1
|
Garcia-Santamarina S, Kuhn M, Devendran S, Maier L, Driessen M, Mateus A, Mastrorilli E, Brochado AR, Savitski MM, Patil KR, Zimmermann M, Bork P, Typas A. Emergence of community behaviors in the gut microbiota upon drug treatment. Cell 2024; 187:6346-6357.e20. [PMID: 39321801 DOI: 10.1016/j.cell.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Pharmaceuticals can directly inhibit the growth of gut bacteria, but the degree to which such interactions manifest in complex community settings is an open question. Here, we compared the effects of 30 drugs on a 32-species synthetic community with their effects on each community member in isolation. While most individual drug-species interactions remained the same in the community context, communal behaviors emerged in 26% of all tested cases. Cross-protection during which drug-sensitive species were protected in community was 6 times more frequent than cross-sensitization, the converse phenomenon. Cross-protection decreased and cross-sensitization increased at higher drug concentrations, suggesting that the resilience of microbial communities can collapse when perturbations get stronger. By metabolically profiling drug-treated communities, we showed that both drug biotransformation and bioaccumulation contribute mechanistically to communal protection. As a proof of principle, we molecularly dissected a prominent case: species expressing specific nitroreductases degraded niclosamide, thereby protecting both themselves and sensitive community members.
Collapse
Affiliation(s)
- Sarela Garcia-Santamarina
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany; European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany
| | - Michael Kuhn
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany
| | - Saravanan Devendran
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany
| | - Lisa Maier
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany
| | - Marja Driessen
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany
| | - André Mateus
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany
| | - Eleonora Mastrorilli
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany
| | - Ana Rita Brochado
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany
| | - Mikhail M Savitski
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany
| | - Kiran R Patil
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany.
| | - Michael Zimmermann
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany.
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| | - Athanasios Typas
- European Molecular Biology Laboratory, Genome Biology, Heidelberg, Germany; European Molecular Biology Laboratory, Structural and Computational Biology, Heidelberg, Germany.
| |
Collapse
|
2
|
Molina-Panadero I, Morales-Tenorio M, García-Rubia A, Ginex T, Eskandari K, Martinez A, Gil C, Smani Y. Discovery of new antimicrobial thiophene derivatives with activity against drug-resistant Gram negative-bacteria. Front Pharmacol 2024; 15:1412797. [PMID: 39228527 PMCID: PMC11368766 DOI: 10.3389/fphar.2024.1412797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/03/2024] [Indexed: 09/05/2024] Open
Abstract
Our aim is to identify new small molecules with antimicrobial potential, especially against colistin-resistant (Col-R) Acinetobacter baumannii and Escherichia coli. After initial hits identification by fingerprint similarity, MIC of 24 heterocyclic derivatives for A. baumannii and E. coli reference strains, and bactericidal activity of selected thiophenes against Col-R strains were determined. We analyzed changes in bacterial membrane permeability and the OMPs profile. Additionally, we determined bacterial adherence to host cells and performed molecular docking studies to assess their binding to bacterial targets. The compounds' MICs ranged from 4 to >64 mg/L. Thiophene derivatives 4, 5 and 8 exhibited MIC50 values between 16 and 32 mg/L for Col-R A. baumannii and 8 and 32 mg/L for Col-R E. coli. The time-kill curve assay demonstrated that thiophenes 4 and 8 had bactericidal effects against Col-R A. baumannii and E. coli. Furthermore, treatment with them resulted in increased membrane permeabilization and reduced adherence of these isolates to host cells. Finally, the docking studies showed a stronger binding affinity to CarO1 and Omp33 of A. baumannii and OmpW and OmpC of E. coli. These findings indicate that thiophene derivatives possess antibacterial activity against Col-R A. baumannii and E. coli, suggesting that they may enhance the repertoire of drug treatments against bacteria.
Collapse
Affiliation(s)
- Irene Molina-Panadero
- Andalusian Center of Developmental Biology, CSIC, University of Pablo de Olavide - Seville, Seville, Spain
| | | | | | - Tiziana Ginex
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Khalil Eskandari
- Andalusian Center of Developmental Biology, CSIC, University of Pablo de Olavide - Seville, Seville, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Younes Smani
- Andalusian Center of Developmental Biology, CSIC, University of Pablo de Olavide - Seville, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, University of Pablo de Olavide, Seville, Spain
| |
Collapse
|
3
|
Ortiz-Miravalles L, Sánchez-Angulo M, Sanz JM, Maestro B. Drug Repositioning as a Therapeutic Strategy against Streptococcus pneumoniae: Cell Membrane as Potential Target. Int J Mol Sci 2023; 24:ijms24065831. [PMID: 36982905 PMCID: PMC10058218 DOI: 10.3390/ijms24065831] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
A collection of repurposing drugs (Prestwick Chemical Library) containing 1200 compounds was screened to investigate the drugs' antimicrobial effects against planktonic cultures of the respiratory pathogen Streptococcus pneumoniae. After four discrimination rounds, a set of seven compounds was finally selected, namely (i) clofilium tosylate; (ii) vanoxerine; (iii) mitoxantrone dihydrochloride; (iv) amiodarone hydrochloride; (v) tamoxifen citrate; (vi) terfenadine; and (vii) clomiphene citrate (Z, E). These molecules arrested pneumococcal growth in a liquid medium and induced a decrease in bacterial viability between 90.0% and 99.9% at 25 µM concentration, with minimal inhibitory concentrations (MICs) also in the micromolar range. Moreover, all compounds but mitoxantrone caused a remarkable increase in the permeability of the bacterial membrane and share a common, minimal chemical structure consisting of an aliphatic amine linked to a phenyl moiety via a short carbon/oxygen linker. These results open new possibilities to tackle pneumococcal disease through drug repositioning and provide clues for the design of novel membrane-targeted antimicrobials with a related chemical structure.
Collapse
Affiliation(s)
- Laura Ortiz-Miravalles
- Protein Engineering against Antimicrobial Resistance Group, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
- Department of Animal Health, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Manuel Sánchez-Angulo
- Department of Vegetal Production and Microbiology, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Jesús M Sanz
- Protein Engineering against Antimicrobial Resistance Group, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Beatriz Maestro
- Protein Engineering against Antimicrobial Resistance Group, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| |
Collapse
|
4
|
Abstract
Objectives Tamoxifen (TAM), which is used for treating breast cancer, has exhibited another important function as an antimicrobial agent. The objective of this study is to investigate the antibacterial action of TAM against the bacteria present in the human oral cavity. Materials and Methods The bacteria present in the human oral cavity were isolated from healthy individuals. Different concentrations of TAM were tested against the isolated bacteria. Additionally, bactericidal and bacteriostatic effects of TAM were also determined. Results Out of 23 isolated bacteria, a greater number of Gram-positive bacteria were highly susceptible to the low concentrations of TAM than Gram-negative bacteria. Kytococcus sedentarius, which is Gram-positive bacterium, and Pseudomonas stutzeri, which is Gram-negative bacterium, needed a high minimum inhibitory concentration value of TAM (2.5 mg/mL) to be inhibited by TAM's bacteriostatic action. Resistance to TAM was also observed in three strains of Gram-positive and four strains of Gram-negative bacteria. Conclusion TAM has shown a potential antibacterial effect against the bacteria present in the oral cavity, especially against Gram-positive bacteria. This effect is mostly bacteriostatic. This study also found bacterial resistance toward TAM.
Collapse
|
5
|
Abstract
The concept of repurposing previously approved medications to the treatment of new indications by taking advantage of off-target effects has gained traction in recent years, particularly in areas of medicine that do not offer large profits to pharmaceutical firms. As infectious disease discovery research has declined among large pharmaceutical companies, the potential payoff of repurposing has become attractive. The concept of repurposing previously approved medications to the treatment of new indications by taking advantage of off-target effects has gained traction in recent years, particularly in areas of medicine that do not offer large profits to pharmaceutical firms. As infectious disease discovery research has declined among large pharmaceutical companies, the potential payoff of repurposing has become attractive. From these efforts, the triphenylethylene class of selective estrogen receptor modulators related to tamoxifen has shown activity against a wide range of medically important human pathogens, including bacteria, fungi, parasites, and viruses. Because it has activity against many pathogens affecting people in resource-limited areas of the world, TAM and related drugs may be particularly useful. Here, we review the in vitro, in vivo, and mechanistic studies of the anti-infective activity of tamoxifen, toremifene, clomiphene, and their analogs. We also discuss the pharmacologic properties of this privileged scaffold and its potential utility in treating infectious diseases.
Collapse
|
6
|
Hussein MH, Schneider EK, Elliott AG, Han M, Reyes-Ortega F, Morris F, Blaskovich MAT, Jasim R, Currie B, Mayo M, Baker M, Cooper MA, Li J, Velkov T. From Breast Cancer to Antimicrobial: Combating Extremely Resistant Gram-Negative “Superbugs” Using Novel Combinations of Polymyxin B with Selective Estrogen Receptor Modulators. Microb Drug Resist 2017; 23:640-650. [DOI: 10.1089/mdr.2016.0196] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Maytham H. Hussein
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Elena K. Schneider
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Alysha G. Elliott
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Meiling Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Felisa Reyes-Ortega
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Faye Morris
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Mark A. T. Blaskovich
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Raad Jasim
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Bart Currie
- Menzies School of Health Research, Casuarina, Australia
| | - Mark Mayo
- Menzies School of Health Research, Casuarina, Australia
| | - Mark Baker
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, Australia
| | - Matthew A. Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Parkville, Australia
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| |
Collapse
|
7
|
Antibiotic-non-antibiotic combinations for combating extremely drug-resistant Gram-negative 'superbugs'. Essays Biochem 2017; 61:115-125. [PMID: 28258235 DOI: 10.1042/ebc20160058] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
Abstract
The emergence of antimicrobial resistance of Gram-negative pathogens has become a worldwide crisis. The status quo for combating resistance is to employ synergistic combinations of antibiotics. Faced with this fast-approaching post-antibiotic era, it is critical that we devise strategies to prolong and maximize the clinical efficacy of existing antibiotics. Unfortunately, reports of extremely drug-resistant (XDR) Gram-negative pathogens have become more common. Combining antibiotics such as polymyxin B or the broad-spectrum tetracycline and minocycline with various FDA-approved non-antibiotic drugs have emerged as a novel combination strategy against otherwise untreatable XDR pathogens. This review surveys the available literature on the potential benefits of employing antibiotic-non-antibiotic drug combination therapy. The apex of this review highlights the clinical utility of this novel therapeutic strategy for combating infections caused by 'superbugs'.
Collapse
|
8
|
El Arbi M, Théolier J, Pigeon P, Jellali K, Trigui F, Top S, Aifa S, Fliss I, Jaouen G, Hammami R. Antibacterial properties and mode of action of new triaryl butene citrate compounds. Eur J Med Chem 2014; 76:408-13. [DOI: 10.1016/j.ejmech.2014.02.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
|
9
|
Adenylate kinase release as a high-throughput-screening-compatible reporter of bacterial lysis for identification of antibacterial agents. Antimicrob Agents Chemother 2012; 57:26-36. [PMID: 23027196 DOI: 10.1128/aac.01640-12] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Adenylate kinase (AK) is a ubiquitous intracellular enzyme that is released into the extracellular space upon cell lysis. We have shown that AK release serves as a useful reporter of bactericidal agent activity and can be exploited for antimicrobial screening purposes. The AK assay exhibits improved sensitivity over that of growth-based assays and can detect agents that are active against bacteria in clinically relevant growth states that are difficult to screen using conventional approaches, such as small colony variants (SCV) and bacteria within established biofilms. The usefulness of the AK assay was validated by screening a library of off-patent drugs for agents that exhibit antimicrobial properties toward a variety of bacterial species, including Escherichia coli and all members of the "ESKAPE" pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). The assay detected antibiotics within the library that were expected to be active against the organism screened. Moreover, 38 drugs with no previously reported antibacterial activity elicited AK release. Four of these were acquired, and all were verified to exhibit antimicrobial activity by standard susceptibility testing. Two of these molecules were further characterized. The antihistamine, terfenadine, was active against S. aureus planktonic, SCV population, and biofilm-associated cells. Tamoxifen, an estrogen receptor antagonist, was active toward E. faecium in vitro and also reduced E. faecium pathogenesis in a Galleria mellonella infection model. Our data demonstrate that the AK assay provides an attractive screening approach for identifying new antimicrobial agents. Further, terfenadine and tamoxifen may represent novel antimicrobial drug development scaffolds.
Collapse
|
10
|
Pereira SP, Fernandes MAS, Martins JD, Santos MS, Moreno AJM, Vicente JAF, Videira RA, Jurado AS. Toxicity assessment of the herbicide metolachlor comparative effects on bacterial and mitochondrial model systems. Toxicol In Vitro 2009; 23:1585-90. [PMID: 19607910 DOI: 10.1016/j.tiv.2009.06.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 06/02/2009] [Accepted: 06/30/2009] [Indexed: 11/18/2022]
Abstract
Metolachlor is one of the most intensively used chloroacetamide herbicides. However, its effects on the environment and on non-target animals and humans as well as its interference at a cell/molecular level have not yet been fully elucidated. The aim of this study was: firstly, to evaluate the potential toxicity of metolachlor at a cell/subcellular level by using two in vitro biological model systems (a strain of Bacillus stearothermophilus and rat liver mitochondria); secondly, to evaluate the relative sensibility of these models to xenobiotics to reinforce their suitability for pollutant toxicity assessment. Our results show that metolachlor inhibits growth and impairs the respiratory activity of B.stearothermophilus at concentrations two to three orders of magnitude higher than those at which bacterial cells are affected by other pesticides. Also at concentrations significantly higher than those of other pesticides, metolachlor depressed the respiratory control ratio, membrane potential and respiration of rat liver mitochondria when malate/glutamate or succinate were used as respiratory substrates. Moreover, metolachlor impaired the respiratory activity of rat liver mitochondria in the same concentration range at which it inhibited bacterial respiratory system (0.4-5.0 micromol/mg of protein). In conclusion, the high concentration range at which metolachlor induces toxicity in vitro suggests that this compound is safer than other pesticides previously studied in our laboratory, using the same model systems. The good parallelism between metolachlor effects on both models and the toxicity data described in the literature, together with results obtained in our laboratory with other compounds, indicate the suitability of these systems to assess toxicity in vitro.
Collapse
Affiliation(s)
- Susana P Pereira
- CNC - Centre for Neuroscience and Cell Biology, Zoology Department, Coimbra University, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Qi LF, Xu ZR, Li Y, Jiang X, Han XY. In vitro effects of chitosan nanoparticles on proliferation of human gastric carcinoma cell line MGC803 cells. World J Gastroenterol 2005; 11:5136-41. [PMID: 16127742 PMCID: PMC4320385 DOI: 10.3748/wjg.v11.i33.5136] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of chitosan nanoparticles on proliferation of human gastric carcinoma cell line MGC803 in vitro and the possible mechanisms involved.
METHODS: Chitosan nanoparticles were characterized by particle size, zeta potential, and morphology. After treatment with various concentrations of chitosan nanoparticles (25, 50, 75, 100 μg/mL) at various time intervals, cell proliferation, ultrastructural changes, DNA fragmentation, mitochondrial membrane potential (MMP), cell cycle phase distribution and apoptotic peaks of MGC803 cells were analyzed by MTT assay, electron microscopy, DNA agarose gel electrophoresis, and flow cytometry.
RESULTS: Chitosan nanoparticles exhibited a small particle size as 65 nm and a high surface charge as 52 mV. Chitosan nanoparticles markedly inhibited cell proliferation of MGC803 cells with an IC50 value of 5.3 μg/mL 48 h after treatment. After treatment with chitosan nanoparticles, the typical necrotic cell morphology was observed by electron microscopy, a typical DNA degradation associated with necrosis was determined by DNA agarose electrophoresis. Flow cytometry showed the loss of MMP and occurrence of apoptosis in chitosan nanoparticles-treated cells.
CONCLUSION: Chitosan nanoparticles effectively inhibit the proliferation of human gastric carcinoma cell line MGC803 in vitro through multiple mechanisms, and may be a beneficial agent against human carcinoma.
Collapse
Affiliation(s)
- Li-Feng Qi
- Nano-biology Laboratory of Animal Science College, Zhejiang University, Hangzhou 310029, Zhejiang Province, China.
| | | | | | | | | |
Collapse
|