant overlap existed in requirements that encompassed professionalism plus cultural sensitivity, professionalism plus self-awareness, and self-awareness plus leadership. Some activities benefited the school or community, while others solely contributed to individual member professional development. CONCLUSION: Professional pharmacy fraternities provide ample co-curricular learning opportunities among the breadth of affective domains. The mission and vision of these organizations foster affective domain skill development in both school- and community-facing events as well as dedicated individual professional development.

2.
ACS Infect Dis ; 10(5): 1612-1623, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38597503

ABSTRACT

Fusobacterium nucleatum, a pathobiont inhabiting the oral cavity, contributes to opportunistic diseases, such as periodontal diseases and gastrointestinal cancers, which involve microbiota imbalance. Broad-spectrum antimicrobial agents, while effective against F. nucleatum infections, can exacerbate dysbiosis. This necessitates the discovery of more targeted narrow-spectrum antimicrobial agents. We therefore investigated the potential for the fusobacterial enoyl-ACP reductase II (ENR II) isoenzyme FnFabK (C4N14_ 04250) as a narrow-spectrum drug target. ENRs catalyze the rate-limiting step in the bacterial fatty acid synthesis pathway. Bioinformatics revealed that of the four distinct bacterial ENR isoforms, F. nucleatum specifically encodes FnFabK. Genetic studies revealed that fabK was indispensable for F. nucleatum growth, as the gene could not be deleted, and silencing of its mRNA inhibited growth under the test conditions. Remarkably, exogenous fatty acids failed to rescue growth inhibition caused by the silencing of fabK. Screening of synthetic phenylimidazole analogues of a known FabK inhibitor identified an inhibitor (i.e., 681) of FnFabK enzymatic activity and F. nucleatum growth, with an IC50 of 2.1 µM (1.0 µg/mL) and a MIC of 0.4 µg/mL, respectively. Exogenous fatty acids did not attenuate the activity of 681 against F. nucleatum. Furthermore, FnFabK was confirmed as the intracellular target of 681 based on the overexpression of FnFabK shifting MICs and 681-resistant mutants having amino acid substitutions in FnFabK or mutations in other genetic loci affecting fatty acid biosynthesis. 681 had minimal activity against a range of commensal flora, and it was less active against streptococci in physiologic fatty acids. Taken together, FnFabK is an essential enzyme that is amenable to drug targeting for the discovery and development of narrow-spectrum antimicrobial agents.


Subject(s)
Anti-Bacterial Agents , Fusobacterium nucleatum , Fusobacterium nucleatum/enzymology , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Fatty Acids/chemistry , Fusobacterium Infections/microbiology , Fusobacterium Infections/drug therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
3.
Biochem Biophys Res Commun ; 705: 149740, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38458032

ABSTRACT

Clostridioides difficile, a gram-positive anaerobic bacterium, is one of the most frequent causes of nosocomial infections. C. difficile infection (CDI) results in almost a half a million infections and approximately 30,000 deaths in the U.S. each year. Broad-spectrum antibacterial use is a strong risk factor for development of recurring CDI. There is a critical need for narrow-spectrum antibacterials with activity limited to C. difficile. The C. difficile enoyl-acyl carrier protein (ACP) reductase II enzyme (CdFabK), an essential and rate-limiting enzyme in the organism's fatty acid biosynthesis pathway (FAS-2), is an attractive target for narrow-spectrum CDI therapeutics as it is not present in many of the non-pathogenic gut organisms. We have previously characterized inhibitors of the CdFabK enzyme with narrow-spectrum anti-difficile activity and favorable in vivo efficacy, ADME, and low dysbiosis. To expand our knowledge of the structural requirements for CdFabK inhibition, we seek to identify new inhibitors with novel chemical scaffolds. Herein we present the optimization of a thermo-FMN biophysical assay based on the principles of differential scanning fluorimetry, or thermal shift, which leverages the fluorescence signal of the FabK enzyme's FMN prosthetic group. The optimized assay was validated by pilot testing a 10K diversity-based chemical library and novel scaffold hit compounds were identified and biochemically characterized. Additionally, we show that the thermo-FMN assay can be used to determine the thermodynamic dissociation constant, Kd, of CdFabK inhibitors.


Subject(s)
Clostridioides difficile , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Clostridioides difficile/metabolism , Base Composition , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
4.
Antimicrob Agents Chemother ; 68(3): e0122223, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38265216

ABSTRACT

Clostridioides difficile infection (CDI) is a leading cause of hospital-acquired diarrhea, which often stems from disruption of the gut microbiota by broad-spectrum antibiotics. The increasing prevalence of antibiotic-resistant C. difficile strains, combined with disappointing clinical trial results for recent antibiotic candidates, underscores the urgent need for novel CDI antibiotics. To this end, we investigated C. difficile enoyl ACP reductase (CdFabK), a crucial enzyme in de novo fatty acid synthesis, as a drug target for microbiome-sparing antibiotics. To test this concept, we evaluated the efficacy and in vivo spectrum of activity of the phenylimidazole analog 296, which is validated to inhibit intracellular CdFabK. Against major CDI-associated ribotypes 296 had an Minimum inhibitory concentration (MIC90) of 2 µg/mL, which was comparable to vancomycin (1 µg/mL), a standard of care antibiotic. In addition, 296 achieved high colonic concentrations and displayed dosed-dependent efficacy in mice with colitis CDI. Mice that were given 296 retained colonization resistance to C. difficile and had microbiomes that resembled the untreated mice. Conversely, both vancomycin and fidaxomicin induced significant changes to mice microbiomes, in a manner consistent with prior reports. CdFabK, therefore, represents a potential target for microbiome-sparing CDI antibiotics, with phenylimidazoles providing a good chemical starting point for designing such agents.


Subject(s)
Clostridioides difficile , Clostridium Infections , Animals , Mice , Vancomycin/pharmacology , Oxidoreductases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Fidaxomicin/pharmacology , Clostridium Infections/drug therapy
5.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37790427

ABSTRACT

Clostridioides difficile infection (CDI) is a leading cause of hospital-acquired diarrhea, which often stem from disruption of the gut microbiota by broad-spectrum antibiotics. The increasing prevalence of antibiotic-resistant C. difficile strains, combined with disappointing clinical trials results for recent antibiotic candidates, underscore the urgent need for novel CDI antibiotics. To this end, we investigated C. difficile enoyl ACP reductase (CdFabK), a crucial enzyme in de novo fatty acid synthesis, as a drug target for microbiome-sparing antibiotics. To test this concept, we evaluated the efficacy and in vivo spectrum of activity of the phenylimidazole analog 296, which is validated to inhibit intracellular CdFabK. Against major CDI-associated ribotypes 296 had an MIC90 of 2 µg/ml, which was comparable to vancomycin (1 µg/ml), a standard of care antibiotic. In addition, 296 achieved high colonic concentrations and displayed dosed-dependent efficacy in mice with colitis CDI. Mice that were given 296 retained colonization resistance to C. difficile and had microbiomes that resembled the untreated mice. Conversely, both vancomycin and fidaxomicin induced significant changes to mice microbiomes, in a manner consistent with prior reports. CdFabK therefore represents a potential target for microbiome-sparing CDI antibiotics, with phenylimidazoles providing a good chemical starting point for designing such agents.

6.
Nat Commun ; 14(1): 4130, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438331

ABSTRACT

Severe outbreaks and deaths have been linked to the emergence and global spread of fluoroquinolone-resistant Clostridioides difficile over the past two decades. At the same time, metronidazole, a nitro-containing antibiotic, has shown decreasing clinical efficacy in treating C. difficile infection (CDI). Most metronidazole-resistant C. difficile exhibit an unusual resistance phenotype that can only be detected in susceptibility tests using molecularly intact heme. Here, we describe the mechanism underlying this trait. We find that most metronidazole-resistant C. difficile strains carry a T-to-G mutation (which we term PnimBG) in the promoter of gene nimB, resulting in constitutive transcription. Silencing or deleting nimB eliminates metronidazole resistance. NimB is related to Nim proteins that are known to confer resistance to nitroimidazoles. We show that NimB is a heme-dependent flavin enzyme that degrades nitroimidazoles to amines lacking antimicrobial activity. Furthermore, occurrence of the PnimBG mutation is associated with a Thr82Ile substitution in DNA gyrase that confers fluoroquinolone resistance in epidemic strains. Our findings suggest that the pandemic of fluoroquinolone-resistant C. difficile occurring over the past few decades has also been characterized by widespread resistance to metronidazole.


Subject(s)
Clostridioides difficile , Nitroimidazoles , Metronidazole/pharmacology , Clostridioides difficile/genetics , Fluoroquinolones/pharmacology , Nitroimidazoles/pharmacology , Clostridioides , Heme , Pandemics
7.
Bioorg Med Chem ; 88-89: 117330, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37224699

ABSTRACT

Previously, 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-(pyridin-2-ylthio)thiazol-2-yl)urea bearing a p-bromine substitution was shown to possess selective inhibitory activity against the Clostridioides difficile enoyl-acyl carrier protein (ACP) reductase II enzyme, FabK. Inhibition of CdFabK by this compound translated to promising antibacterial activity in the low micromolar range. In these studies, we sought to expand our knowledge of the SAR of the phenylimidazole CdFabK inhibitor series while improving the potency of the compounds. Three main series of compounds were synthesized and evaluated based on: 1) pyridine head group modifications including the replacement with a benzothiazole moiety, 2) linker explorations, and 3) phenylimidazole tail group modifications. Overall, improvement in the CdFabK inhibition was achieved, while maintaining the whole cell antibacterial activity. Specifically, compounds 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-((3-(trifluoromethyl)pyridin-2-yl)thio)thiazol-2-yl)urea, 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(6-(trifluoromethyl)benzo[d]thiazol-2-yl)urea, and 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(6-chlorobenzo[d]thiazol-2-yl)urea showed CdFabK inhibition (IC50 = 0.10 to 0.24 µM), a 5 to 10-fold improvement in biochemical activity relative to 1-((4-(4-bromophenyl)-1H-imidazol-2-yl)methyl)-3-(5-(pyridin-2-ylthio)thiazol-2-yl)urea, with anti-C. difficile activity ranging from 1.56 to 6.25 µg/mL. Detailed analysis of the expanded SAR, supported by computational analysis, is presented.


Subject(s)
Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Urea , Urea/pharmacology , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship
8.
Angew Chem Int Ed Engl ; 61(31): e202203225, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35594368

ABSTRACT

A short total synthesis of tunicamycin V (1), a non-selective phosphotransferase inhibitor, is achieved via a Büchner-Curtius-Schlotterbeck type reaction. Tunicamycin V can be synthesized in 15 chemical steps from D-galactal with 21 % overall yield. The established synthetic scheme is operationally very simple and flexible to introduce building blocks of interest. The inhibitory activity of one of the designed analogues 28 against human dolichyl-phosphate N-acetylglucosaminephosphotransferase 1 (DPAGT1) is 12.5 times greater than 1. While tunicamycins are cytotoxic molecules with a low selectivity, the novel analogue 28 displays selective cytostatic activity against breast cancer cell lines including a triple-negative breast cancer.


Subject(s)
Antineoplastic Agents , Cytostatic Agents , Antineoplastic Agents/pharmacology , Humans , Tunicamycin/chemistry , Tunicamycin/pharmacology
9.
ACS Infect Dis ; 7(8): 2522-2535, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34260210

ABSTRACT

Invasive and superficial infections caused by the Candida species result in significant global morbidity and mortality. As the pathogenicity of these organisms is intimately intertwined with host immune response, therapies to target both the fungus and host inflammation may be warranted. Structural similarities exist between established inhibitors of the NLRP3 inflammasome and those of fungal acetohydroxyacid synthase (AHAS). Therefore, we leveraged this information to conduct an in silico molecular docking screen to find novel polypharmacologic inhibitors of these targets that resulted in the identification of 12 candidate molecules. Of these, compound 10 significantly attenuated activation of the NLPR3 inflammasome by LPS + ATP, while also demonstrating growth inhibitory activity against C. albicans that was alleviated in the presence of exogenous branched chain amino acids, consistent with targeting of fungal AHAS. SAR studies delineated an essential molecular scaffold required for dual activity. Ultimately, 10 and its analog 10a resulted in IC50 (IL-1ß release) and MIC50 (fungal growth) values with low µM potency against several Candida species. Collectively, this work demonstrates promising potential of dual-target approaches for improved management of fungal infections.


Subject(s)
Acetolactate Synthase , Inflammasomes , Antifungal Agents/pharmacology , Candida albicans , Molecular Docking Simulation
10.
ACS Infect Dis ; 7(5): 1044-1058, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33471519

ABSTRACT

The successful treatment of Helicobacter pylori infections is becoming increasingly difficult due to the rise of resistance against current broad spectrum triple therapy regimens. In the search for narrow-spectrum agents against H. pylori, a high-throughput screen identified two structurally related thienopyrimidine compounds that selectively inhibited H. pylori over commensal members of the gut microbiota. To develop the structure-activity relationship (SAR) of the thienopyrimidines against H. pylori, this study employed four series of modifications in which systematic substitution to the thienopyrimidine core was explored and ultimately side-chain elements optimized from the two original hits were merged into lead compounds. During the development of this series, the mode of action studies identified H. pylori's respiratory complex I subunit NuoD as the target for lead thienopyrimidines. As this enzyme complex is uniquely essential for ATP synthesis in H. pylori, a homology model of the H. pylori NuoB-NuoD binding interface was generated to help rationalize the SAR and guide further development of the series. From these studies, lead compounds emerged with increased potency against H. pylori, improved safety indices, and a good overall pharmacokinetic profile with the exception of high protein binding and poor solubility. Although lead compounds in the series demonstrated efficacy in an ex vivo infection model, the compounds had no efficacy in a mouse model of H. pylori infection. Additional optimization of pharmacological properties of the series to increase solubility and free-drug levels at the sequestered sites of H. pylori infection would potentially result in a gain of in vivo efficacy. The thienopyrimidine series developed in this study demonstrates that NuoB-NuoD of the respiratory complex I can be targeted for development of novel narrow spectrum agents against H. pylori and that thienopyrimines can serve as the basis for future advancement of these studies.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Animals , Anti-Bacterial Agents/pharmacology , Electron Transport Complex I , Helicobacter Infections/drug therapy , Mice , Pyrimidines
11.
Phytochemistry ; 181: 112545, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33217722

ABSTRACT

Alarming rate of resistance to the existing antibiotics exhibits the importance of developing new antibiotic molecules from relatively under explored sources as well as implementing alternative approaches like antibiotic adjuvants. Six previously undescribed fungal polyketides, kaneoheoic acids A-F (1-6) were isolated from a fungal strain Fusarium sp. FM701 which was collected from a muddy sample of Hawaiian beach. The structures of these six compounds were elucidated by spectroscopic interpretation, including HRESIMS and NMR, and electronic circular dichroism (ECD) analysis. All six compounds that were inactive when tested alone showed significant antibacterial activity against Staphylococcus aureus and Bacillus subtilis, in the range of 10-80 µg/mL when assayed in combination with either chloramphenicol (half of the MIC, 1 µg/mL), an FDA approved antibiotic or disulfiram (6 µg/mL), an established antibiotic adjuvant that augmented the activity of antibiotics.


Subject(s)
Fusarium , Polyketides , Anti-Bacterial Agents/pharmacology , Fungi , Hawaii , Microbial Sensitivity Tests , Polyketides/pharmacology
12.
J Med Chem ; 63(19): 10855-10878, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32886511

ABSTRACT

Capuramycin displays a narrow spectrum of antibacterial activity by targeting bacterial translocase I (MraY). In our program of development of new N-acetylglucosaminephosphotransferase1 (DPAGT1) inhibitors, we have identified that a capuramycin phenoxypiperidinylbenzylamide analogue (CPPB) inhibits DPAGT1 enzyme with an IC50 value of 200 nM. Despite a strong DPAGT1 inhibitory activity, CPPB does not show cytotoxicity against normal cells and a series of cancer cell lines. However, CPPB inhibits migrations of several solid cancers including pancreatic cancers that require high DPAGT1 expression in order for tumor progression. DPAGT1 inhibition by CPPB leads to a reduced expression level of Snail but does not reduce E-cadherin expression level at the IC50 (DPAGT1) concentration. CPPB displays a strong synergistic effect with paclitaxel against growth-inhibitory action of a patient-derived pancreatic adenocarcinoma, PD002: paclitaxel (IC50: 1.25 µM) inhibits growth of PD002 at 0.0024-0.16 µM in combination with 0.10-2.0 µM CPPB (IC50: 35 µM).


Subject(s)
Aminoglycosides/pharmacology , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Neoplasms/pathology , Aminoglycosides/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/chemistry , Humans , Paclitaxel/pharmacology , Snail Family Transcription Factors/antagonists & inhibitors , Structure-Activity Relationship
13.
Article in English | MEDLINE | ID: mdl-31712208

ABSTRACT

Repurposing of currently approved medications is an attractive option for the development of novel treatment strategies against physiological and infectious diseases. The antidiabetic sulfonylurea glyburide has demonstrated off-target capacity to inhibit activation of the NLRP3 inflammasome in a variety of disease models, including vaginal candidiasis, caused primarily by the fungal pathogen Candida albicans Therefore, we sought to determine which of the currently approved sulfonylurea drugs prevent the release of interleukin 1ß (IL-1ß), a major inflammasome effector, during C. albicans challenge of the human macrophage-like THP1 cell line. Findings revealed that the second-generation antidiabetics (glyburide, glisoxepide, gliquidone, and glimepiride), which exhibit greater antidiabetic efficacy than prior iterations, demonstrated anti-inflammatory effects with various degrees of potency as determined by calculation of 50% inhibitory concentrations (IC50s). These same compounds were also effective in reducing IL-1ß release during noninfectious inflammasome activation (e.g., induced by lipopolysaccharide [LPS] plus ATP), suggesting that their anti-inflammatory activity is not specific to C. albicans challenge. Moreover, treatment with sulfonylurea drugs did not impact C. albicans growth and filamentation or THP1 viability. Finally, the use of ECE1 and Candidalysin deletion mutants, along with isogenic NLRP3-/- cells, demonstrated that both Candidalysin and NLRP3 are required for IL-1ß secretion, further confirming that sulfonylureas suppress inflammasome signaling. Moreover, challenge of THP1 cells with synthetic Candidalysin peptide demonstrated that this toxin is sufficient to activate the inflammasome. Treatment with the experimental inflammasome inhibitor MCC950 led to similar blockade of IL-1ß release, suggesting that Candidalysin-mediated inflammasome activation can be inhibited independently of potassium efflux. Together, these results demonstrate that the second-generation antidiabetic sulfonylureas retain anti-inflammatory activity and may be considered for repurposing against immunopathological diseases, including vaginal candidiasis.


Subject(s)
Candida albicans/drug effects , Candidiasis, Vulvovaginal/drug therapy , Fungal Proteins/genetics , Hypoglycemic Agents/pharmacology , Inflammasomes/antagonists & inhibitors , Interleukin-1beta/metabolism , Sulfonylurea Compounds/pharmacology , Candida albicans/genetics , Candida albicans/growth & development , Candidiasis, Vulvovaginal/microbiology , Cell Line , Female , Fungal Proteins/metabolism , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Indenes , Inflammasomes/genetics , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sequence Deletion , Signal Transduction/drug effects , Sulfonamides , Sulfones/pharmacology
14.
J Antimicrob Chemother ; 75(4): 859-867, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31873741

ABSTRACT

OBJECTIVES: To describe, for the first time (to the best of our knowledge), the genetic mechanisms of vancomycin resistance in clinical isolates of Clostridioides difficile ribotype 027. METHODS: Clinical isolates and laboratory mutants were analysed: genomically to identify resistance mutations; by transcriptional analysis of vanGCd, the vancomycin resistance operon encoding lipid II d-alanine-d-serine that is less bound by vancomycin than native lipid II d-alanine-d-alanine; by imaging of vancomycin binding to cell walls; and for changes in vancomycin bactericidal activity and autolysis. RESULTS: Vancomycin-resistant laboratory mutants and clinical isolates acquired mutations to the vanSR two-component system that regulates vanGCd. The substitutions impaired VanSR's function, resulting in constitutive transcription of vanGCd. Resistance was reversed by silencing vanG, encoding d-alanine-d-serine ligase in the vanGCd operon. In resistant cells, vancomycin was less bound to the cell wall septum, the site where vancomycin interacts with lipid II. Vancomycin's bactericidal activity was reduced against clinical isolates and laboratory mutants (64 and ≥1024 mg/L, respectively) compared with WT strains (4 mg/L). Truncation of the potassium transporter TrkA occurred in laboratory mutants, which were refractory to autolysis, accounting for their survival in high drug concentrations. CONCLUSIONS: Ribotype 027 evolved first-step resistance to vancomycin by constitutively expressing vanGCd, which is otherwise silent. Experimental evolutions and bactericidal assays show that ribotype 027 can acquire mutations to drastically enhance its tolerance to vancomycin. Thus, further epidemiological studies are warranted to examine the extent to which vancomycin resistance impacts clinical outcomes and the potential for these strains to evolve higher-level resistance, which would be devastating.


Subject(s)
Clostridioides , Vancomycin Resistance , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Microbial Sensitivity Tests , Operon , Vancomycin/pharmacology
15.
Biochem Biophys Res Commun ; 516(2): 333-338, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31204053

ABSTRACT

Herein we report the first structure of topoisomerase I determined from the gram-positive bacterium, S. mutans. Bacterial topoisomerase I is an ATP-independent type 1A topoisomerase that uses the inherent torsional strain within hyper-negatively supercoiled DNA as an energy source for its critical function of DNA relaxation. Interest in the enzyme has gained momentum as it has proven to be essential in various bacterial organisms. In order to aid in further biochemical characterization, the apo 65-kDa amino-terminal fragment of DNA topoisomerase I from the gram-positive model organism Streptococcus mutans was crystalized and a three-dimensional structure was determined to 2.06 Šresolution via x-ray crystallography. The overall structure illustrates the four classic major domains that create the traditional topoisomerase I "lock" formation comprised of a sizable toroidal aperture atop what is considered to be a highly dynamic body. A catalytic tyrosine residue resides at the interface between two domains and is known to form a 5' phosphotyrosine DNA-enzyme intermediate during transient single-stranded cleavage required for enzymatic relaxation of hyper negative DNA supercoils. Surrounding the catalytic tyrosine residue is the remainder of the highly conserved active site. Within 5 Šfrom the catalytic center, only one dissimilar residue is observed between topoisomerase I from S. mutans and the gram-negative model organism E. coli. Immediately adjacent to the conserved active site, however, S. mutans topoisomerase I displays a somewhat unique nine residue loop extension not present in any bacterial topoisomerase I structures previously determined other than that of an extremophile.


Subject(s)
DNA Topoisomerases, Type I/chemistry , Streptococcus mutans/enzymology , Amino Acid Sequence , Crystallization , Crystallography, X-Ray , Models, Molecular
16.
Article in English | MEDLINE | ID: mdl-31209003

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) strains that are resistant to all forms of penicillin have become an increasingly common and urgent problem threatening human health. They are responsible for a wide variety of infectious diseases ranging from minor skin abscesses to life-threatening severe infections. The vra operon that is conserved among S. aureus strains encodes a three-component signal transduction system (vraTSR) that is responsible for sensing and responding to cell wall stress. We developed a novel and multifaceted assay to identify compounds that potentiate the activity of oxacillin, essentially restoring efficacy of oxacillin against MRSA, and performed high-throughput screening (HTS) to identify oxacillin potentiators. HTS of 13,840 small-molecule compounds from an antimicrobial-focused Life Chemicals library, using the MRSA cell-based assay, identified three different inhibitor scaffolds. Checkerboard assays for synergy with oxacillin, reverse transcriptase PCR (RT-PCR) assays against vraR expression, and direct confirmation of interaction with VraS by surface plasmon resonance (SPR) further verified them to be viable hit compounds. A subsequent structure-activity relationship (SAR) study of the best scaffold with diverse analogs was utilized to improve potency and provides a strong foundation for further development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Oxacillin/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Histidine Kinase/genetics , Histidine Kinase/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Structure-Activity Relationship
17.
ACS Chem Biol ; 14(7): 1528-1535, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31184849

ABSTRACT

Clostridioides difficile infection (CDI) is a leading cause of significant morbidity, mortality, and healthcare-related costs in the United States. After standard therapy, recurrence rates remain high, and multiple recurrences are not uncommon. Causes include treatments employing broad-spectrum agents that disrupt the normal host microbiota, as well as treatment-resistant spore formation by C. difficile. Thus, novel druggable anti-C. difficile targets that promote narrow-spectrum eradication and inhibition of sporulation are desired. As a critical rate-limiting step within the FAS-II bacterial fatty acid synthesis pathway, which supplies precursory component phospholipids found in bacterial cytoplasmic and spore-mediated membranes, enoyl-acyl carrier protein (ACP) reductase II (FabK) represents such a target. FabK is essential in C. difficile (CdFabK) and is structurally and mechanistically distinct from other isozymes found in gut microbiota species, making CdFabK an attractive narrow-spectrum target. We report here the kinetic evaluation of CdFabK, the biochemical activity of a series of phenylimidazole analogues, and microbiological data suggesting these compounds' selective antibacterial activity against C. difficile versus several other prominent gut organisms. The compounds display promising, selective, low micromolar CdFabK inhibitory activity without significantly affecting the growth of other gut organisms, and the series prototype (1b) is shown to be competitive for the CdFabK cofactor and uncompetitive for the substrate. A series analogue (1g) shows maintained inhibitory activity while also possessing increased solubility. These findings represent the basis for future drug discovery efforts by characterizing the CdFabK enzyme while demonstrating its druggability and potential role as a narrow-spectrum antidifficile target.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Clostridioides difficile/chemistry , Clostridioides difficile/metabolism , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
18.
mSphere ; 4(1)2019 02 06.
Article in English | MEDLINE | ID: mdl-30728284

ABSTRACT

Calcium is a critically important secondary messenger of intracellular signal transduction in eukaryotes but must be maintained at low levels in the cytoplasm of resting cells to avoid toxicity. This is achieved by several pumps that actively transport excess cytoplasmic Ca2+ out of the cell across the plasma membrane and into other intracellular compartments. In fungi, the vacuole serves as the major storage site for excess Ca2+, with two systems actively transporting cytoplasmic calcium ions into the vacuole. The H+/Ca2+ exchanger, Vcx1p, harnesses the proton-motive force across the vacuolar membrane (generated by the V-ATPase) to drive Ca2+ transport, while the P-type ATPase Pmc1p uses ATP hydrolysis to translocate Ca2+ into the vacuole. Ca2+-dependent signaling is required for the prevalent human fungal pathogen Candida albicans to endure exposure to the azole antifungals and to cause disease within the mammalian host. The purpose of this study was to determine if the Pmc1p or Vcx1p Ca2+ pumps are required for C. albicans pathogenicity and if these pumps impact antifungal resistance. Our results indicate that Pmc1p is required by C. albicans to transition from yeast to hyphal growth, to form biofilms in vitro, and to cause disease in a mouse model of disseminated infection. Moreover, loss of Pmc1p function appears to enhance C. albicans azole tolerance in a temperature-dependent manner.IMPORTANCE Maintenance of Ca2+ homeostasis is important for fungal cells to respond to a multitude of stresses, as well as antifungal treatment, and for virulence in animal models. Here, we demonstrate that a P-type ATPase, Pmc1p, is required for Candidaalbicans to respond to a variety of stresses, affects azole susceptibility, and is required to sustain tissue invasive hyphal growth and to cause disease in a mouse model of disseminated infection. Defining the mechanisms responsible for maintaining proper Ca2+ homeostasis in this important human pathogen can ultimately provide opportunities to devise new chemotherapeutic interventions that dysregulate intracellular signaling and induce Ca2+ toxicity.


Subject(s)
Candida albicans/genetics , Candida albicans/pathogenicity , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Antifungal Agents/pharmacology , Azoles/pharmacology , Biofilms/growth & development , Candida albicans/drug effects , Candidiasis, Invasive/microbiology , Drug Resistance, Fungal , Fungal Proteins , Hyphae/growth & development , Mice , Mice, Inbred BALB C , Plasma Membrane Calcium-Transporting ATPases/genetics , Temperature , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Virulence
19.
ACS Infect Dis ; 5(2): 208-217, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30501172

ABSTRACT

Clostridium difficile infection (CDI) is an antibiotic-induced microbiota shift disease of the large bowel. While there is a need for narrow-spectrum CDI antibiotics, it is unclear which cellular proteins are appropriate drug targets to specifically inhibit C. difficile. We evaluated the enoyl-acyl carrier protein (ACP) reductase II (FabK), which catalyzes the final step of bacterial fatty acid biosynthesis. Bioinformatics showed that C. difficile uses FabK as its sole enoyl-ACP reductase, unlike several major microbiota species. The essentiality of fabK for C. difficile growth was confirmed by failure to delete this gene using ClosTron mutagenesis and by growth inhibition upon gene silencing with CRISPR interference antisense to fabK transcription or by blocking protein translation. Inhibition of C. difficile's FASII pathway could not be circumvented by supply of exogenous fatty acids, either during fabK's gene silencing or upon inhibition of the enzyme with a phenylimidazole-derived inhibitor (1). The inability of fatty acids to bypass FASII inhibition is likely due to the function of the transcriptional repressor FapR. Inhibition of FabK also inhibited spore formation, reflecting the enzyme's role in de novo fatty acid biosynthesis for the formation of spore membrane lipids. Compound 1 did not inhibit growth of key microbiota species. These findings suggest that C. difficile FabK is a druggable target for discovering narrow-spectrum anti- C. difficile drugs that treat CDI but avoid collateral damage to the gut microbiota.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Biosynthetic Pathways , CRISPR-Cas Systems , Clostridioides difficile/enzymology , Clostridioides difficile/genetics , Crystallography, X-Ray , DNA, Antisense , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Fatty Acids/biosynthesis , Gene Silencing
20.
Methods Enzymol ; 610: 265-309, 2018.
Article in English | MEDLINE | ID: mdl-30390802

ABSTRACT

High-throughput screening assays have become nearly ubiquitous in the search for small compounds or peptides that can modulate biological processes for therapeutic purposes. While many assays have become quite robust, with well-established protocols, the subsequent steps of validating the hits and choosing the best ones to take forward into leads for further chemical development are less established. In this chapter, we describe a variety of approaches, including chemical assessment, the use of various computational approaches, a variety of counter-screens, and "orthogonal" biophysical assays using nuclear magnetic resonance, surface plasmon resonance, isothermal titration calorimetry or thermal shift assays as methods for validating and assessing the quality of hits.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Calorimetry/methods , Computer-Aided Design , Humans , Magnetic Resonance Spectroscopy/methods , Structure-Activity Relationship , Surface Plasmon Resonance/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...