1
|
Shideler S, Bookout T, Qasim A, Bowron L, Wu Q, Duan K, Treu R, Reckseidler-Zenteno S, Lewenza S. Biosensor-guided detection of outer membrane-specific antimicrobial activity against Pseudomonas aeruginosa from fungal cultures and medicinal plant extracts. Microbiol Spectr 2023; 11:e0153623. [PMID: 37882578 PMCID: PMC10714926 DOI: 10.1128/spectrum.01536-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE New approaches are needed to discover novel antimicrobials, particularly antibiotics that target the Gram-negative outer membrane. By exploiting bacterial sensing and responses to outer membrane (OM) damage, we used a biosensor approach consisting of polymyxin resistance gene transcriptional reporters to screen natural products and a small drug library for biosensor activity that indicates damage to the OM. The diverse antimicrobial compounds that cause induction of the polymyxin resistance genes, which correlates with outer membrane damage, suggest that these LPS and surface modifications also function in short-term repair to sublethal exposure and are required against broad membrane stress conditions.
Collapse
Affiliation(s)
- Steve Shideler
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Tyson Bookout
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Azka Qasim
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Lauren Bowron
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Qiaolian Wu
- College of Life Sciences, Northwest University, Xian, China
| | - Kangmin Duan
- Department of Oral Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Roland Treu
- Faculty of Science and Technology, Athabasca University, Athabasca, Alberta, Canada
| | - Shauna Reckseidler-Zenteno
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Faculty of Science and Technology, Athabasca University, Athabasca, Alberta, Canada
| | - Shawn Lewenza
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Faculty of Science and Technology, Athabasca University, Athabasca, Alberta, Canada
| |
Collapse
|
2
|
Alkatheri AH, Yap PSX, Abushelaibi A, Lai KS, Cheng WH, Erin Lim SH. Microbial Genomics: Innovative Targets and Mechanisms. Antibiotics (Basel) 2023; 12:190. [PMID: 36830101 PMCID: PMC9951906 DOI: 10.3390/antibiotics12020190] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Multidrug resistance (MDR) has become an increasing threat to global health because bacteria can develop resistance to antibiotics over time. Scientists worldwide are searching for new approaches that go beyond traditional antibiotic discovery and development pipelines. Advances in genomics, however, opened up an unexplored therapeutic opportunity for the discovery of new antibacterial agents. Genomic approaches have been used to discover several novel antibiotics that target critical processes for bacterial growth and survival, including histidine kinases (HKs), LpxC, FabI, peptide deformylase (PDF), and aminoacyl-tRNA synthetases (AaRS). In this review, we will discuss the use of microbial genomics in the search for innovative and promising drug targets as well as the mechanisms of action for novel antimicrobial agents. We will also discuss future directions on how the utilization of the microbial genomics approach could improve the odds of antibiotic development having a more successful outcome.
Collapse
Affiliation(s)
- Asma Hussain Alkatheri
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Polly Soo-Xi Yap
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor 47500, Malaysia
| | - Aisha Abushelaibi
- Office of Campus Director, Abu Dhabi Colleges, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Wan-Hee Cheng
- Faculty of Health and Life Sciences, INTI International University, Persiaran Perdana BBN, Nilai 71800, Malaysia
| | - Swee-Hua Erin Lim
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| |
Collapse
|
3
|
Kepplinger B, Mardiana L, Cowell J, Morton-Laing S, Dashti Y, Wills C, Marrs ECL, Perry JD, Gray J, Goodfellow M, Errington J, Probert MR, Clegg W, Bogaerts J, Herrebout W, Allenby NEE, Hall MJ. Discovery, isolation, heterologous expression and mode-of-action studies of the antibiotic polyketide tatiomicin from Amycolatopsis sp. DEM30355. Sci Rep 2022; 12:15579. [PMID: 36114335 PMCID: PMC9481585 DOI: 10.1038/s41598-022-18726-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/18/2022] [Indexed: 11/08/2022] Open
Abstract
A genomic and bioactivity informed analysis of the metabolome of the extremophile Amycolatopsis sp. DEM30355 has allowed for the discovery and isolation of the polyketide antibiotic tatiomicin. Identification of the biosynthetic gene cluster was confirmed by heterologous expression in Streptomyces coelicolor M1152. Structural elucidation, including absolute stereochemical assignment, was performed using complementary crystallographic, spectroscopic and computational methods. Tatiomicin shows antibiotic activity against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). Cytological profiling experiments suggest a putative antibiotic mode-of-action, involving membrane depolarisation and chromosomal decondensation of the target bacteria.
Collapse
Affiliation(s)
- Bernhard Kepplinger
- Biopharmaceutical Bioprocessing Technology Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK.
- Demuris Limited, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne, NE4 5BX, UK.
- Department of Molecular Microbiology, Faculty of Biotechnology, University of Wrocław, 50-383, Wrocław, Poland.
| | - Lina Mardiana
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Joseph Cowell
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Stephanie Morton-Laing
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Yousef Dashti
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK
| | - Corinne Wills
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Emma C L Marrs
- Department of Microbiology, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - John D Perry
- Department of Microbiology, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Joe Gray
- Pinnacle Laboratory, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK
| | - Michael Goodfellow
- Biology, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Jeff Errington
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE2 4AX, UK
- Demuris Limited, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne, NE4 5BX, UK
| | - Michael R Probert
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - William Clegg
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Jonathan Bogaerts
- Molecular Spectroscopy, Department of Chemistry, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Wouter Herrebout
- Molecular Spectroscopy, Department of Chemistry, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Nick E E Allenby
- Demuris Limited, The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne, NE4 5BX, UK.
| | - Michael J Hall
- Chemistry, School of Environmental and Natural Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
4
|
Yuan T, Werman JM, Sampson NS. The pursuit of mechanism of action: uncovering drug complexity in TB drug discovery. RSC Chem Biol 2021; 2:423-440. [PMID: 33928253 PMCID: PMC8081351 DOI: 10.1039/d0cb00226g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Whole cell-based phenotypic screens have become the primary mode of hit generation in tuberculosis (TB) drug discovery during the last two decades. Different drug screening models have been developed to mirror the complexity of TB disease in the laboratory. As these culture conditions are becoming more and more sophisticated, unraveling the drug target and the identification of the mechanism of action (MOA) of compounds of interest have additionally become more challenging. A good understanding of MOA is essential for the successful delivery of drug candidates for TB treatment due to the high level of complexity in the interactions between Mycobacterium tuberculosis (Mtb) and the TB drug used to treat the disease. There is no single "standard" protocol to follow and no single approach that is sufficient to fully investigate how a drug restrains Mtb. However, with the recent advancements in -omics technologies, there are multiple strategies that have been developed generally in the field of drug discovery that have been adapted to comprehensively characterize the MOAs of TB drugs in the laboratory. These approaches have led to the successful development of preclinical TB drug candidates, and to a better understanding of the pathogenesis of Mtb infection. In this review, we describe a plethora of efforts based upon genetic, metabolomic, biochemical, and computational approaches to investigate TB drug MOAs. We assess these different platforms for their strengths and limitations in TB drug MOA elucidation in the context of Mtb pathogenesis. With an emphasis on the essentiality of MOA identification, we outline the unmet needs in delivering TB drug candidates and provide direction for further TB drug discovery.
Collapse
Affiliation(s)
- Tianao Yuan
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| | - Joshua M. Werman
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| | - Nicole S. Sampson
- Department of Chemistry, Stony Brook UniversityStony BrookNY 11794-3400USA+1-631-632-5738+1-631-632-7952
| |
Collapse
|
5
|
Wex KW, Saur JS, Handel F, Ortlieb N, Mokeev V, Kulik A, Niedermeyer THJ, Mast Y, Grond S, Berscheid A, Brötz-Oesterhelt H. Bioreporters for direct mode of action-informed screening of antibiotic producer strains. Cell Chem Biol 2021; 28:1242-1252.e4. [PMID: 33761329 DOI: 10.1016/j.chembiol.2021.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/27/2021] [Accepted: 02/23/2021] [Indexed: 01/17/2023]
Abstract
A big challenge in natural product research of today is rapid dereplication of already known substances, to free capacities for the exploration of new agents. Prompt information on bioactivities and mode of action (MOA) speeds up the lead discovery process and is required for rational compound optimization. Here, we present a bioreporter approach as a versatile strategy for combined bioactivity- and MOA-informed primary screening for antimicrobials. The approach is suitable for directly probing producer strains grown on agar, without need for initial compound enrichment or purification, and works along the entire purification pipeline with culture supernatants, extracts, fractions, and pure substances. The technology allows for MOA-informed purification to selectively prioritize activities of interest. In combination with high-resolution mass spectrometry, the biosensor panel is an efficient and sensitive tool for compound deconvolution. Concomitant information on the affected metabolic pathway enables the selection of appropriate follow-up assays to elucidate the molecular target.
Collapse
Affiliation(s)
- Katharina W Wex
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Julian S Saur
- Biomolecular Chemistry, Institute of Organic Chemistry, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Franziska Handel
- Department of Microbiology and Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Nico Ortlieb
- Department of Microbiology and Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Vladislav Mokeev
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Cluster of Excellence EXC 2124 - Controlling Microbes to Fight Infections, Tuebingen, Baden-Württemberg 72076, Germany
| | - Andreas Kulik
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Department of Microbiology and Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Cluster of Excellence EXC 2124 - Controlling Microbes to Fight Infections, Tuebingen, Baden-Württemberg 72076, Germany
| | - Timo H J Niedermeyer
- Department of Microbiology and Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Department of Pharmaceutical Biology/Pharmacognosy Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Sachsen-Anhalt 06120, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Yvonne Mast
- Department of Microbiology and Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Department Bioresources for Bioeconomy and Health Research, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Niedersachsen 38124, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Stephanie Grond
- Biomolecular Chemistry, Institute of Organic Chemistry, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Cluster of Excellence EXC 2124 - Controlling Microbes to Fight Infections, Tuebingen, Baden-Württemberg 72076, Germany
| | - Anne Berscheid
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany
| | - Heike Brötz-Oesterhelt
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; German Center for Infection Research (DZIF), Partner Site Tuebingen, Tuebingen, Baden-Württemberg 72076, Germany; Cluster of Excellence EXC 2124 - Controlling Microbes to Fight Infections, Tuebingen, Baden-Württemberg 72076, Germany.
| |
Collapse
|
6
|
Emami K, Wu LJ, Errington J. A Small Molecule Inhibitor of CTP Synthetase Identified by Differential Activity on a Bacillus subtilis Mutant Deficient in Class A Penicillin-Binding Proteins. Front Microbiol 2020; 11:2001. [PMID: 32973723 PMCID: PMC7479849 DOI: 10.3389/fmicb.2020.02001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/28/2020] [Indexed: 12/03/2022] Open
Abstract
In the course of screening for compounds with differential growth inhibition activity on a mutant of Bacillus subtilis lacking all four class A penicillin-binding proteins (Δ4), we came across an isoquinoline derivative, IQ-1 carboxylic acid (IQC) with relatively high activity on the mutant compared to the wild type strain. Treated cells were slightly elongated and had altered chromosome morphology. Mutants of Δ4 resistant to IQC were isolated and subjected to whole genome sequencing. Most of the mutants were affected in the gene, pyrG, encoding CTP synthetase (CTPS). Purified wild type CTPS was inhibited in vitro by IQC. Two of the three mutant proteins purified showed decreased sensitivity to IQC in vitro. Finally, inhibition by IQC was rescued by addition of cytidine but not uridine to the growth medium, consistent with the notion that IQC acts by reducing the synthesis of CTP or a related compound. IQC provides a promising new starting point for antibiotic inhibitors of CTPS.
Collapse
Affiliation(s)
- Kaveh Emami
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ling Juan Wu
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jeff Errington
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
7
|
Kénanian G, Morvan C, Weckel A, Pathania A, Anba-Mondoloni J, Halpern D, Gaillard M, Solgadi A, Dupont L, Henry C, Poyart C, Fouet A, Lamberet G, Gloux K, Gruss A. Permissive Fatty Acid Incorporation Promotes Staphylococcal Adaptation to FASII Antibiotics in Host Environments. Cell Rep 2020; 29:3974-3982.e4. [PMID: 31851927 DOI: 10.1016/j.celrep.2019.11.071] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/01/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023] Open
Abstract
The essentiality of fatty acid synthesis (FASII) products in the human pathogen Staphylococcus aureus is the underlying rationale for FASII-targeted antimicrobial drug design. Reports of anti-FASII efficacy in animals support this choice. However, restricted test conditions used previously led us to investigate this postulate in a broader, host-relevant context. We report that S. aureus rapidly adapts to FASII antibiotics without FASII mutations when exposed to host environments. FASII antibiotic administration upon signs of infection, rather than just after inoculation as commonly practiced, fails to eliminate S. aureus in a septicemia model. In vitro, serum lowers S. aureus membrane stress, leading to a greater retention of the substrates required for environmental fatty acid (eFA) utilization: eFAs and the acyl carrier protein. In this condition, eFA occupies both phospholipid positions, regardless of anti-FASII selection. Our results identify S. aureus membrane plasticity in host environments as a main limitation for using FASII antibiotics in monotherapeutic treatments.
Collapse
Affiliation(s)
- Gérald Kénanian
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Claire Morvan
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Antonin Weckel
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014 Paris, France
| | - Amit Pathania
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Jamila Anba-Mondoloni
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - David Halpern
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Marine Gaillard
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014 Paris, France
| | - Audrey Solgadi
- SAMM, UMS IPSIT, Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Laetitia Dupont
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Céline Henry
- PAPPSO Platform, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Claire Poyart
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014 Paris, France; Centre National de Référence des Streptocoques, Hôpitaux Universitaires Paris Centre Site Cochin, APHP, Paris, France
| | - Agnès Fouet
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014 Paris, France
| | - Gilles Lamberet
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Karine Gloux
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Alexandra Gruss
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France.
| |
Collapse
|
8
|
Richter MF, Hergenrother PJ. The challenge of converting Gram-positive-only compounds into broad-spectrum antibiotics. Ann N Y Acad Sci 2018; 1435:18-38. [PMID: 29446459 DOI: 10.1111/nyas.13598] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 12/14/2022]
Abstract
Multidrug resistant Gram-negative bacterial infections are on the rise, and there is a lack of new classes of drugs to treat these pathogens. This drug shortage is largely due to the challenge of finding antibiotics that can permeate and persist inside Gram-negative species. Efforts to understand the molecular properties that enable certain compounds to accumulate in Gram-negative bacteria based on retrospective studies of known antibiotics have not been generally actionable in the development of new antibiotics. A recent assessment of the ability of >180 diverse small molecules to accumulate in Escherichia coli led to predictive guidelines for compound accumulation in E. coli. These "eNTRy rules" state that compounds are most likely to accumulate if they contain a nonsterically encumbered ionizable Nitrogen (primary amines are the best), have low Three-dimensionality (globularity ≤ 0.25), and are relatively Rigid (rotatable bonds ≤ 5). In this review, we look back through 50+ years of antibacterial research and 1000s of derivatives and assess this historical data set through the lens of these predictive guidelines. The results are consistent with the eNTRy rules, suggesting that the eNTRy rules may provide an actionable and general roadmap for the conversion of Gram-positive-only compounds into broad-spectrum antibiotics.
Collapse
Affiliation(s)
- Michelle F Richter
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
9
|
Kepplinger B, Morton-Laing S, Seistrup KH, Marrs ECL, Hopkins AP, Perry JD, Strahl H, Hall MJ, Errington J, Allenby NEE. Mode of Action and Heterologous Expression of the Natural Product Antibiotic Vancoresmycin. ACS Chem Biol 2018; 13:207-214. [PMID: 29185696 DOI: 10.1021/acschembio.7b00733] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Antibiotics that interfere with the bacterial cytoplasmic membrane have long-term potential for the treatment of infectious diseases as this mode of action is anticipated to result in low resistance frequency. Vancoresmycin is an understudied natural product antibiotic consisting of a terminal tetramic acid moiety fused to a linear, highly oxygenated, stereochemically complex polyketide chain. Vancoresmycin shows minimum inhibitory concentrations (MICs) from 0.125 to 2 μg/mL against a range of clinically relevant, antibiotic-resistant Gram-positive bacteria. Through a comprehensive mode-of-action study, utilizing Bacillus subtilis reporter strains, DiSC3(5) depolarization assays, and fluorescence microscopy, we have shown that vancoresmycin selectively targets the cytoplasmic membrane of Gram-positive bacteria via a non-pore-forming, concentration-dependent depolarization mechanism. Whole genome sequencing of the producing strain allowed identification of the 141 kbp gene cluster encoding for vancoresmycin biosynthesis and a preliminary model for its biosynthesis. The size and complex structure of vancoresmycin could confound attempts to generate synthetic analogues. To overcome this problem and facilitate future studies, we identified, cloned, and expressed the 141 kbp biosynthetic gene cluster in Streptomyces coelicolor M1152. Elucidation of the mode-of-action of vancoresmycin, together with the heterologous expression system, will greatly facilitate further studies of this and related molecules.
Collapse
Affiliation(s)
- Bernhard Kepplinger
- Centre for Bacterial Cell Biology, Newcastle University
, Newcastle upon Tyne NE2 4BN, United Kingdom
- Demuris Limited , Newcastle Biomedicine Bio-Incubators
, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Stephanie Morton-Laing
- Demuris Limited , Newcastle Biomedicine Bio-Incubators
, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Kenneth Holst Seistrup
- Centre for Bacterial Cell Biology, Newcastle University
, Newcastle upon Tyne NE2 4BN, United Kingdom
| | | | - Adam Paul Hopkins
- Demuris Limited , Newcastle Biomedicine Bio-Incubators
, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - John David Perry
- Microbiology Department, Freeman Hospital
, Newcastle upon Tyne NE7 7DN, United Kingdom
| | - Henrik Strahl
- Centre for Bacterial Cell Biology, Newcastle University
, Newcastle upon Tyne NE2 4BN, United Kingdom
| | - Michael John Hall
- School of Chemistry, Newcastle University
, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Jeff Errington
- Centre for Bacterial Cell Biology, Newcastle University
, Newcastle upon Tyne NE2 4BN, United Kingdom
- Demuris Limited , Newcastle Biomedicine Bio-Incubators
, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Nicholas Edward Ellis Allenby
- Demuris Limited , Newcastle Biomedicine Bio-Incubators
, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| |
Collapse
|
10
|
Domínguez Á, Muñoz E, López MC, Cordero M, Martínez JP, Viñas M. Transcriptomics as a tool to discover new antibacterial targets. Biotechnol Lett 2017; 39:819-828. [PMID: 28289911 DOI: 10.1007/s10529-017-2319-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
The emergence of antibiotic-resistant pathogens, multiple drug-resistance, and extremely drug-resistant strains demonstrates the need for improved strategies to discover new drug-based compounds. The development of transcriptomics, proteomics, and metabolomics has provided new tools for global studies of living organisms. However, the compendium of expression profiles produced by these methods has introduced new scientific challenges into antimicrobial research. In this review, we discuss the practical value of transcriptomic techniques as well as their difficulties and pitfalls. We advocate the construction of new databases of transcriptomic data, using standardized formats in addition to standardized models of bacterial and yeast similar to those used in systems biology. The inclusion of proteomic and metabolomic data is also essential, as the resulting networks can provide a landscape to rationally predict and exploit new drug targets and to understand drug synergies.
Collapse
Affiliation(s)
- Ángel Domínguez
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain.
| | - Elisa Muñoz
- Department of Cell Biology & Pathology, Universidad de Salamanca, Salamanca, Spain
| | - M Carmen López
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain
| | - Miguel Cordero
- Department of Medicine, Universidad de Salamanca, Salamanca, Spain
| | - José Pedro Martínez
- Department of Microbiology & Ecology, Universitat de Valencia/Estudi General (UVEG), Valencia, Spain
| | - Miguel Viñas
- Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
de la Cruz M, González I, Parish CA, Onishi R, Tormo JR, Martín J, Peláez F, Zink D, El Aouad N, Reyes F, Genilloud O, Vicente F. Production of Ramoplanin and Ramoplanin Analogs by Actinomycetes. Front Microbiol 2017; 8:343. [PMID: 28321210 PMCID: PMC5337499 DOI: 10.3389/fmicb.2017.00343] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/20/2017] [Indexed: 01/01/2023] Open
Abstract
Ramoplanin is a glycolipodepsipeptide antibiotic obtained from fermentation of Actinoplanes sp. ATCC 33076 that exhibits activity against clinically important multi-drug-resistant, Gram-positive pathogens including vancomycin-resistant Enterococcus (VRE), methicillin-resistant Staphylococcus aureus (MRSA), and vancomycin-intermediate resistant Clostridium difficile. It disrupts bacterial cell wall through a unique mechanism of action by sequestering the peptidoglycan intermediate Lipid II and therefore does not show cross-resistance with other antibiotics. However, while demonstrating excellent antimicrobial activity in systemic use in animal models of infection, ramoplanin presents low local tolerability when injected intravenously. As a consequence of this limitation, new derivatives are desirable to overcome this issue. During a natural product screening program developed to discover compounds that disrupt bacterial cell wall synthesis by inhibiting peptidoglycan transglycosylation through binding to the intermediate Lipid II, 49 actinomycete strains were identified by HR-LCMS as producers of ramoplanin-related compounds. The producing strains were isolated from environmental samples collected worldwide comprising both tropical and temperate areas. To assess the diversity of this microbial population, the 49 isolates were initially identified to the genus level on the basis of their micromorphology, and 16S sequencing confirmed the initial identification of the strains. These analyses resulted in the identification of members of genus Streptomyces, as well as representatives of the families Micromonosporaceae, Nocardiaceae, Thermomonosporaceae, and Pseudonocardiaceae, suggesting that the production of ramoplanins is relatively widespread among Actinomycetes. In addition, all of these isolates were tested against a panel of Gram-positive and Gram-negative bacteria, filamentous fungi, and yeast in order to further characterize their antimicrobial properties. This work describes the diversity of actinomycete strains that produced ramoplanin-related compounds, and the analysis of the antimicrobial activity exhibited by these isolates. Our results strongly suggest the presence of new ramoplanin-analogs among these actinomycete producers.
Collapse
Affiliation(s)
- Mercedes de la Cruz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | - Ignacio González
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | | | | | - José R Tormo
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | - Jesús Martín
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | - Fernando Peláez
- Biotechnology Programme, Spanish National Cancer Research Centre Madrid, Spain
| | - Debbie Zink
- Merck Research Laboratories, Merck Kenilworth, NJ, USA
| | | | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía Granada, Spain
| |
Collapse
|
12
|
Bioluminescent Reporters for Rapid Mechanism of Action Assessment in Tuberculosis Drug Discovery. Antimicrob Agents Chemother 2016; 60:6748-6757. [PMID: 27572410 DOI: 10.1128/aac.01178-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/26/2016] [Indexed: 12/12/2022] Open
Abstract
The tuberculosis (TB) drug discovery pipeline is fueled by compounds identified in whole-cell screens against the causative agent, Mycobacterium tuberculosis Phenotypic screening enables the selection of molecules that inhibit essential cellular functions in live, intact bacilli grown under a chosen in vitro condition. However, deducing the mechanism of action (MOA), which is important to avoid promiscuous targets, often requires significant biological resources in a lengthy process that risks decoupling medicinal chemistry and biology efforts. Therefore, there is a need to develop methods enabling rapid MOA assessment of putative "actives" for triage decisions. Here, we describe a modified version of a bioluminescence reporter assay that allows nondestructive detection of compounds targeting either of two macromolecular processes in M. tuberculosis: cell wall biosynthesis or maintenance of DNA integrity. Coupling the luxCDABE operon from Photorhabdus luminescens to mycobacterial promoters driving expression of the iniBAC operon (PiniB-LUX) or the DNA damage-inducible genes, recA (PrecA-LUX) or radA (PradA-LUX), provided quantitative detection in real time of compounds triggering expression of any of these promoters over an extended 10- to 12-day incubation. Testing against known anti-TB agents confirmed the specificity of each reporter in registering the MOA of the applied antibiotic in M. tuberculosis, independent of bactericidal or bacteriostatic activity. Moreover, profiles obtained for experimental compounds indicated the potential to infer complex MOAs in which multiple cellular processes are disrupted. These results demonstrate the utility of the reporters for early triage of compounds based on the provisional MOA and suggest their application to investigate polypharmacology in known and experimental anti-TB agents.
Collapse
|
13
|
Silver LL. Natural products as a source of drug leads to overcome drug resistance. Future Microbiol 2015; 10:1711-8. [DOI: 10.2217/fmb.15.67] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Novel antibacterials may be found that can enhance the pipeline of therapeutics capable of overcoming antibiotic resistance by a return to exploration of natural products. Such novel products may be derived from both standard and previously uncultivable sources, and enriched by expression of previously unseen antibiotics predicted by genome mining of productive bacterial genera. Hypersensitive whole cell phenotypic screens can be used to detect novel secondary metabolites from both standard and newly uncovered sources.
Collapse
Affiliation(s)
- Lynn L Silver
- LL Silver Consulting, LLC, 955 South Springfield Avenue, Unit C403, Springfield, NJ 07081, USA
| |
Collapse
|
14
|
Target mechanism-based whole-cell screening identifies bortezomib as an inhibitor of caseinolytic protease in mycobacteria. mBio 2015; 6:e00253-15. [PMID: 25944857 PMCID: PMC4436076 DOI: 10.1128/mbio.00253-15] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A novel type of antibacterial screening method, a target mechanism-based whole-cell screening method, was developed to combine the advantages of target mechanism- and whole-cell-based approaches. A mycobacterial reporter strain with a synthetic phenotype for caseinolytic protease (ClpP1P2) activity was engineered, allowing the detection of inhibitors of this enzyme inside intact bacilli. A high-throughput screening method identified bortezomib, a human 26S proteasome drug, as a potent inhibitor of ClpP1P2 activity and bacterial growth. A battery of secondary assays was employed to demonstrate that bortezomib indeed exerts its antimicrobial activity via inhibition of ClpP1P2: Down- or upmodulation of the intracellular protease level resulted in hyper- or hyposensitivity of the bacteria, the drug showed specific potentiation of translation error-inducing aminoglycosides, ClpP1P2-specific substrate WhiB1 accumulated upon exposure, and growth inhibition potencies of bortezomib derivatives correlated with ClpP1P2 inhibition potencies. Furthermore, molecular modeling showed that the drug can bind to the catalytic sites of ClpP1P2. This work demonstrates the feasibility of target mechanism-based whole-cell screening, provides chemical validation of ClpP1P2 as a target, and identifies a drug in clinical use as a new lead compound for tuberculosis therapy. During the last decade, antibacterial drug discovery relied on biochemical assays, rather than whole-cell approaches, to identify molecules that interact with purified target proteins derived by genomics. This approach failed to deliver antibacterial compounds with whole-cell activity, either because of cell permeability issues that medicinal chemistry cannot easily fix or because genomic data of essentiality insufficiently predicted the vulnerability of the target identified. As a consequence, the field largely moved back to a whole-cell approach whose main limitation is its black-box nature, i.e., that it requires trial-and-error chemistry because the cellular target is unknown. We developed a novel type of antibacterial screening method, target mechanism-based whole-cell screening, to combine the advantages of both approaches. We engineered a mycobacterial reporter strain with a synthetic phenotype allowing us to identify inhibitors of the caseinolytic protease (ClpP1P2) inside the cell. This approach identified bortezomib, an anticancer drug, as a specific inhibitor of ClpP1P2. We further confirmed the specific “on-target” activity of bortezomib by independent approaches including, but not limited to, genetic manipulation of the target level (over- and underexpressing strains) and by establishing a dynamic structure-activity relationship between ClpP1P2 and growth inhibition. Identifying an “on-target” compound is critical to optimize the efficacy of the compound without compromising its specificity. This work demonstrates the feasibility of target mechanism-based whole-cell screening methods, validates ClpP1P2 as a druggable target, and delivers a lead compound for tuberculosis therapy.
Collapse
|
15
|
Monciardini P, Iorio M, Maffioli S, Sosio M, Donadio S. Discovering new bioactive molecules from microbial sources. Microb Biotechnol 2014; 7:209-20. [PMID: 24661414 PMCID: PMC3992017 DOI: 10.1111/1751-7915.12123] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/16/2014] [Accepted: 02/16/2014] [Indexed: 01/01/2023] Open
Abstract
There is an increased need for new drug leads to treat diseases in humans, animals and plants. A dramatic example is represented by the need for novel and more effective antibiotics to combat multidrug-resistant microbial pathogens. Natural products represent a major source of approved drugs and still play an important role in supplying chemical diversity, despite a decreased interest by large pharmaceutical companies. Novel approaches must be implemented to decrease the chances of rediscovering the tens of thousands of known natural products. In this review, we present an overview of natural product screening, focusing particularly on microbial products. Different approaches can be implemented to increase the probability of finding new bioactive molecules. We thus present the rationale and selected examples of the use of hypersensitive assays; of accessing unexplored microorganisms, including the metagenome; and of genome mining. We then focus our attention on the technology platform that we are currently using, consisting of approximately 70,000 microbial strains, mostly actinomycetes and filamentous fungi, and discuss about high-quality screening in the search for bioactive molecules. Finally, two case studies are discussed, including the spark that arose interest in the compound: in the case of orthoformimycin, the novel mechanism of action predicted a novel structural class; in the case of NAI-112, structural similarity pointed out to a possible in vivo activity. Both predictions were then experimentally confirmed.
Collapse
|
16
|
Double recombinant Mycobacterium bovis BCG strain for screening of primary and rationale-based antimycobacterial compounds. Antimicrob Agents Chemother 2013; 58:1389-96. [PMID: 24342633 DOI: 10.1128/aac.01301-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Conventional antimycobacterial screening involves CFU analysis, which poses a great challenge due to slow growth of mycobacteria. Recombinant strains carrying reporter genes under the influence of constitutive promoters allow rapid and wide screening of compounds but without revealing their modes of action. Reporter strains using pathway-specific promoters provide a better alternative but allow a limited screening of compounds interfering with only a particular metabolic pathway. This reduces these strains to merely a second-line screening system, as they fail to identify even the more potent compounds if they are not inhibiting the pathway of interest. In this study, we have generated a double recombinant Mycobacterium bovis BCG strain carrying firefly and Renilla luciferase genes as two reporters under the control of a constitutive and an inducible mycobacterial promoter. The presence of dual reporters allows simultaneous expression and analysis of two reporter enzymes within a single system. The expression profile of the firefly luciferase gene, rendered by a constitutive mycobacterial promoter, coincides with the decline in bacterial growth in response to a wide range of antimycobacterial drugs, while the enhanced expression of Renilla luciferase mirrors the selective induction of the reporter gene expression as a result of pathway-specific inhibition. Thus, the double recombinant strain allows the screening of both primary and rationally synthesized antimycobacterial compounds in a single assay. The inhibiting response of drugs was monitored with a dual-luciferase reporter assay which can be easily adapted in high-throughput mode.
Collapse
|
17
|
Silver LL. Antibacterial Discovery: Problems and Possibilities. Antibiotics (Basel) 2013. [DOI: 10.1002/9783527659685.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
18
|
Ooi N, Chopra I, Eady A, Cove J, Bojar R, O'Neill AJ. Antibacterial activity and mode of action of tert-butylhydroquinone (TBHQ) and its oxidation product, tert-butylbenzoquinone (TBBQ). J Antimicrob Chemother 2013; 68:1297-304. [PMID: 23463211 DOI: 10.1093/jac/dkt030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES The antioxidant tert-butylhydroquinone (TBHQ) is a food additive reported to have antibacterial activity, and may therefore have application in the healthcare setting. This study sought to characterize the antibacterial activity and mode of action of TBHQ and its oxidation product, tert-butylbenzoquinone (TBBQ). METHODS The stability of TBHQ/TBBQ was studied in buffer. Susceptibility testing was performed by broth microdilution, and killing and lytic activity were evaluated by viable counting and culture turbidity measurements. Mode of action studies included following the incorporation of radiolabelled precursors into macromolecules. The effect of TBHQ/TBBQ upon bacterial and mammalian membranes was assessed using the BacLight(TM) assay and by monitoring the haemolysis of equine erythrocytes. RESULTS TBHQ underwent oxidation in solution to form TBBQ. When oxidation was prevented, TBHQ lacked useful antibacterial activity, indicating that TBBQ is responsible for the antibacterial activity attributed to TBHQ. TBBQ demonstrated activity against Staphylococcus aureus SH1000 (MIC 8 mg/L) and against a panel of clinical S. aureus isolates (MIC90 16 mg/L). TBBQ at 4× MIC caused a >4 log10 drop in cell viability within 6 h without lysis, and eradicated staphylococcal biofilms at 8× MIC. TBBQ did not display preferential inhibition of any single macromolecular synthetic pathway, but caused loss of staphylococcal membrane integrity without haemolytic activity. CONCLUSIONS TBBQ is responsible for the antibacterial activity previously ascribed to TBHQ. TBBQ prompts loss of staphylococcal membrane integrity; it is rapidly and extensively bactericidal, but is non-lytic. In view of the potent and selective bactericidal activity of TBBQ, this compound warrants further investigation as a candidate antistaphylococcal agent.
Collapse
Affiliation(s)
- N Ooi
- Antimicrobial Research Centre and School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | | | | | | | | | | |
Collapse
|
19
|
Raut N, O'Connor G, Pasini P, Daunert S. Engineered cells as biosensing systems in biomedical analysis. Anal Bioanal Chem 2012; 402:3147-59. [PMID: 22311427 DOI: 10.1007/s00216-012-5756-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/07/2012] [Accepted: 01/16/2012] [Indexed: 01/08/2023]
Abstract
Over the past two decades there have been great advances in biotechnology, including use of nucleic acids, proteins, and whole cells to develop a variety of molecular analytical tools for diagnostic, screening, and pharmaceutical applications. Through manipulation of bacterial plasmids and genomes, bacterial whole-cell sensing systems have been engineered that can serve as novel methods for analyte detection and characterization, and as more efficient and cost-effective alternatives to traditional analytical techniques. Bacterial cell-based sensing systems are typically sensitive, specific and selective, rapid, easy to use, low-cost, and amenable to multiplexing, high-throughput, and miniaturization for incorporation into portable devices. This critical review is intended to provide an overview of available bacterial whole-cell sensing systems for assessment of a variety of clinically relevant analytes. Specifically, we examine whole-cell sensing systems for detection of bacterial quorum sensing molecules, organic and inorganic toxic compounds, and drugs, and for screening of antibacterial compounds for identification of their mechanisms of action. Methods used in the design and development of whole-cell sensing systems are also reviewed.
Collapse
Affiliation(s)
- Nilesh Raut
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | | | | | | |
Collapse
|
20
|
|
21
|
Barker CA, Farha MA, Brown ED. Chemical Genomic Approaches to Study Model Microbes. ACTA ACUST UNITED AC 2010; 17:624-32. [DOI: 10.1016/j.chembiol.2010.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 05/05/2010] [Accepted: 05/06/2010] [Indexed: 12/15/2022]
|
22
|
Ye BC, Zhang Y, Yu H, Yu WB, Liu BH, Yin BC, Yin CY, Li YY, Chu J, Zhang SL. Time-resolved transcriptome analysis of Bacillus subtilis responding to valine, glutamate, and glutamine. PLoS One 2009; 4:e7073. [PMID: 19763274 PMCID: PMC2743287 DOI: 10.1371/journal.pone.0007073] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Accepted: 08/21/2009] [Indexed: 12/19/2022] Open
Abstract
Microorganisms can restructure their transcriptional output to adapt to environmental conditions by sensing endogenous metabolite pools. In this paper, an Agilent customized microarray representing 4,106 genes was used to study temporal transcript profiles of Bacillus subtilis in response to valine, glutamate and glutamine pulses over 24 h. A total of 673, 835, and 1135 amino-acid-regulated genes were identified having significantly changed expression at one or more time points in response to valine, glutamate, and glutamine, respectively, including genes involved in cell wall, cellular import, metabolism of amino-acids and nucleotides, transcriptional regulation, flagellar motility, chemotaxis, phage proteins, sporulation, and many genes of unknown function. Different amino acid treatments were compared in terms of both the global temporal profiles and the 5-minute quick regulations, and between-experiment differential genes were identified. The highlighted genes were analyzed based on diverse sources of gene functions using a variety of computational tools, including T-profiler analysis, and hierarchical clustering. The results revealed the common and distinct modes of action of these three amino acids, and should help to elucidate the specific signaling mechanism of each amino acid as an effector.
Collapse
Affiliation(s)
- Bang-Ce Ye
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kocabaş P, Çalık P, Çalık G, Özdamar TH. Microarray Studies inBacillus subtilis. Biotechnol J 2009; 4:1012-27. [DOI: 10.1002/biot.200800330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
24
|
|
25
|
Gupta N, Singh B. Decipheringkasoperon locus inMycobacterium aurumand genesis of a recombinant strain for rational-based drug screening. J Appl Microbiol 2008; 105:1703-10. [DOI: 10.1111/j.1365-2672.2008.03888.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
De Pascale G, Grigoriadou C, Losi D, Ciciliato I, Sosio M, Donadio S. Validation for high-throughput screening of a VanRS-based reporter gene assay for bacterial cell wall inhibitors. J Appl Microbiol 2008; 103:133-40. [PMID: 17584459 DOI: 10.1111/j.1365-2672.2006.03231.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIMS The present study was undertaken to validate, for antibiotic discovery, a reporter gene assay based on a Bacillus subtilis strain expressing the Enterococcusfaecium vanRS genes and a vanH-lacZ fusion, which produced beta-galactosidase activity in the presence of cell wall inhibitors (CWI) and lysozyme. METHODS AND RESULTS The reporter assay was miniaturized, automated and validated with antibiotics and tested against portions of chemical and microbial extract libraries. The assay is simple, fast and reproducible and can detect all CWI, sometimes at concentrations lower than those necessary to inhibit bacterial growth. However, some membrane-interfering compounds also generate comparable signals. While most CWI elicit a signal that is transcription-dependent and abolished in an osmoprotective medium, transcription is not required for beta-galactosidase activity brought about by the membrane-interfering compounds. CONCLUSIONS At least two distinct mechanisms appear to lead to enzymatic activity in the reporter strain. Effective counterscreens can be designed to discard the undesired classes of compounds. SIGNIFICANCE AND IMPACT OF THE STUDY Extensive validation is required before introducing a reporter assay in high-throughput screening. However, the ease of operation and manipulation makes the reporter assays powerful tools for antibiotic discovery.
Collapse
|
27
|
Walsh TA. The emerging field of chemical genetics: potential applications for pesticide discovery. PEST MANAGEMENT SCIENCE 2007; 63:1165-71. [PMID: 17912687 DOI: 10.1002/ps.1452] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The use of small molecules to probe biological systems, generally described as 'chemical genetics', has grown considerably in the past 7 years, especially in areas related to human biology and therapeutics. This review describes some aspects of chemical genetics technologies that can be usefully applied to pesticide target discovery and lead generation. The chemical genetics approach (consisting of a phenotype screen, a chemical library and a robust target identification methodology) is compared with conventional and target-based screening. The outcomes of a chemical genetics approach are novel protein targets coupled with in vivo-active chemical ligands. The 'chemistry-first' paradigm of the chemical genetics approach can circumvent some of the obstacles that have emerged for the exploitation of novel but chemically unvalidated targets identified from genetic or genomic screens. Some of the advantages and challenges in using chemical genetics approaches are reviewed.
Collapse
Affiliation(s)
- Terence A Walsh
- Dow AgroSciences, 9330 Zionsville Road, Indianapolis, IN 46268, USA.
| |
Collapse
|
28
|
Urban A, Eckermann S, Fast B, Metzger S, Gehling M, Ziegelbauer K, Rübsamen-Waigmann H, Freiberg C. Novel whole-cell antibiotic biosensors for compound discovery. Appl Environ Microbiol 2007; 73:6436-43. [PMID: 17720843 PMCID: PMC2075059 DOI: 10.1128/aem.00586-07] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cells containing reporters which are specifically induced via selected promoters are used in pharmaceutical drug discovery and in environmental biology. They are used in screening for novel drug candidates and in the detection of bioactive compounds in environmental samples. In this study, we generated and validated a set of five Bacillus subtilis promoters fused to the firefly luciferase reporter gene suitable for cell-based screening, enabling the as yet most-comprehensive high-throughput diagnosis of antibiotic interference in the major biosynthetic pathways of bacteria: the biosynthesis of DNA by the yorB promoter, of RNA by the yvgS promoter, of proteins by the yheI promoter, of the cell wall by the ypuA promoter, and of fatty acids by the fabHB promoter. The reporter cells mainly represent novel antibiotic biosensors compatible with high-throughput screening. We validated the strains by developing screens with a set of 14,000 pure natural products, representing a source of highly diverse chemical entities, many of them with antibiotic activity (6% with anti-Bacillus subtilis activity of </=25 mug/ml]). Our screening approach is exemplified by the discovery of classical and novel DNA synthesis and translation inhibitors. For instance, we show that the mechanistically underexplored antibiotic ferrimycin A1 selectively inhibits protein biosynthesis.
Collapse
Affiliation(s)
- Andreas Urban
- Pharma Research & Development, Discovery Europe, Bayer HealthCare AG, D-42096 Wuppertal, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Affiliation(s)
- Michael F Loughlin
- The University of Nottingham, Division of Food Sciences, School of Biosciences, Sutton Bonnington Campus, Loughborough, Leicestershire, LE12 5RD, UK ;
| |
Collapse
|
30
|
von Nussbaum F, Brands M, Hinzen B, Weigand S, Häbich D. Antibacterial natural products in medicinal chemistry--exodus or revival? Angew Chem Int Ed Engl 2007; 45:5072-129. [PMID: 16881035 DOI: 10.1002/anie.200600350] [Citation(s) in RCA: 467] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
To create a drug, nature's blueprints often have to be improved through semisynthesis or total synthesis (chemical postevolution). Selected contributions from industrial and academic groups highlight the arduous but rewarding path from natural products to drugs. Principle modification types for natural products are discussed herein, such as decoration, substitution, and degradation. The biological, chemical, and socioeconomic environments of antibacterial research are dealt with in context. Natural products, many from soil organisms, have provided the majority of lead structures for marketed anti-infectives. Surprisingly, numerous "old" classes of antibacterial natural products have never been intensively explored by medicinal chemists. Nevertheless, research on antibacterial natural products is flagging. Apparently, the "old fashioned" natural products no longer fit into modern drug discovery. The handling of natural products is cumbersome, requiring nonstandardized workflows and extended timelines. Revisiting natural products with modern chemistry and target-finding tools from biology (reversed genomics) is one option for their revival.
Collapse
Affiliation(s)
- Franz von Nussbaum
- Bayer HealthCare AG, Medicinal Chemistry Europe, 42096 Wuppertal, Germany.
| | | | | | | | | |
Collapse
|
31
|
Fischer HP, Freiberg C. Applications of transcriptional profiling in antibiotics discovery and development. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2007; 64:21, 23-47. [PMID: 17195470 DOI: 10.1007/978-3-7643-7567-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
This chapter will review specific applications of microarray technology and related data analysis strategies in antibacterial research and development. We present examples of microarray applications spanning the entire antibiotics research and development pipeline, from target discovery, assay development, pharmacological evaluation, to compound safety studies. This review emphasizes the utility of microarrays for a systematic evaluation of novel chemistry as antibiotic agents. Transcriptional profiling has revolutionized the process of target elucidation and has the potential to offer substantial guidance in the identification of new targets. Microarrays will continue to be a workhorse of anti-infectives discovery programs ranging from efficacy assessments of antibiotics ('forward pharmacology') to drug safety evaluations ('toxicogenomics').
Collapse
|
32
|
Freiberg C, Brunner N, Macko L, Fischer HP. Discovering Antibiotic Efficacy Biomarkers. Mol Cell Proteomics 2006; 5:2326-35. [PMID: 16940496 DOI: 10.1074/mcp.m600127-mcp200] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
As current antibiotic therapy is increasingly challenged by emerging drug-resistant bacteria, new technologies are required to identify and develop novel classes of antibiotics. A major bottleneck in today's discovery efforts, however, is a lack of an efficient and standardized method for assaying the efficacy of a drug candidate. We propose a new high content screening approach for identifying efficacious molecules suitable for development of antibiotics. Key to our approach is a new microarray-based efficacy biomarker discovery strategy. We first produced a large dataset of transcriptional responses of Bacillus subtilis to numerous structurally diverse antibacterial drugs. Second we evaluated different protocols to optimize drug concentration and exposure time selection for profiling compounds of unknown mechanism. Finally we identified a surprisingly low number of gene transcripts (approximately 130) that were sufficient for identifying the mechanism of novel substances with reasonable accuracy (approximately 90%). We show that the statistics-based approach reveals a physiologically meaningful set of biomarkers that can be related to major bacterial defense mechanisms against antibiotics. We provide statistical evidence that a parallel measurement of the expression of the biomarkers guarantees optimal performance when using expression systems for screening libraries of novel substances. The general approach is also applicable to drug discovery for medical indications other than infectious diseases.
Collapse
Affiliation(s)
- Christoph Freiberg
- Pharma Global Drug Discovery, European Research Center, Bayer HealthCare AG, D-42096 Wuppertal, Germany
| | | | | | | |
Collapse
|
33
|
von Nussbaum F, Brands M, Hinzen B, Weigand S, Häbich D. Antibakterielle Naturstoffe in der medizinischen Chemie – Exodus oder Renaissance? Angew Chem Int Ed Engl 2006. [DOI: 10.1002/ange.200600350] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
34
|
Abstract
The ribosome is one of the main antibiotic targets in the cell. Recent years brought important insights into the mode of interaction of antibiotics with the ribosome and mechanisms of antibiotic action. Ribosome crystallography provided a detailed view of the interactions between antibiotics and rRNA. Advances in biochemical techniques let us better understand how the binding of small organic molecules can interfere with functions of an enzyme four orders of magnitude larger than the inhibitor. These and other achievements paved the way for the development of new ribosome-targeting antibiotics, some of which have already entered medical practice. The recent progress, problems and new directions of research of ribosome-targeting antibiotics are discussed in this review.
Collapse
Affiliation(s)
- Tanel Tenson
- Institute of Technology, University of Tartu, Estonia.
| | | |
Collapse
|
35
|
Mills SD. When will the genomics investment pay off for antibacterial discovery? Biochem Pharmacol 2006; 71:1096-102. [PMID: 16387281 DOI: 10.1016/j.bcp.2005.11.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Revised: 11/17/2005] [Accepted: 11/28/2005] [Indexed: 12/11/2022]
Abstract
Effective solutions to antibacterial resistance are among the key unmet medical needs driving the antibacterial industry. A major thrust in a number of companies is the development of agents with new modes of action in order to bypass the increasing emergence of antibacterial resistance. However, few antibacterials marketed in the last 30 years have novel modes of action. Most recently, genomics and target-based screening technologies have been emphasized as a means to facilitate this and expedite the antibacterial discovery process. And although no new antibacterials have yet been marketed as result of these technologies, genomics has delivered well-validated novel bacterial targets as well as a host of genetic approaches to support the antibacterial discovery process. Likewise, high throughput screening technologies have delivered the capacity to perform robust screenings of large compound collections to identify target inhibitors for lead generation. One of the principal challenges still facing antibacterial discovery is to become proficient at optimizing target inhibitors into broad-spectrum antibacterials with appropriate in vivo properties. Genomics-based technologies clearly have the potential for additional application throughout the discovery process especially in the areas of structural biology and safety assessment.
Collapse
Affiliation(s)
- Scott D Mills
- Infection Discovery, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, MA 02451 USA.
| |
Collapse
|
36
|
Lerner CG, Kakavas SJ, Wagner C, Chang RT, Merta PJ, Ruan X, Metzger RE, Beutel BA. Novel approach to mapping of resistance mutations in whole genomes by using restriction enzyme modulation of transformation efficiency. Antimicrob Agents Chemother 2005; 49:2767-77. [PMID: 15980348 PMCID: PMC1168657 DOI: 10.1128/aac.49.7.2767-2777.2005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Restriction enzyme modulation of transformation efficiencies (REMOTE) is a method that makes use of genome restriction maps and experimentally observed differences in transformation efficiencies of genomic DNA restriction digests to discover the location of mutations in genomes. The frequency with which digested genomic DNA from a resistant strain transforms a susceptible strain to resistance is primarily determined by the size of the fragment containing the resistance mutation and the distance of the mutation to the end of the fragment. The positions of restriction enzyme cleavage sites immediately flanking the resistance mutation define these parameters. The mapping procedure involves a process of elimination in which digests that transform with high frequency indicate that the restriction enzyme cleavage sites are relatively far away from the mutation, while digests that transform with low frequency indicate that the sites are close to the mutation. The transformation data are compared computationally to the genome restriction map to identify the regions that best fit the data. Transformations with PCR amplicons encompassing candidate regions identify the resistance locus and enable identification of the mutation. REMOTE was developed using Haemophilus influenzae strains with mutations in gyrA, gyrB, and rpsE that confer resistance to ciprofloxacin, novobiocin, and spectinomycin, respectively. We applied REMOTE to identify mutations that confer resistance to two novel antibacterial compounds. The resistance mutations were found in genes that can decrease the intracellular concentration of compounds: acrB, which encodes a subunit of the AcrAB-TolC efflux pump; and fadL, which encodes a long-chain fatty acid transporter.
Collapse
Affiliation(s)
- Claude G Lerner
- Global Pharmaceutical Research Division, Abbott Laboratories, R4CC, AP10-1, 100 Abbott Park Road, Abbott Park, Illinois 60064, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
O'Neill AJ, Chopra I. Preclinical evaluation of novel antibacterial agents by microbiological and molecular techniques. Expert Opin Investig Drugs 2005; 13:1045-63. [PMID: 15268641 DOI: 10.1517/13543784.13.8.1045] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The defining property of an antibacterial agent is its ability to selectively interfere with bacterial growth and/or survival. Consequently, a considerable and crucial part of the preclinical evaluation of any novel antibacterial drug involves judging and characterising its effects on bacteria in vitro. These critical stages in drug development are sometimes made to appear somewhat trivial, sandwiched as they are between the highly demanding antibacterial discovery process and the formidable task of demonstrating safety and efficacy in vivo. However, careful biological evaluation in vitro is key to quantifying and understanding the basis of the antibacterial activity, providing preliminary indications and evaluations of therapeutic potential, assessing the likelihood for the development of bacterial resistance, guiding chemical refinement and assisting subsequent stages of the appraisal of any new antibacterial drug. This review covers concepts in, and strategies for, the in vitro microbiological and molecular evaluation of antibacterial drug candidates.
Collapse
Affiliation(s)
- Alex J O'Neill
- Antimicrobial Research Centre, School of Biochemistry and Microbiology, University of Leeds, Leeds, LS2 9JT, UK.
| | | |
Collapse
|
38
|
Bandow JE. Proteomic approaches to antibiotic drug discovery. CURRENT PROTOCOLS IN MICROBIOLOGY 2005; Chapter 1:Unit 1F.2. [PMID: 18770548 DOI: 10.1002/9780471729259.mc01f02s00] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The global analysis of changes in the protein composition of bacterial cells in response to treatment with antibiotic agents grants insight into the physiological response of cells to inhibition of vital cellular functions. This unit gives an overview of how these global proteomic studies can impact antibacterial drug discovery by identifying or validating compound mechanism of action and by increasing the confidence in the value of genes with unknown function as potential new targets. It describes the design and function of a reference compendium of proteomic responses to inhibition of vital cellular functions through antibacterial agents or genetic down-regulation of potential target genes. An overview of the workflow for two-dimensional gel electrophoresis-based experiments is also presented.
Collapse
|
39
|
Abstract
Antibacterial drug discovery has experienced a paradigm shift from phenotypic screening for antibacterial activity to rational inhibition of preselected targets. Functional genomics techniques are implemented at various stages of the early drug discovery process and play a central role in target validation and mode of action determination. The spectrum of methods ranges from genetic manipulations (e.g. knockout studies, mutation analyses and the construction of conditional mutants) to transcriptome and proteome expression profiling. Functional genomics supports antibacterial drug discovery by improving knowledge on gene function, bacterial physiology and virulence and the effects of antibiotics on bacterial metabolism.
Collapse
|
40
|
Brötz-Oesterhelt H, Bandow JE, Labischinski H. Bacterial proteomics and its role in antibacterial drug discovery. MASS SPECTROMETRY REVIEWS 2005; 24:549-565. [PMID: 15389844 DOI: 10.1002/mas.20030] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Gene-expression profiling technologies in general, and proteomic technologies in particular have proven extremely useful to study the physiological response of bacterial cells to various environmental stress conditions. Complex protein toolkits coordinated by sophisticated regulatory networks have evolved to accommodate bacterial survival under ever-present stress conditions such as varying temperatures, nutrient availability, or antibiotics produced by other microorganisms that compete for habitat. In the last decades, application of man-made antibacterial agents resulted in additional bacterial exposure to antibiotic stress. Whereas the targeted use of antibiotics has remarkably reduced human suffering from infectious diseases, the ever-increasing emergence of bacteria that are resistant to antibiotics has led to an urgent need for novel antibiotic strategies. The intent of this review is to present an overview of the major achievements of proteomic approaches to study adaptation networks that are crucial for bacterial survival with a special emphasis on the stress induced by antibiotic treatment. A further focus will be the review of the, so far few, published efforts to exploit the knowledge derived from bacterial proteomic studies directly for the antibacterial drug-discovery process.
Collapse
|
41
|
Freiberg C, Fischer HP, Brunner NA. Discovering the mechanism of action of novel antibacterial agents through transcriptional profiling of conditional mutants. Antimicrob Agents Chemother 2005; 49:749-59. [PMID: 15673760 PMCID: PMC547252 DOI: 10.1128/aac.49.2.749-759.2005] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We present a new strategy for predicting novel antibiotic mechanisms of action based on the analysis of whole-genome microarray data. We first built up a reference compendium of Bacillus subtilis expression profiles induced by 14 different antibiotics. This data set was expanded by adding expression profiles from mutants that showed downregulation of genes coding for proven or emerging antibacterial targets. Here, we investigate conditional mutants underexpressing ileS, pheST, fabF, and accDA, each of which is essential for growth. Our proof-of-principle analyses reveal that conditional mutants can be used to mimic chemical inhibition of the corresponding gene products. Moreover, we show that a statistical data analysis combined with thorough pathway and regulon analysis can pinpoint the molecular target of uncharacterized antibiotics. We apply this approach to two novel antibiotics: a recently published phenyl-thiazolylurea derivative and the natural product moiramide B. Our results support recent findings suggesting that the phenyl-thiazolylurea derivative is a novel phenylalanyl-tRNA synthetase inhibitor. Finally, we propose a completely novel antibiotic mechanism of action for moiramide B based on inhibition of the bacterial acetyl coenzyme A carboxylase.
Collapse
MESH Headings
- Algorithms
- Amides/pharmacology
- Anti-Bacterial Agents/pharmacology
- Bacillus subtilis/drug effects
- Bacillus subtilis/genetics
- Bacteria/drug effects
- Bacteria/genetics
- DNA, Bacterial/biosynthesis
- DNA, Bacterial/genetics
- DNA, Complementary/biosynthesis
- DNA, Complementary/genetics
- Escherichia coli/drug effects
- Escherichia coli/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Bacterial/drug effects
- Gene Expression Regulation, Bacterial/genetics
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Isoenzymes/biosynthesis
- Isoenzymes/genetics
- Microbial Sensitivity Tests
- Models, Statistical
- Mutation/genetics
- Mutation/physiology
- Oligonucleotide Array Sequence Analysis
- RNA, Bacterial/biosynthesis
- RNA, Bacterial/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Regulon/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Statistics, Nonparametric
- Succinimides/pharmacology
Collapse
Affiliation(s)
- C Freiberg
- Bayer HealthCare AG, Pharma Research, 42096 Wuppertal, Germany
| | | | | |
Collapse
|
42
|
Li X, Zolli-Juran M, Cechetto JD, Daigle DM, Wright GD, Brown ED. Multicopy suppressors for novel antibacterial compounds reveal targets and drug efflux susceptibility. ACTA ACUST UNITED AC 2005; 11:1423-30. [PMID: 15489169 DOI: 10.1016/j.chembiol.2004.08.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Revised: 08/02/2004] [Accepted: 08/09/2004] [Indexed: 11/28/2022]
Abstract
Gene dosage has frequently been exploited to select for genetic interactions between a particular mutant and clones from a random genomic library at high copy. We report here the first use of multicopy suppression as a forward genetic method to determine cellular targets and potential resistance mechanisms for novel antibacterial compounds identified through high-throughput screening. A screen of 8640 small molecules for growth inhibition of a hyperpermeable strain of Escherichia coli led to the identification of 49 leads for suppressor selection from clones harboring an E. coli genomic library. The majority of suppressors were found to encode the multidrug efflux pump AcrB, indicating that those compounds were substrates for efflux. Two leads, which produced clones containing the gene folA, encoding dihydrofolate reductase (DHFR), proved to target DHFR in vivo and were competitive inhibitors in vitro.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Biochemistry, Antimicrobial Research Centre, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Brown ED, Wright GD. New Targets and Screening Approaches in Antimicrobial Drug Discovery. Chem Rev 2005; 105:759-74. [PMID: 15700964 DOI: 10.1021/cr030116o] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Eric D Brown
- Antimicrobial Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada L8N 3Z5
| | | |
Collapse
|
44
|
Fischer HP. Towards quantitative biology: integration of biological information to elucidate disease pathways and to guide drug discovery. BIOTECHNOLOGY ANNUAL REVIEW 2005; 11:1-68. [PMID: 16216773 DOI: 10.1016/s1387-2656(05)11001-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Developing a new drug is a tedious and expensive undertaking. The recently developed high-throughput experimental technologies, summarised by the terms genomics, transcriptomics, proteomics and metabolomics provide for the first time ever the means to comprehensively monitor the molecular level of disease processes. The "-omics" technologies facilitate the systematic characterisation of a drug target's physiology, thereby helping to reduce the typically high attrition rates in discovery projects, and improving the overall efficiency of pharmaceutical research processes. Currently, the bottleneck for taking full advantage of the new experimental technologies are the rapidly growing volumes of automatically produced biological data. A lack of scalable database systems and computational tools for target discovery has been recognised as a major hurdle. In this review, an overview will be given on recent progress in computational biology that has an impact on drug discovery applications. The focus will be on novel in silico methods to reconstruct regulatory networks, signalling cascades, and metabolic pathways, with an emphasis on comparative genomics and microarray-based approaches. Promising methods, such as the mathematical simulation of pathway dynamics are discussed in the context of applications in discovery projects. The review concludes by exemplifying concrete data-driven studies in pharmaceutical research that demonstrate the value of integrated computational systems for drug target identification and validation, screening assay development, as well as drug candidate efficacy and toxicity evaluations.
Collapse
|
45
|
Chan PF, Holmes DJ, Payne DJ. Finding the gems using genomic discovery: antibacterial drug discovery strategies – the successes and the challenges. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.ddstr.2004.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Ren D, Bedzyk LA, Setlow P, England DF, Kjelleberg S, Thomas SM, Ye RW, Wood TK. Differential gene expression to investigate the effect of (5Z)-4-bromo- 5-(bromomethylene)-3-butyl-2(5H)-furanone on Bacillus subtilis. Appl Environ Microbiol 2004; 70:4941-9. [PMID: 15294834 PMCID: PMC492336 DOI: 10.1128/aem.70.8.4941-4949.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
(5Z)-4-Bromo-5-(bromomethylene)-3-butyl-2(5H)-furanone (furanone) from the red marine alga Delisea pulchra was found previously to inhibit the growth, swarming, and biofilm formation of gram-positive bacteria. Using the gram-positive bacterium Bacillus subtilis as a test organism, we observed cell killing by 20 microg of furanone per ml, while 5 microg of furanone per ml inhibited growth approximately twofold without killing the cells. To discover the mechanism of this inhibition on a genetic level and to investigate furanone as a novel antibiotic, full-genome DNA microarrays were used to analyze the gene expression profiles of B. subtilis grown with and without 5 microg of furanone per ml. This agent induced 92 genes more than fivefold (P < 0.05) and repressed 15 genes more than fivefold (P < 0.05). The induced genes include genes involved in stress responses (such as the class III heat shock genes clpC, clpE, and ctsR and the class I heat shock genes groES, but no class II or IV heat shock genes), fatty acid biosynthesis, lichenan degradation, transport, and metabolism, as well as 59 genes with unknown functions. The microarray results for four genes were confirmed by RNA dot blotting. Mutation of a stress response gene, clpC, caused B. subtilis to be much more sensitive to 5 microg of furanone per ml (there was no growth in 8 h, while the wild-type strain grew to the stationary phase in 8 h) and confirmed the importance of the induction of this gene as identified by the microarray analysis.
Collapse
Affiliation(s)
- Dacheng Ren
- Department of Chemical Engineering, University of Connecticut, 191 Auditorium Rd., U-3222, Storrs, CT 06269-3222, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
High-throughput screening (HTS), systematically testing thousands of small molecules to find candidates for lead optimization, primarily involves exposure of purified proteins to arrayed collections of small molecules. More complex phenotypic assays, such as cell-based or whole-organism assays, traditionally have flanked HTS, preceding it to validate new therapeutic targets, and following it to characterize new lead compounds in cellular contexts. Recently, however, cell- and organism-based phenotypic assays have increasingly been adopted as a primary screening platform for annotating small molecules.
Collapse
Affiliation(s)
- Paul A Clemons
- Initiative for Chemical Genetics, ICCB-Broad Institute, Harvard University, 320 Charles Street, Room 184, Cambridge, Massachusetts 02141, USA.
| |
Collapse
|