Mehmet A. Orman's research while affiliated with University of Houston and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (64)


Fig. 1 | Impact of metabolic inhibitors on OFX tolerance. Cells were exposed to OFX (5 μg/ml) after 5 h of cell growth. Metabolic inhibitors were introduced at specified concentrations either 1 h before OFX addition (a: pre-treatment), simultaneously with OFX addition (b: co-treatment), or 1 h after OFX addition (c: posttreatment). Following a 20-h OFX treatment period, clonogenic survival assays were performed to quantify persister cells (see "Materials and methods"). In these assays, cell cultures before and after OFX treatments were washed to reduce chemical concentration, and then, serially diluted and plated on agar media for initial cell and
Fig. 2 | Synergy analysis, revealing higher synergy scores between TDZ and OFX. Survival fractions of cell cultures treated with combined drugs (OFX + metabolic inhibitor) or single drugs (OFX only or metabolic inhibitor only) under pre-, co-, and post-treatment conditions were utilized to determine the synergy scores 29 . a HSA synergy scores for all metabolic inhibitors under pre-treatment conditions
Fig. 3 | Synergy scores obtained from different models. Survival fractions of cell cultures treated with metabolic inhibitors and/or OFX under (a) pre-, (b) co-, and (c) posttreatment conditions were employed to calculate synergy scores using HAS, Bliss, Loewe, and ZIP models. N = 3.
Fig. 4 | The impact of metabolic inhibitors on cellular ATP levels. Metabolic inhibitors were administered at specified concentrations at t = 4 h (a), t = 5 h (b), or t = 6 h of cell growth (c). Culture samples were collected at the indicated time points during the treatments for ATP level quantification. ATP levels were normalized to the number of cells (#) quantified by a flow cytometer. For pairwise comparisons, a one-way ANOVA with Dunnett's post-test was applied. The threshold values for statistical significance analysis were established as *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. The mean value ± standard deviation represents
Unveiling the critical roles of cellular metabolism suppression in antibiotic tolerance
  • Article
  • Full-text available

June 2024

·

1 Read

npj Antimicrobials and Resistance

·

·

Mehmet A. Orman

Metabolic inhibitors are known to exhibit complex interactions with antibiotics in bacteria, potentially acting as antagonists by inducing cell dormancy and promoting cell survival. However, the specific synergistic or antagonistic effects of these inhibitors depend on factors like their mechanisms of action, concentrations, and treatment timings, which require further investigation. In our study, we systematically explored the synergistic interactions of various metabolic inhibitors—such as chloramphenicol (a translation inhibitor), rifampicin (a transcription inhibitor), arsenate (an ATP production inhibitor), and thioridazine (a PMF inhibitor)—in combination with ofloxacin. We conducted this investigation under pre-, co-, and post-treatment conditions, employing a wide concentration range and utilizing four distinct synergy models. Chloramphenicol, rifampicin, and arsenate consistently showed minimal synergy scores, indicating a notable antagonistic relationship with ofloxacin across all models and conditions. In contrast, thioridazine consistently demonstrated elevated synergy scores, especially in pre- and co-treatment scenarios, albeit its synergy decreased during post-treatment conditions. When multivariable linear regression analyses were used for all drugs and conditions examined, a correlation between the synergy of thioridazine and its ability to suppress cellular energy metabolism became evident, underscoring the potential utility of certain metabolic inhibitors as effective anti-persistence adjuvants.

Download
Share

Unraveling CRP/cAMP-Mediated Metabolic Regulation In Escherichia coli Persister Cells

June 2024

·

10 Reads

A substantial gap persists in our comprehension of how bacterial metabolism undergoes rewiring during the transition to a persistent state. Also, it remains unclear which metabolic mechanisms become indispensable for persister cell survival. To address these questions, we directed our efforts towards persister cells in Escherichia coli that emerge during the late stationary phase. These cells have been recognized for their exceptional resilience and are commonly believed to be in a dormant state. Our results demonstrate that the global metabolic regulator Crp/cAMP redirects the metabolism of these antibiotic-tolerant cells from anabolism to oxidative phosphorylation. Although our data indicates that persisters exhibit a reduced metabolic rate compared to rapidly growing exponential-phase cells, their survival still relies on energy metabolism. Extensive genomic-level analyses of metabolomics, proteomics, and single-gene deletions consistently emphasize the critical role of energy metabolism, specifically the tricarboxylic acid (TCA) cycle, electron transport chain (ETC), and ATP synthase, in sustaining the viability of persisters. Altogether, this study provides much-needed clarification regarding the role of energy metabolism in antibiotic tolerance and highlights the importance of using a multipronged approach at the genomic level to obtain a broader picture of the metabolic state of persister cells.


Metabolic disruption impairs ribosomal protein levels, resulting in enhanced aminoglycoside tolerance

June 2024

·

1 Read

Aminoglycoside antibiotics display broad-spectrum activity against Gram-negative and Gram-positive bacteria by targeting their ribosomes. Herein, we have demonstrated that energy metabolism plays a crucial role in aminoglycoside tolerance, as knockout strains associated with the tricarboxylic acid cycle (TCA) and the electron transport chain (ETC) exhibited increased tolerance to aminoglycosides in the mid-exponential growth phase of Escherichia coli cells. Given that aminoglycoside uptake relies on the energy-driven electrochemical potential across the cytoplasmic membrane, our initial expectation was that these genetic perturbations would decrease the proton motive force (PMF), subsequently affecting the uptake of aminoglycosides. However, our results did not corroborate this assumption. We found no consistent metabolic changes, ATP levels, cytoplasmic pH variations, or membrane potential differences in the mutant strains compared to the wild type. Additionally, intracellular concentrations of fluorophore-labeled gentamicin remained similar across all strains. To uncover the mechanism responsible for the observed tolerance in mutant strains, we employed untargeted mass spectrometry to quantify the proteins within these mutants and subsequently compared them to their wild-type counterparts. Our comprehensive analysis, which encompassed protein-protein association networks and functional enrichment, unveiled a noteworthy upregulation of proteins linked to the TCA cycle in the mutant strains during the mid-exponential growth phase, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research has the potential to uncover mechanisms behind aminoglycoside tolerance, paving the way for novel strategies to combat such cells.


Correlation between UV-induced SOS response and mutagenesis. (a) Schematic representation of the methodology for inducing intracellular mutagenesis using UV as the mutagenic agent: Mid-exponential-phase E. coli MG1655 pUA66- PrecA-gfp cells were exposed to UV for different durations and allowed to recover for 24 h. Following the recovery period, cells were plated on rifampicin (RIF)-agar plates to quantify the extent of mutagenesis. The schematic was made using BioRender software. (b) The expression profile of PrecA-gfp at different UV exposure times after a 24-h recovery period. (c) Quantification of mutant cells reported as RIF-resistant colony count per 10⁸ cell population for different UV exposure times. (d) Correlation between recA expression level and normalized RIF-resistant mutants formed during different UV exposure durations. Simple linear regression analysis was performed to fit a straight line passing through the origin (y = 0.0054 x), R² = 0.99 and P < 0.0001 (F-statistics). (e) Validation of the correlation between the UV-induced SOS response and mutagenesis levels utilizing a 96-well plate format. 288 independent cell cultures were similarly exposed to UV for varying times and then a 10 µL sample from each culture was plated on RIF agar plates after a 24-h recovery period. A frequency distribution was generated from the RIF-resistant colony data for each experimental condition and Gaussian distribution-based non-linear regression was employed to derive the probability distribution of RIF-resistant colony generation across various durations of UV exposure. For parts b, c and d: n  =  4; statistical analysis was performed using one-way ANOVA with Dunnett’s post-test, where ****P  <  0.0001; data corresponding to each time point represent mean value  ± standard deviation.
Chemical inhibition of RecA expression can significantly reduce UV-induced SOS mutagenesis. (a) Exponential-phase E. coli MG1655 ΔrecA cells were exposed to UV for 0, 2, 4, 8, 16, 24, and 32 min and then, recovered for 24 h. At the end of recovery, cells were plated on RIF agar plates to quantify RIF-resistant colonies. (b) Mid-exponential phase E. coli MG1655 pUA66- PrecA-gfp cells were treated with chemical inhibitors namely, thioridazine (TDZ; 1 mM), chlorpromazine (CPZ; 0.25 mM), hexachlorophene (HCP; 0.1 mM), perphenazine (PPZ; 1 mM), pentachlorophenol (PCP; 0.3 mM), arsenate (ARS; 1 mM), and chloramphenicol (CAM; 20 µg/mL) and exposed to UV radiation for 16 min and recovered for 24 h. After 24 h, recA expression levels were measured with a plate reader. (c) Quantification of mutant cells involved plating washed samples onto RIF agar plates after a 24-h recovery period. (d and e) Mid-exponential phase cells of Klebsiella pneumoniae (CXY 130 WT) and Acinetobacter baumannii (BAA-1605 WT) were treated with CPZ (0.3 mM and 0.1 mM, respectively) and UV for 16 min and then recovered for 24 h. RIF-resistant colonies were quantified in each case by plating the washed samples after 24 h of recovery onto RIF agar plates. n = 4. Pairwise statistical significance was performed using one-way ANOVA with Dunnett’s post-test. **, P  <  0.01; ***, P  <  0.001; ****, P  <  0.0001. Data corresponding to each column represent mean value  ± standard deviation.
Timing of targeting cellular processes is critical for the effectiveness of the drugs in reducing mutagenesis. (a) Mid-exponential phase E. coli MG1655 cells were exposed to UV radiation for 16 min and recovered for 24 h. An untreated sample was kept as a control. Samples were taken at indicated time points during recovery and plated on RIF agar medium to quantify the extent of UV-induced mutagenesis. (b) Mid-exponential phase E. coli MG1655 cells were exposed to UV for 16 min and the metabolic inhibitors, chlorpromazine (CPZ; 0.25 mM), arsenate (ARS; 1 mM), and chloramphenicol (CAM; 20 µg/mL) were added at the indicated time points (as shown in the schematics) during the recovery (0, 2, 4, and 6 h). After 24 h of recovery, mutant cells were quantified by washing the recovered samples and then plating them on the RIF agar medium. n = 4. Data corresponding to each column represent mean value  ± standard deviation.
Fluoroquinolone antibiotics induce enhanced mutagenesis compared to conventional antibiotics. (a and b) E. coli MG1655 cells were treated with ciprofloxacin (CFX; MIC: 0.02 µg/mL) and ofloxacin (OFX; MIC: 0.07 µg/mL) at indicated concentrations for 24 h, then washed and recovered for 24 h in fresh media. Antibiotic-induced SOS mutagenesis was quantified by plating recovered cells on RIF agar plates. (c) Validation of the correlation between the CFX-induced SOS response and mutagenesis levels utilizing a 96-well plate format. A total of 384 separate cell cultures underwent treatment with 1× MIC of CFX for 24 h, followed by a 24-h recovery in antibiotic-free fresh media. A parallel process was applied to the untreated control, using solvent instead of CFX. After the recovery period, a 10 µL sample from each culture was plated onto RIF agar plates. A frequency distribution was generated from the RIF-resistant colony data for the two experimental conditions and non-linear regression using a Gaussian distribution was conducted to obtain the probability distribution. (d and e) E. coli MG1655 pUA66- PrecA-gfp cells were treated with 1× MIC of conventional antibiotics, ciprofloxacin (CFX; MIC: 0.02 µg/mL), ofloxacin (OFX; MIC: 0.07 µg/mL), gentamycin (GEN; MIC: 0.5 µg/mL), ampicillin (AMP; MIC: 6 µg/mL), and fosfomycin (FOS; MIC: 0.1 µg/mL), for 24 h, and, then washed and recovered for 24 h in fresh media. After 24 h of recovery, recA expression levels were measured and antibiotic-induced SOS mutagenesis was quantified by plating recovered cells on RIF agar plates. n = 6, Pairwise statistical significance was performed using one-way ANOVA with Dunnett’s post-test. ****, P  <  0.0001. Data corresponding to each time point represent mean value  ± standard deviation.
Co-treatment with metabolic inhibitors reduced recA expression levels and antibiotic-induced mutagenesis. (a and b) E. coli MG1655 pUA66- PrecA-gfp cells were co-treated with 1× MIC of ciprofloxacin (CFX; 0.02 µg/mL) and metabolic inhibitors, chlorpromazine (CPZ; 0.25 mM), arsenate (ARS; 1 mM), and chloramphenicol (CAM; 20 µg/mL), for 24 h, followed by a 24-h recovery period in fresh media, during which the metabolic inhibitors were also introduced into the recovery cultures. After 24 h of recovery, recA expression levels were measured and antibiotic-induced SOS mutagenesis was quantified by plating recovered cells on RIF agar plates. (c) E. coli MG1655 pUA66- PrecA-gfp cells were co-treated with CFX (1× MIC) and CPZ (0.25 mM) following the experimental schematics shown on the panel. (d) Mid-exponential phase cells of Klebsiella pneumoniae (CXY 130 WT) were co-treated with 1× MIC of ciprofloxacin (CFX; 3.5 µg/mL) and CPZ (0.3 mM) for 24 h and then recovered for 24 h in fresh media with CPZ. After 24 h of recovery, antibiotic-induced SOS mutagenesis was quantified by plating recovered cells on RIF agar plates. n = 6 and 8. Pairwise statistical significance was performed using one-way ANOVA with Dunnett’s post-test and unpaired t-test. **, P  <  0.01, ****, P  <  0.0001. Data corresponding to each time point represent mean value  ± standard deviation.
Exploring the links between SOS response, mutagenesis, and resistance during the recovery period

March 2024

·

18 Reads

Antimicrobial Agents and Chemotherapy

Antimicrobial Agents and Chemotherapy

Although the mechanistic connections between SOS-induced mutagenesis and antibiotic resistance are well established, our current understanding of the impact of SOS response levels, recovery durations, and transcription/translation activities on mutagenesis remains relatively limited. In this study, when bacterial cells were exposed to mutagens like ultraviolet light for defined time intervals, a compelling connection between the rate of mutagenesis and the RecA-mediated SOS response levels became evident. Our observations also indicate that mutagenesis primarily occurs during the subsequent recovery phase following the removal of the mutagenic agent. When transcription/translation was inhibited or energy molecules were depleted at the onset of treatment or during the early recovery phase, there was a noticeable decrease in SOS response activation and mutagenesis. However, targeting these processes later in the recovery phase does not have the same effect in reducing mutagenesis, suggesting that the timing of inhibiting transcription/translation or depleting energy molecules is crucial for their efficacy in reducing mutagenesis. Active transcription, translation, and energy availability within the framework of SOS response and DNA repair mechanisms appear to be conserved attributes, supported by their consistent manifestation across diverse conditions, including the use of distinct mutagens such as fluoroquinolones and various bacterial strains.


Metabolic disruption impairs ribosomal protein levels, resulting in enhanced aminoglycoside tolerance

February 2024

Aminoglycosides, a class of antibiotics, have been in use for decades, displaying broad-spectrum activity against Gram-negative and Gram-positive bacteria. They target ribosomes and disrupt protein synthesis. Although their use declined due to newer antibiotics with lower toxicity, increasing drug resistance has renewed interest in aminoglycosides. Herein, we have demonstrated that energy metabolism plays a crucial role in aminoglycoside tolerance, as knockout strains with deleted genes associated with the tricarboxylic acid cycle (TCA) and the electron transport chain (ETC) exhibited increased tolerance to aminoglycosides in the mid-exponential growth phase of Escherichia coli cells. Our initial hypothesis posited that genetic perturbations would lead to a reduction in the proton motive force, subsequently affecting the uptake of aminoglycosides. This hypothesis is based on the prevailing notion that aminoglycoside uptake is dependent on the distinctive and energy-driven electrochemical potential across the cytoplasmic membrane. However, our results did not support this hypothesis. Despite genetic perturbations in mutant strains, we found no consistent metabolic changes, ATP levels, cytoplasmic pH variations, or membrane potential differences compared to wild-type strains. Additionally, intracellular concentrations of fluorophore-labeled gentamicin remained similar across all strains. To uncover the mechanism responsible for the observed tolerance in mutant strains, we employed untargeted mass spectrometry to quantify the proteins within these mutants and subsequently compared them to their wild-type counterparts. Our comprehensive analysis, which encompassed protein-protein association networks and functional enrichment, unveiled a noteworthy upregulation of proteins linked to the TCA cycle in the mutant strains, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research has the potential to uncover mechanisms behind aminoglycoside tolerance, paving the way for novel strategies to combat such cells.


Metabolic disruption impairs ribosomal protein levels, resulting in enhanced aminoglycoside tolerance

February 2024

·

2 Reads

Aminoglycosides, a class of antibiotics, have been in use for decades, displaying broad-spectrum activity against Gram-negative and Gram-positive bacteria. They target ribosomes and disrupt protein synthesis. Although their use declined due to newer antibiotics with lower toxicity, increasing drug resistance has renewed interest in aminoglycosides. Herein, we have demonstrated that energy metabolism plays a crucial role in aminoglycoside tolerance, as knockout strains with deleted genes associated with the tricarboxylic acid cycle (TCA) and the electron transport chain (ETC) exhibited increased tolerance to aminoglycosides in the mid-exponential growth phase of Escherichia coli cells. Our initial hypothesis posited that genetic perturbations would lead to a reduction in the proton motive force, subsequently affecting the uptake of aminoglycosides. This hypothesis is based on the prevailing notion that aminoglycoside uptake is dependent on the distinctive and energy-driven electrochemical potential across the cytoplasmic membrane. However, our results did not support this hypothesis. Despite genetic perturbations in mutant strains, we found no consistent metabolic changes, ATP levels, cytoplasmic pH variations, or membrane potential differences compared to wild-type strains. Additionally, intracellular concentrations of fluorophore-labeled gentamicin remained similar across all strains. To uncover the mechanism responsible for the observed tolerance in mutant strains, we employed untargeted mass spectrometry to quantify the proteins within these mutants and subsequently compared them to their wild-type counterparts. Our comprehensive analysis, which encompassed protein-protein association networks and functional enrichment, unveiled a noteworthy upregulation of proteins linked to the TCA cycle in the mutant strains, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research has the potential to uncover mechanisms behind aminoglycoside tolerance, paving the way for novel strategies to combat such cells.


Figure 1. Deletions in the TCA cycle and ETC genes enhance tolerance to aminoglycosides. In the
Figure 2. The observed tolerance in the mutant strains is not linked to cell growth. (A) Growth of E.
Figure 4. Deletion of genes associated with the TCA cycle and electron transport chain showed no
Metabolic disruption impairs ribosomal protein levels, resulting in enhanced aminoglycoside tolerance

December 2023

·

23 Reads

Aminoglycosides, a class of antibiotics, have been in use for decades, displaying broad-spectrum activity against Gram-negative and Gram-positive bacteria. They target ribosomes and disrupt protein synthesis. Although their use declined due to newer antibiotics with lower toxicity, increasing drug resistance has renewed interest in aminoglycosides. Herein, we have demonstrated that energy metabolism plays a crucial role in aminoglycoside tolerance, as knockout strains with deleted genes associated with the tricarboxylic acid cycle (TCA) and the electron transport chain (ETC) exhibited increased tolerance to aminoglycosides in the mid-exponential growth phase of Escherichia coli cells. Our initial hypothesis posited that genetic perturbations would lead to a reduction in the proton motive force, subsequently affecting the uptake of aminoglycosides. This hypothesis is based on the prevailing notion that aminoglycoside uptake is dependent on the distinctive and energy-driven electrochemical potential across the cytoplasmic membrane. However, our results did not support this hypothesis. Despite genetic perturbations in mutant strains, we found no consistent metabolic changes, ATP levels, cytoplasmic pH variations, or membrane potential differences compared to wild-type strains. Additionally, intracellular concentrations of fluorophore-labeled gentamicin remained similar across all strains. To uncover the mechanism responsible for the observed tolerance in mutant strains, we employed untargeted mass spectrometry to quantify the proteins within these mutants and subsequently compared them to their wild-type counterparts. Our comprehensive analysis, which encompassed protein-protein association networks and functional enrichment, unveiled a noteworthy upregulation of proteins linked to the TCA cycle in the mutant strains, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research has the potential to uncover mechanisms behind aminoglycoside tolerance, paving the way for novel strategies to combat such cells.


Systematic design of pulse dosing to eradicate persister bacteria

January 2023

·

49 Reads

·

6 Citations

A small fraction of infectious bacteria use persistence as a strategy to survive exposure to antibiotics. Periodic pulse dosing of antibiotics has long been considered a potentially effective strategy towards eradication of persisters. Recent studies have demonstrated through in vitro experiments that it is indeed feasible to achieve such effectiveness. However, systematic design of periodic pulse dosing regimens to treat persisters is currently lacking. Here we rigorously develop a methodology for the systematic design of optimal periodic pulse dosing strategies for rapid eradication of persisters. A key outcome of the theoretical analysis, on which the proposed methodology is based, is that bactericidal effectiveness of periodic pulse dosing depends mainly on the ratio of durations of the corresponding on and off parts of the pulse. Simple formulas for critical and optimal values of this ratio are derived. The proposed methodology is supported by computer simulations and in vitro experiments.


Fig. 2 Identified chemical hits reduced persister levels in E. coli MG1655 cultures. a-c Biphasic kill curves. Early stationary phase E. coli MG1655 cells, grown in LB, were treated with OFX (5 µg/ml) and the chemicals at indicated concentrations. Throughout treatment cells were plated for viable cell counts at indicated time points. Of note, the control data (OFX only) is the same in all three panels. TDZ Thioridazine; TFP Trifluoperazine; FPZ Fluphenazine; PPZ Perphenazine; CPZ Chlorpromazine; AMT Amitriptyline; HCP Hexachlorophene; PCP Pentachlorophenol; PT Potassium tellurite. n = 4. d Cell survival after quinolone and phenothiazine treatment. Cells (E. coli MG1655) treated with indicated quinolone and/or phenothiazines for 20 h were plated for viable cell counts. NOR: norfloxacin (0.8 µg/ml); MXF Moxifloxacin (0.8 µg/ml), LVX Levofloxacin (0.3 µg/ml); CIP Ciprofloxacin (0.2 µg/ml). n = 4. Statistical analysis was performed using one-way ANOVA with Dunnett's post-test, where *P < 0.05, ***P < 0.001, ****P < 0.0001. Data corresponding to each time point represent mean value ± standard deviation.
Fig. 3 Phenothiazine drugs reduced persister levels of Gram-negative bacteria. Early stationary phase cells of P. aeruginosa, K. pneumonia, and A. baumannii strains (see Supplementary Fig. 1 for growth curves) were treated
Fig. 4 Phenothiazine drugs perturb OFX-induced RecA expression during recovery. Early stationary phase E. coli MG1655 cells with P recA -gfp (grown in LB) were treated with (a) OFX or (b) OFX + TDZ for 20 h. After treatment, cells were washed and transferred to liquid LB medium for persister recovery. At designated time points during persister recovery, 10 µl cells samples were spotted on 1% agarose pads for phase contrast and fluorescent microscopy. A representative image is shown; all independent biological replicates have similar results. Green arrow: elongated cells with high GFP, blue arrow: dividing exponential phase cells with low GFP. n = 3. Scale bar: 25 µm.
Fig. 5 Phenothiazine drugs reduced energy metabolism in E. coli MG1655. a Mass spectrometry analysis of TDZ-treated and control cells. E. coli MG1655 cells grown in LB were treated with 0.25 mM TDZ in early stationary phase (t = 5 h) for 20 h. After treatment, cells were collected and analyzed by mass spectrometry. Unsupervised hierarchical clustering was performed on the metabolic data. The data was standardized across all samples for each metabolite to obtain a mean value of 0 and a standard deviation of 1. Color codes show how a data point deviates from the mean value. Each column represents a biological replicate. n = 4. b, c Pathway enrichment maps. Metabolites of the TCA cycle and glycolysis from TDZ-treated cells were compared to those of untreated cells. The circle size is proportional to the ratio of the normalized intensities of metabolites between TDZ-treated and untreated cells. Blue (P ≤ 0.05) and red (P ≤ 0.05 for dark red; 0.05 < P < 0.10 for light red) colors represent the metabolites that are significantly upregulated (red) or downregulated (blue) in the treatment group compared to the control. Gray indicates that there was no significant difference between the groups. n = 4. d Relative amounts of carbon source metabolites in the treatment and control groups obtained from mass spectrometry analysis. n = 4. e RSG staining of phenothiazine-treated cells. Early stationary phase cells (t = 5 h) were treated with phenothiazines for 20 h and then stained with RSG prior to analysis by flow cytometry. The mean fluorescence intensity of each treatment group was normalized to that of the control (untreated) group. n = 6. f ATP levels in
Fig. 6 Phenothiazine can transiently permeabilize the cell membrane by inhibiting membrane-bound metabolic proteins. a Phenothiazine treatment induced membrane permeabilization. E. coli MG1655 cells at early stationary phase (grown in LB) were treated with phenothiazine drugs at indicated concentrations. Following treatment, samples were collected and stained with PI at indicated time points for flow cytometry analysis. Live cells and ethanoltreated dead cells were used as negative and positive controls, respectively, to gate the PI negative (−) and PI positive (+) populations (see Supplementary Fig. 8b). n = 3. b Screening E. coli (K-12 BW25113) Keio Knockout Collection. Cells at early stationary phase (grown in LB) were treated with 0.25 mM TDZ for 1 h and then stained with PI for flow cytometry analysis to quantify PI (+) cell fractions (%). n = 1. c Phenothiazine-induced membrane permeabilization in E. coli MG1655 mutant strains. The mutant strains at early stationary phase were treated with 0.25 mM TDZ for 1 h and then stained with PI for flow cytometry analysis. n = 4. Statistical analysis was performed using one-way ANOVA with Dunnett's post-test. ****P < 0.0001. FSC-H: Forward scatter. Data corresponding to each time point represent mean value ± standard deviation.
Pleiotropic actions of phenothiazine drugs are detrimental to Gram-negative bacterial persister cells

December 2022

·

115 Reads

·

8 Citations

Communications Biology

Bacterial persister cells are temporarily tolerant to bactericidal antibiotics but are not necessarily dormant and may exhibit physiological activities leading to cell damage. Based on the link between fluoroquinolone-mediated SOS responses and persister cell recovery, we screened chemicals that target fluoroquinolone persisters. Metabolic inhibitors (e.g., phenothiazines) combined with ofloxacin (OFX) perturbed persister levels in metabolically active cell populations. When metabolically stimulated, intrinsically tolerant stationary phase cells also became OFX-sensitive in the presence of phenothiazines. The effects of phenothiazines on cell metabolism and physiology are highly pleiotropic: at sublethal concentrations, phenothiazines reduce cellular metabolic, transcriptional, and translational activities; impair cell repair and recovery mechanisms; transiently perturb membrane integrity; and disrupt proton motive force by dissipating the proton concentration gradient across the cell membrane. Screening a subset of mutant strains lacking membrane-bound proteins revealed the pleiotropic effects of phenothiazines potentially rely on their ability to inhibit a wide range of critical metabolic proteins. Altogether, our study further highlights the complex roles of metabolism in persister cell formation, survival and recovery, and suggests metabolic inhibitors such as phenothiazines can be selectively detrimental to persister cells.


Figure 1. Bexarotene effects on Aβ40 fibrillization kinetics. A-C, schematic illustrations of binding of bexarotene (purple) to an Aβ40 monomer (gold) (A) and an Aβ40 oligomer (B) and capping of the fibril tip (C). D, the evolution of thioflavin T (ThT) fluorescence at 488 nm in 50 μM solutions of Aβ40 in the absence and presence of bexarotene at 17 and 50 μM. Inset, the chemical structure of bexarotene. Aβ, amyloid β; Aβ40, 40-residue Aβ isoform.
Suppression of amyloid β fibril growth by drug-engineered polymorph transformation

November 2022

·

67 Reads

·

1 Citation

Journal of Biological Chemistry

Sima Mafimoghaddam

·

·

Michael B. Sherman

·

[...]

·

Fibrillization of the protein amyloid β is assumed to trigger Alzheimer’s pathology. Approaches that target amyloid plaques, however, have garnered limited clinical success and their failures may relate to the scarce understanding of the impact of potential drugs on the intertwined stages of fibrillization. Here we demonstrate that bexarotene, a T-cell lymphoma medication with anti-amyloid activity both in vitro and in vivo, suppresses amyloid fibrillization by promoting an alternative fibril structure. We employ time-resolved in situ atomic force microscopy to quantify the kinetics of growth of individual fibrils and supplement it with structure characterization by cryogenic electron microscopy. We show that fibrils with structure engineered by the drug nucleate and grow substantially slower than “normal” fibrils; remarkably, growth remains stunted even in drug-free solutions. We find that the suppression of fibril growth by bexarotene is not due to the drug binding to the fibril tips or to the peptides in the solution. Kinetic analyses attribute the slow growth of drug-enforced fibril polymorph to the distinctive dynamics of peptide chain association to their tips. As an additional benefit, the bexarotene fibrils kill primary rat hippocampal neurons less efficiently than normal fibrils. In conclusion, the suggested drug-driven polymorph transformation presents a mode of action to irreversibly suppress toxic aggregates not only in Alzheimer’s, but also potentially in myriad diverse pathologies that originate with protein condensation.


Citations (43)


... In that study, the length of the periodic break in between antibiotics impacted efficacy, with an optimal length that sensitized the biofilm to repeat treatment without allowing resistance. The optimal length of time in between pulses to rapidly eradicate persisters has been recently addressed using a methodology where the bactericidal effectiveness of periodic pulse dosing depended primarily on the ratio of durations of the corresponding on and off parts of the pulse [227]. The methodology relies on explicit formulas that make use of easily obtainable data from time-growth and time-kill experiments with a bacterial population exposed to antibiotics [227] and could be conducted for both Borrelia burgdorferi and Bartonella henselae since both have biofilm/persister forms in culture and in vitro [82][83][84]153,158,160,228,229]. ...

Reference:

Combining Double-Dose and High-Dose Pulsed Dapsone Combination Therapy for Chronic Lyme Disease/Post-Treatment Lyme Disease Syndrome and Co-Infections, Including Bartonella: A Report of 3 Cases and a Literature Review
Systematic design of pulse dosing to eradicate persister bacteria
PLOS Computational Biology

PLOS Computational Biology

... A variety of intervention strategies can be envisioned to slow the progression of diseases like Alzheimer's, Parkinson's, and HD: e.g. antibody-based therapeutics, protein-lowering strategies, and small molecule aggregation modulators (18)(19)(20)(21). Much recent effort has gone into the protein-lowering approaches, but we hypothesize that methods that target the downstream aggregation process may be a valuable complement, also given recent challenges in clinical tests of Htt lowering in patients. ...

Suppression of amyloid β fibril growth by drug-engineered polymorph transformation

Journal of Biological Chemistry

... A promising avenue for developing antibacterial therapies involves identifying a shared mechanism spanning diverse cell types, potentially found within the metabolic processes of bacterial cells. One potential target is the bacterial proton motive force (PMF), which may apply to both antibiotic-sensitive and resistant bacteria 12,13 . However, this may seem counterintuitive for tolerant cells like persisters or stationary phase cells, typically considered non-growing and dormant 14 . ...

Proton Motive Force Inhibitors Are Detrimental to Methicillin-Resistant Staphylococcus aureus Strains

Microbiology Spectrum

... A promising avenue for developing antibacterial therapies involves identifying a shared mechanism spanning diverse cell types, potentially found within the metabolic processes of bacterial cells. One potential target is the bacterial proton motive force (PMF), which may apply to both antibiotic-sensitive and resistant bacteria 12,13 . However, this may seem counterintuitive for tolerant cells like persisters or stationary phase cells, typically considered non-growing and dormant 14 . ...

Pleiotropic actions of phenothiazine drugs are detrimental to Gram-negative bacterial persister cells

Communications Biology

... guaA is essential for the growth of Mtb H37Rv, and deletion of the guaA gene resulted in Mtb lethality [25]. In E. coli, guaA is essential for the formation of persister cells, as their absence significantly enhances cell sensitivity to various antibiotics [43]. GuaA acted as an upstream reaction in the ppGpp biosynthesis pathway and may affect persistence by disrupting ppGpp regulation [44]. ...

High-Throughput Screening of a Promoter Library Reveals New Persister Mechanisms in Escherichia Coli

Microbiology Spectrum

... 22 Glycolysis was suppressed while enrichment was observed in bile acid metabolism, fatty acid metabolism, and adipogenesis, confirming altered metabolism in DTPs. 11,23 Apart from H358, in PC9, A375, and GTL-16, enrichment was also observed in gene sets, including inflammatory response, IL6-JAK-STAT3 signaling, and tumor necrosis factor-a signaling via NF-kB, interferon-g, and interferon-a response, raising the possibility that active inflammation signaling may contribute to the suppression of drug responses in persisters. Other established features of persisters, including apoptosis, ROS pathway, and EMT transition, were also observed consistently by RNA-seq. 10 To comprehensively investigate the difference of persisters derived from multiple cancer origins, we defined each signature Article ll OPEN ACCESS enrichment score as a characteristic of every DTP dataset and calculated the distance between every pair of two samples based on the average value of differences over all characteristics (Table S2; see STAR Methods). ...

A Transient Metabolic State in Melanoma Persister Cells Mediated by Chemotherapeutic Treatments

Frontiers in Molecular Biosciences

... Regarding peptidases and proteases, clpP2 emerged as the most intriguing candidate, exhibiting altered expression under treatments in both growth phases. This finding is compelling because Clp family chaperones and peptidases are closely linked to the activation of type II TA systems [47,48,51], suggesting their joint participation, which warrants further investigation in subsequent studies. Additionally, it has been demonstrated that changes in chaperone expression influence the targets of Clp protease complexes, regulating, for example, the abundance of misfolded proteins (or total proteins) in the cell, a factor associated with persister cells [18,20]. ...

lon Deletion Impairs Persister Cell Resuscitation in Escherichia coli

... We sought to determine whether activation of the MRTF pathway contributes to Vemresistance. Using CCG-257081, an MRTF-pathway inhibitor (MRTFi) acting downstream of ROCK [24,25] we tested effects of the compound on measures of Rho/MRTF signaling and cell sensitivity to Vem. Treating YUMM1.7_R and YUMMER_R with 10 µM of CCG-257081 for 24 h significantly reduced actin-stress fibers (Figure 4a-d). ...

BRAF-Inhibitor-Induced Metabolic Alterations in A375 Melanoma Cells

Metabolites

... Self-digestion or autophagy of cells of a subpopulation, including the intracellular degradation of RNA, protein and lipids, is an important survival strategy for a bacterial population to combat unfavored conditions through recycling of energy harvesting molecules or acceleration of dormancy. 32 In E. coli stationary phase populations, a correlation between self-digestion and persister formation was proposed. 33 In our work, we used an early log phase population to generate persisters where starvation is unlikely to occur. ...

Cellular Self-Digestion and Persistence in Bacteria

Microorganisms

... Ethanol (70% vol/vol)treated cells (i.e., dead cells) were used as a positive control (PI-positive cells), and PI-stained live cells (PI-negative cells) served as a negative control (Fig. S2). Forward and side scatter parameters obtained from the untreated live cells were used to gate the cell populations on the flow cytometry diagram (80). For the fluorescence measurement, the cells were excited at a 561-nm wavelength and detected with a 615/20-nm bandpass filter. ...

Monitoring Persister Resuscitation with Flow Cytometry
  • Citing Chapter
  • September 2021

Methods in molecular biology (Clifton, N.J.)