1
|
Campillo R, García-Penas I, López N, Sánchez A, Fau A, Gómez D, Berdejo D, García-Gonzalo D, Pagán R. Ciprofloxacin-resistant Salmonella Typhimurium demonstrates cross-tolerance to heat treatments in liquid food matrices. Food Res Int 2025; 210:116330. [PMID: 40306802 DOI: 10.1016/j.foodres.2025.116330] [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: 01/11/2025] [Revised: 02/28/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
The alarming occurrence of antimicrobial resistance (AMR) in human bacterial isolates indicates that prevention and control protocols are not adequately managing this global threat. The agri-food chain plays a noteworthy role in the dissemination of AMR via the handling and consumption of contaminated food products. However, it remains unclear whether acquisition of AMR in bacteria might indirectly enhance bacterial tolerance to food preservation methods (i.e., cross-tolerance), resulting in defective pathogen reduction. In this study, five ciprofloxacin (CIP) resistant variants (RVs) were generated after exposing Salmonella Typhimurium LT2 (SeT) to an upward CIP gradient. We thereupon observed up to 125-fold increases in the minimum inhibitory concentration to CIP in all five RVs. Moreover, two RVs showed reduced sensitivity to heat in laboratory media compared to SeT. The most tolerant strain displayed mutations in genes previously implicated in AMR, coding for RNA polymerase subunits (rpoD), regulatory protein RamR (ramR) and enzyme adenylate cyclase (cyaA). Validation in liquid food matrices revealed enhanced thermotolerance of the RV to treatments performed at 50 °C in orange juice (×986.7 survival risk after 15 min of treatment), and 54 °C in milk (more than ×10,000 survival risk after 30 min) and liquid-whole egg (×976.7 survival risk after 40 min). Furthermore, virulence assays in nematode Caenorhabditis elegans showed mutations conferring AMR and cross-tolerance did not result in a substantial loss of pathogenicity. Hence, exposures to CIP might lead to the selection of S. Typhimurium variants that pose limits to heat treatment efficacy, thereby increasing their survival risk and ultimately allowing them to reach the end consumer - thus also limiting the scope of antibiotic action during eventual infection.
Collapse
Affiliation(s)
- Raúl Campillo
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Ivo García-Penas
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Noelia López
- CNTA, Centro Nacional de Tecnología y Seguridad Alimentaria, San Adrián, Spain
| | - Ana Sánchez
- CNTA, Centro Nacional de Tecnología y Seguridad Alimentaria, San Adrián, Spain
| | - Alberto Fau
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Diego Gómez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Daniel Berdejo
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Diego García-Gonzalo
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain
| | - Rafael Pagán
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Zaragoza, Spain.
| |
Collapse
|
2
|
Kerek Á, Román I, Szabó Á, Kovács D, Kardos G, Kovács L, Jerzsele Á. Antibiotic resistance genes in Escherichia coli - literature review. Crit Rev Microbiol 2025:1-35. [PMID: 40249005 DOI: 10.1080/1040841x.2025.2492156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Antimicrobial resistance threatens humans and animals worldwide and is recognized as one of the leading global public health issues. Escherichia coli (E. coli) has an unquestionable role in carrying and transmitting antibiotic resistance genes (ARGs), which in many cases are encoded on plasmids or phage, thus creating the potential for horizontal gene transfer. In this literature review, the authors summarize the major antibiotic resistance genes occurring in E. coli bacteria, through the major antibiotic classes. The aim was not only listing the resistance genes against the clinically relevant antibiotics, used in the treatment of E. coli infections, but also to cover the entire resistance gene carriage in E. coli, providing a more complete picture. We started with the long-standing antibiotic groups (beta-lactams, aminoglycosides, tetracyclines, sulfonamides and diaminopyrimidines), then moved toward the newer groups (phenicols, peptides, fluoroquinolones, nitrofurans and nitroimidazoles), and in every group we summarized the resistance genes grouped by the mechanism of their action (enzymatic inactivation, antibiotic efflux, reduced permeability, etc.). We observed that the frequency of antibiotic resistance mechanisms changes in the different groups.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Dóra Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Gábor Kardos
- One Health Institute, University of Debrecen, Debrecen, Hungary
- National Public Health Center, Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Nyíregyháza, Hungary
| | - László Kovács
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
- Department of Animal Hygiene, Herd Health and Mobile Clinic, University of Veterinary Medicine, Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, Budapest, Hungary
| |
Collapse
|
3
|
Kerek Á, Román I, Szabó Á, Pézsa NP, Jerzsele Á. Antibiotic Resistance Gene Expression in Veterinary Probiotics: Two Sides of the Coin. Vet Sci 2025; 12:217. [PMID: 40266902 PMCID: PMC11945515 DOI: 10.3390/vetsci12030217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 04/25/2025] Open
Abstract
The rapid proliferation of antimicrobial resistance has emerged as one of the most pressing animal and public health challenges of our time. Probiotics, extensively employed in human and veterinary medicine, are instrumental in maintaining a balanced microbiome and mitigating its disruption during antibiotic therapy. While their numerous benefits are well documented, probiotics also present potential risks, notably the capacity to harbor antimicrobial resistance genes. This genetic reservoir could contribute to the emergence and spread of antimicrobial resistance by facilitating the horizontal transfer of resistance genes to pathogenic bacteria within the gut. This review critically examines the presence of antimicrobial resistance genes in commonly used probiotic strains, explores the underlying mechanisms of resistance, and provides a balanced analysis of the benefits and risks associated with their use. By addressing these dual aspects, this paper highlights the need for vigilant evaluation of probiotics to preserve their therapeutic potential while minimizing public health risks.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| |
Collapse
|
4
|
Herrera-Espejo S, Rubio A, Ceballos-Romero L, Pachón J, Cordero E, Pérez-Pulido AJ, Pachón-Ibáñez ME. Detection of Possible Resistance Mechanisms in Uropathogenic Escherichia coli Strains Isolated from Kidney Transplant Recipients Based on Whole Genome Sequencing. Biomolecules 2025; 15:260. [PMID: 40001563 PMCID: PMC11853403 DOI: 10.3390/biom15020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Urinary tract infections are a global health concern, with uropathogenic Escherichia coli (UPEC) accounting for 80-90% of cases. Given the rise in antimicrobial resistance, our aim was to elucidate the genetic mechanisms behind low-level resistance to ciprofloxacin and fosfomycin (LLCR and LLFR) in UPEC strains, using whole-genome sequencing (WGS) to identify point mutations in chromosomal and plasmid genes. METHODS A cohort UPEC was collected from kidney transplant recipients at the Virgen del Rocío University Hospital, Spain. Minimum inhibitory concentrations were determined for ciprofloxacin and fosfomycin to categorize strains into LLCR and LLFR. Twenty strains were selected for WGS, with genome annotations. Point mutations were identified and analyzed using alignment tools, and protein stability changes were predicted. RESULTS LLCR strains exhibited mutations in key quinolone resistance-determining regions of the gyrA gene, in 83% of cases. The qnrS1 plasmid gene was found in 17% of LLCR strains. LLFR strains showed mutations in the glpT and cyaA genes. Mutations in the uhp gene family were linked to the fosfomycin-resistant phenotype, suggesting a multi-step resistance evolution mechanism. CONCLUSIONS This study highlights the complex interplay between chromosomal and plasmid genes in UPEC's resistance to ciprofloxacin and fosfomycin. The findings contribute to understanding low-level resistance mechanisms and may guide the development of novel therapeutic strategies to combat multidrug-resistant strains.
Collapse
Affiliation(s)
- Soraya Herrera-Espejo
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (L.C.-R.); (M.E.P.-I.)
| | - Alejandro Rubio
- Andalusian Centre for Developmental Biology (CABD, UPO-CSIC-JA), Faculty of Experimental Sciences (Genetics Area), University Pablo de Olavide, 41013 Seville, Spain;
| | - Lucía Ceballos-Romero
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (L.C.-R.); (M.E.P.-I.)
| | - Jerónimo Pachón
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain;
- Department of Medicine, School of Medicine, University of Seville, 41004 Seville, Spain
| | - Elisa Cordero
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (L.C.-R.); (M.E.P.-I.)
- Department of Medicine, School of Medicine, University of Seville, 41004 Seville, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio J. Pérez-Pulido
- Andalusian Centre for Developmental Biology (CABD, UPO-CSIC-JA), Faculty of Experimental Sciences (Genetics Area), University Pablo de Olavide, 41013 Seville, Spain;
| | - María Eugenia Pachón-Ibáñez
- Clinical Unit of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (L.C.-R.); (M.E.P.-I.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Knak T, Takada S, Illarionov B, Krisilia V, Pessanha de Carvalho L, Lungerich B, Sakamoto Y, Höfmann S, Bacher A, Kalscheuer R, Held J, Fischer M, Tanaka N, Kurz T. Expanding the Chemical Space of Reverse Fosmidomycin Analogs. ACS Med Chem Lett 2025; 16:136-143. [PMID: 39811140 PMCID: PMC11726376 DOI: 10.1021/acsmedchemlett.4c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Multidrug-resistant pathogens pose a major threat to human health, necessitating the identification of new drug targets and lead compounds that are not susceptible to cross-resistance. This study demonstrates that novel reverse thia analogs of the phosphonohydroxamic acid antibiotic fosmidomycin inhibit 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR), an essential enzyme for Plasmodium falciparum, Escherichia coli, and Mycobacterium tuberculosis that is absent in humans. Some novel analogs with large α-phenyl substituents exhibited strong inhibition across these three DXR orthologues, surpassing the inhibitory activity of fosmidomycin. Despite nanomolar target inhibition, the new DXR inhibitors demonstrated mainly weak or no in vitro growth inhibition of the pathogens. Crystallographic studies revealed that compounds 12a and 12b induce an open PfDXR conformation and that the enzyme selectively binds the S-enantiomers. The study underscores the difficulties of achieving potent cellular activity despite strong DXR inhibition and emphasizes the need for novel structural optimization strategies and comprehensive pharmacokinetic studies.
Collapse
Affiliation(s)
- Talea Knak
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sana Takada
- School
of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan
| | - Boris Illarionov
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
| | - Violetta Krisilia
- Institute
of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr.
1, 40225 Düsseldorf, Germany
| | - Lais Pessanha de Carvalho
- Institut
für Tropenmedizin, Eberhard Karls
Universität Tübingen, Wilhelmstr. 27, 72074 Tübingen, Germany
| | - Beate Lungerich
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Yasumitsu Sakamoto
- School
of
Pharmacy, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Stefan Höfmann
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Adelbert Bacher
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
- TUM
School of Natural Sciences, Technical University
of Munich, Boltzmannstr.
10, 85748 Garching, Germany
| | - Rainer Kalscheuer
- Institute
of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr.
1, 40225 Düsseldorf, Germany
| | - Jana Held
- Institut
für Tropenmedizin, Eberhard Karls
Universität Tübingen, Wilhelmstr. 27, 72074 Tübingen, Germany
- German
Center for Infection Research (DZIF),
partner site Tübingen, 72074 Tübingen, Germany
| | - Markus Fischer
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
| | - Nobutada Tanaka
- School
of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan
| | - Thomas Kurz
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| |
Collapse
|
6
|
Kesharwani S, Eeba, Tandi M, Agarwal N, Sundriyal S. Design and synthesis of non-hydroxamate lipophilic inhibitors of 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR): in silico, in vitro and antibacterial studies. RSC Adv 2024; 14:27530-27554. [PMID: 39221132 PMCID: PMC11362829 DOI: 10.1039/d4ra05083e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
1-Deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) is a key enzyme of the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway operating in several pathogens, including Mycobacterium and Plasmodium. Since a DXR homologue is not present in humans, it is an important antimicrobial target. Fosmidomycin (FSM) and its analogues inhibit DXR function by chelating the divalent metal (Mn2+ or Mg2+) in its active site via a hydroxamate metal binding group (MBG). The latter, however, enhances the polarity of molecules and is known to display metabolic instability and toxicity issues. While attempts have been made to increase the lipophilicity of FSM by substituting the linker chain and prodrug approach, very few efforts have been made to replace the hydroxamate group with other lipophilic MBGs. We report a systematic in silico and experimental investigation to identify novel MBGs for designing non-hydroxamate lipophilic DXR inhibitors. The SAR studies with selected MBG fragments identified novel inhibitors of E. Coli DXR with IC50 values ranging from 0.29 to 106 μM. The promising inhibitors were also screened against ESKAPE pathogens and M. tuberculosis.
Collapse
Affiliation(s)
- Sharyu Kesharwani
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani (BITS) Pilani Campus, Vidya Vihar, Pilani Rajasthan 333 031 India
| | - Eeba
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Mile Stone, Gurugram-Faridabad Expressway Faridabad 121001 Haryana India
| | - Mukesh Tandi
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani (BITS) Pilani Campus, Vidya Vihar, Pilani Rajasthan 333 031 India
| | - Nisheeth Agarwal
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Mile Stone, Gurugram-Faridabad Expressway Faridabad 121001 Haryana India
| | - Sandeep Sundriyal
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani (BITS) Pilani Campus, Vidya Vihar, Pilani Rajasthan 333 031 India
| |
Collapse
|
7
|
Mazzone F, Hoeppner A, Reiners J, Gertzen CG, Applegate V, Abdullaziz MA, Gottstein J, Degrandi D, Wesemann M, Kurz T, Smits SH, Pfeffer K. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase as target for anti Toxoplasma gondii agents: crystal structure, biochemical characterization and biological evaluation of inhibitors. Biochem J 2024; 481:1075-1096. [PMID: 39105673 PMCID: PMC11346426 DOI: 10.1042/bcj20240110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/07/2024]
Abstract
Toxoplasma gondii is a widely distributed apicomplexan parasite causing toxoplasmosis, a critical health issue for immunocompromised individuals and for congenitally infected foetuses. Current treatment options are limited in number and associated with severe side effects. Thus, novel anti-toxoplasma agents need to be identified and developed. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) is considered the rate-limiting enzyme in the non-mevalonate pathway for the biosynthesis of the isoprenoid precursors isopentenyl pyrophosphate and dimethylallyl pyrophosphate in the parasite, and has been previously investigated for its key role as a novel drug target in some species, encompassing Plasmodia, Mycobacteria and Escherichia coli. In this study, we present the first crystal structure of T. gondii DXR (TgDXR) in a tertiary complex with the inhibitor fosmidomycin and the cofactor NADPH in dimeric conformation at 2.5 Å resolution revealing the inhibitor binding mode. In addition, we biologically characterize reverse α-phenyl-β-thia and β-oxa fosmidomycin analogues and show that some derivatives are strong inhibitors of TgDXR which also, in contrast with fosmidomycin, inhibit the growth of T. gondii in vitro. Here, ((3,4-dichlorophenyl)((2-(hydroxy(methyl)amino)-2-oxoethyl)thio)methyl)phosphonic acid was identified as the most potent anti T. gondii compound. These findings will enable the future design and development of more potent anti-toxoplasma DXR inhibitors.
Collapse
Affiliation(s)
- Flaminia Mazzone
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| | - Astrid Hoeppner
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Jens Reiners
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Christoph G.W. Gertzen
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Violetta Applegate
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Mona A. Abdullaziz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
- National Research Centre (NRC), Dokki, Cairo, Egypt
| | - Julia Gottstein
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Degrandi
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| | - Martina Wesemann
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Sander H.J. Smits
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
8
|
Gil-Gil T, Martínez JL. Role of the phosphotransferase system in the transport of fosfomycin in Escherichia coli. Int J Antimicrob Agents 2024; 63:107027. [PMID: 37926273 DOI: 10.1016/j.ijantimicag.2023.107027] [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: 08/17/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
The inducible inner membrane transporters, UhpT and GlpT are considered to be unique fosfomycin transporters. Glucose-6-phosphate, the substrate for UhpT, enhances fosfomycin activity. Previous work indicates that the fructose phosphotransferase system (PTS) might be involved in fosfomycin transport in the bacterial species, Stenotrophomonas maltophilia. Fosfomycin transport in Escherichia coli has been extensively studied and characterised. The current paper addresses the potential fosfomycin transport activity of the fructose PTS in E. coli. Notably, the deletion of both fructose-specific and general PTS proteins in E. coli increases fosfomycin resistance, which indicates that fructose PTS is involved in fosfomycin transport in E. coli. Further, although inactivation of UhpT, the canonical fosfomycin transporter, in E. coli increases fosfomycin resistance by 2-fold, inactivation of genes encoding the PTS increases it by up to 256-fold. Moreover, intracellular accumulation declines in the absence of both transporters, being mutations in the PTS associated with a larger decline. The results presented in this paper re-open the study of fosfomycin transport and reveal the role of the PTS in the transport of this bactericidal antibiotic in E. coli.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Centro Nacional de Biotecnología, CSIC. Darwin 3. 28049, Madrid, Spain.
| | | |
Collapse
|
9
|
The Multifaceted MEP Pathway: Towards New Therapeutic Perspectives. Molecules 2023; 28:molecules28031403. [PMID: 36771066 PMCID: PMC9919496 DOI: 10.3390/molecules28031403] [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: 01/10/2023] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Isoprenoids, a diverse class of natural products, are present in all living organisms. Their two universal building blocks are synthesized via two independent pathways: the mevalonate pathway and the 2-C-methyl-ᴅ-erythritol 4-phosphate (MEP) pathway. The presence of the latter in pathogenic bacteria and its absence in humans make all its enzymes suitable targets for the development of novel antibacterial drugs. (E)-4-Hydroxy-3-methyl-but-2-enyl diphosphate (HMBPP), the last intermediate of this pathway, is a natural ligand for the human Vγ9Vδ2 T cells and the most potent natural phosphoantigen known to date. Moreover, 5-hydroxypentane-2,3-dione, a metabolite produced by Escherichia coli 1-deoxy-ᴅ-xylulose 5-phosphate synthase (DXS), the first enzyme of the MEP pathway, structurally resembles (S)-4,5-dihydroxy-2,3-pentanedione, a signal molecule implied in bacterial cell communication. In this review, we shed light on the diversity of potential uses of the MEP pathway in antibacterial therapies, starting with an overview of the antibacterials developed for each of its enzymes. Then, we provide insight into HMBPP, its synthetic analogs, and their prodrugs. Finally, we discuss the potential contribution of the MEP pathway to quorum sensing mechanisms. The MEP pathway, providing simultaneously antibacterial drug targets and potent immunostimulants, coupled with its potential role in bacterial cell-cell communication, opens new therapeutic perspectives.
Collapse
|
10
|
Over 40 Years of Fosmidomycin Drug Research: A Comprehensive Review and Future Opportunities. Pharmaceuticals (Basel) 2022; 15:ph15121553. [PMID: 36559004 PMCID: PMC9782300 DOI: 10.3390/ph15121553] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
To address the continued rise of multi-drug-resistant microorganisms, the development of novel drugs with new modes of action is urgently required. While humans biosynthesize the essential isoprenoid precursors isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP) via the established mevalonate pathway, pathogenic protozoa and certain pathogenic eubacteria use the less well-known methylerythritol phosphate pathway for this purpose. Important pathogens using the MEP pathway are, for example, Plasmodium falciparum, Mycobacterium tuberculosis, Pseudomonas aeruginosa and Escherichia coli. The enzymes of that pathway are targets for antiinfective drugs that are exempt from target-related toxicity. 2C-Methyl-D-erythritol 4-phosphate (MEP), the second enzyme of the non-mevalonate pathway, has been established as the molecular target of fosmidomycin, an antibiotic that has so far failed to be approved as an anti-infective drug. This review describes the development and anti-infective properties of a wide range of fosmidomycin derivatives synthesized over the last four decades. Here we discuss the DXR inhibitor pharmacophore, which comprises a metal-binding group, a phosphate or phosphonate moiety and a connecting linker. Furthermore, non-fosmidomycin-based DXRi, bisubstrate inhibitors and several prodrug concepts are described. A comprehensive structure-activity relationship (SAR) of nearly all inhibitor types is presented and some novel opportunities for further drug development of DXR inhibitors are discussed.
Collapse
|
11
|
Ortiz-Padilla M, Portillo-Calderón I, Velázquez-Escudero A, Rodríguez-Baño J, Pascual Á, Rodríguez-Martínez JM, Docobo-Pérez F. Effect of Glycerol on Fosfomycin Activity against Escherichia coli. Antibiotics (Basel) 2022; 11:antibiotics11111612. [PMID: 36421256 PMCID: PMC9686493 DOI: 10.3390/antibiotics11111612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Fosfomycin is an antimicrobial that inhibits the biosynthesis of peptidoglycan by entering the bacteria through two channels (UhpT and GlpT). Glycerol is clinically used as a treatment for elevated intracranial pressure and induces the expression of glpT in Escherichia coli. Glycerol might offer synergistic activity by increasing fosfomycin uptake. The present study evaluates the use of glycerol at physiological concentrations in combination with fosfomycin against a collection of isogenic mutants of fosfomycin-related genes in E. coli strains. Induction of fosfomycin transporters, susceptibility tests, interaction assays, and time-kill assays were performed. Our results support the notion that glycerol allows activation of the GlpT transporter, but this induction is delayed over time and is not homogeneous across the bacterial population, leading to contradictory results regarding the enhancement of fosfomycin activity. The susceptibility assays showed an increase in fosfomycin activity with glycerol in the disk diffusion assay but not in the agar dilution or broth microdilution assays. Similarly, in the time-kill assays, the effect of glycerol was absent by the emergence of fosfomycin-resistant subpopulations. In conclusion, glycerol may not be a good candidate for use as an adjuvant with fosfomycin.
Collapse
Affiliation(s)
- Miriam Ortiz-Padilla
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
| | - Inés Portillo-Calderón
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - Ana Velázquez-Escudero
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
| | - Jesús Rodríguez-Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Álvaro Pascual
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - José Manuel Rodríguez-Martínez
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
- Correspondence: (J.M.R.-M.); (F.D.-P.); Tel.: +34-95-455-6100 (J.M.R.-M. & F.D.-P.)
| | - Fernando Docobo-Pérez
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, 41009 Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
- Correspondence: (J.M.R.-M.); (F.D.-P.); Tel.: +34-95-455-6100 (J.M.R.-M. & F.D.-P.)
| |
Collapse
|
12
|
GAPDH mediates drug resistance and metabolism in Plasmodium falciparum malaria parasites. PLoS Pathog 2022; 18:e1010803. [PMID: 36103572 PMCID: PMC9512246 DOI: 10.1371/journal.ppat.1010803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 09/26/2022] [Accepted: 08/09/2022] [Indexed: 11/19/2022] Open
Abstract
Efforts to control the global malaria health crisis are undermined by antimalarial resistance. Identifying mechanisms of resistance will uncover the underlying biology of the Plasmodium falciparum malaria parasites that allow evasion of our most promising therapeutics and may reveal new drug targets. We utilized fosmidomycin (FSM) as a chemical inhibitor of plastidial isoprenoid biosynthesis through the methylerythritol phosphate (MEP) pathway. We have thus identified an unusual metabolic regulation scheme in the malaria parasite through the essential glycolytic enzyme, glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Two parallel genetic screens converged on independent but functionally analogous resistance alleles in GAPDH. Metabolic profiling of FSM-resistant gapdh mutant parasites indicates that neither of these mutations disrupt overall glycolytic output. While FSM-resistant GAPDH variant proteins are catalytically active, they have reduced assembly into the homotetrameric state favored by wild-type GAPDH. Disrupted oligomerization of FSM-resistant GAPDH variant proteins is accompanied by altered enzymatic cooperativity and reduced susceptibility to inhibition by free heme. Together, our data identifies a new genetic biomarker of FSM-resistance and reveals the central role of GAPDH in MEP pathway control and antimalarial sensitivity. Malaria is a life-threatening mosquito-borne infection that remains an enormous public health threat worldwide, with over 600,000 deaths reported in 2020 alone. The parasites that cause malaria invade and replicate within human red blood cells. This unique environment provides the malaria parasite with almost unlimited supply of sugar in the form of glucose, which the parasite uses for energy and as building blocks to grow and divide. Parasites break down glucose, and must use these breakdown products to make new molecules, including a very important class of compounds called isoprenoids. Malaria parasites normally die when they are treated with a drug, called fosmidomycin, that inhibits this process. To understand how parasites regulate this critical function, in this study we identified parasites that were resistant to fosmidomycin. These fosmidomycin-resistant cells had mutations in an enzyme that is critical for sugar breakdown, called glyceraldehyde phosphate dehydrogenase (GAPDH). We find that parasites with mutant GAPDH enzymes still break down sugar normally, but are not inhibited by other changes in the cell that happen upon fosmidomycin treatment. These results reveal a new and important role for the enzyme GAPDH as a control-point for downstream metabolism in malaria parasites.
Collapse
|
13
|
Oral Fosfomycin Formulation in Bacterial Prostatitis: New Role for an Old Molecule-Brief Literature Review and Clinical Considerations. Infect Dis Rep 2022; 14:621-634. [PMID: 36005269 PMCID: PMC9408554 DOI: 10.3390/idr14040067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022] Open
Abstract
Bacterial prostatitis infections are described as infections that are difficult-to-treat, due to prostate anatomic characteristics along with clinical difficulty in terms of diagnosis and management. Furthermore, the emergence of multidrug resistant (MDR) bacteria, such as extended-spectrum beta-lactamase (ESBL) producer Escherichia coli, also representing the main causative pathogen in prostatitis, poses major problems in terms of antibiotic management and favorable clinical outcome. Oral fosfomycin, an antibiotic commonly used for the treatment of uncomplicated urinary tract infections (UTIs), has been recently evaluated for the treatment of bacterial prostatitis due to its favorable pharmacokinetic profile, its activity against MDR gram-positive and gram-negative bacteria, safety profile, and multiple synergic effect with other antibiotics as well as the low resistance rate. This review addresses fosfomycin pharmacokinetics and pharmacodynamics and discusses the latest clinical evidence on its clinical use to treat acute and chronic bacterial prostatitis in hospitalized patients and in outpatients. As described in several reports, oral fosfomycin may represent a valid therapeutic option to treat susceptible germs commonly causing prostatitis, such as E. coli and other Enterobacterales as well as Enterococcus faecium, even as a first-line regimen in particular clinical settings (patients with previous treatment failure, with allergies or outpatients). Stronger data from further studies, including randomized controlled trials, would be helpful to establish the proper dosage and specific indications.
Collapse
|
14
|
Liebrenz K, Frare R, Gómez C, Pascuan C, Brambilla S, Soldini D, Maguire V, Carrio A, Ruiz O, McCormick W, Soto G, Ayub N. Multiple ways to evade the bacteriostatic action of glyphosate in rhizobia include the mutation of the conserved serine 90 of the nitrogenase subunit NifH to alanine. Res Microbiol 2022; 173:103952. [PMID: 35436545 DOI: 10.1016/j.resmic.2022.103952] [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: 01/11/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
The genome resequencing of spontaneous glyphosate-resistant mutants derived from the soybean inoculant E109 allowed identifying genes most likely associated with the uptake (gltL and cya) and metabolism (zigA and betA) of glyphosate, as well as with nitrogen fixation (nifH). Mutations in these genes reduce the lag phase and improve nodulation under glyphosate stress. In addition to providing glyphosate resistance, the amino acid exchange Ser90Ala in NifH increased the citrate synthase activity, growth rate and plant growth-promoting efficiency of E109 in the absence of glyphosate stress, suggesting roles for this site during both the free-living and symbiotic growth stages.
Collapse
Affiliation(s)
- Karen Liebrenz
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Romina Frare
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Cristina Gómez
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Cecilia Pascuan
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Silvina Brambilla
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Diego Soldini
- Estación Experimental Agropecuaria Marcos Juárez, INTA, Córdoba, Argentina
| | - Vanina Maguire
- Instituto Tecnológico Chascomús (INTECH-CONICET), Buenos Aires, Argentina
| | - Alejandro Carrio
- Estación Experimental Agropecuaria Marcos Juárez, INTA, Córdoba, Argentina
| | - Oscar Ruiz
- Instituto Tecnológico Chascomús (INTECH-CONICET), Buenos Aires, Argentina
| | - Wayne McCormick
- Ottawa Research and Development Centre (AAFC), Ottawa, ON, Canada
| | - Gabriela Soto
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina
| | - Nicolás Ayub
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Buenos Aires, Argentina; Instituto de Genética (IGEAF), Buenos Aires, Argentina.
| |
Collapse
|
15
|
Differences in fosfomycin resistance mechanisms between Pseudomonas aeruginosa and Enterobacterales. Antimicrob Agents Chemother 2021; 66:e0144621. [PMID: 34807759 DOI: 10.1128/aac.01446-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Multidrug-resistant (MDR) Pseudomonas aeruginosa presents a serious threat to public health due to its widespread resistance to numerous antibiotics. P. aeruginosa commonly causes nosocomial infections including urinary tract infections (UTI) which have become increasingly difficult to treat. The lack of effective therapeutic agents has renewed interest in fosfomycin, an old drug discovered in the 1960s and approved prior to the rigorous standards now required for drug approval. Fosfomycin has a unique structure and mechanism of action, making it a favorable therapeutic alternative for MDR pathogens that are resistant to other classes of antibiotics. The absence of susceptibility breakpoints for fosfomycin against P. aeruginosa limits its clinical use and interpretation due to extrapolation of breakpoints established for Escherichia coli or Enterobacterales without supporting evidence. Furthermore, fosfomycin use and efficacy for treatment of P. aeruginosa is also limited by both inherent and acquired resistance mechanisms. This narrative review provides an update on currently identified resistance mechanisms to fosfomycin, with a focus on those mediated by P. aeruginosa such as peptidoglycan recycling enzymes, chromosomal Fos enzymes, and transporter mutation. Additional fosfomycin resistance mechanisms exhibited by Enterobacterales including mutations in transporters and associated regulators, plasmid mediated Fos enzymes, kinases, and murA modification, are also summarized and contrasted. These data highlight that different fosfomycin resistance mechanisms may be associated with elevated MIC values in P. aeruginosa compared to Enterobacterales, emphasizing that extrapolation of E. coli breakpoints to P. aeruginosa should be avoided.
Collapse
|
16
|
Ball HS, Girma MB, Zainab M, Soojhawon I, Couch RD, Noble SM. Characterization and Inhibition of 1-Deoxy-d-Xylulose 5-Phosphate Reductoisomerase: A Promising Drug Target in Acinetobacter baumannii and Klebsiella pneumoniae. ACS Infect Dis 2021; 7:2987-2998. [PMID: 34672535 PMCID: PMC8594541 DOI: 10.1021/acsinfecdis.1c00132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
The ESKAPE pathogens
comprise a group of multidrug-resistant bacteria
that are the leading cause of nosocomial infections worldwide. The
prevalence of antibiotic resistant strains and the relative ease by
which bacteria acquire resistance genes highlight the continual need
for the development of novel antibiotics against new drug targets.
The methylerythritol phosphate (MEP) pathway is an attractive target
for the development of new antibiotics. The MEP pathway governs the
synthesis of isoprenoids, which are key lipid precursors for vital
cell components such as ubiquinone and bacterial hopanoids. Additionally,
the MEP pathway is entirely distinct from the corresponding mammalian
pathway, the mevalonic acid (MVA) pathway, making the first committed
enzyme of the MEP pathway, 1-deoxy-d-xylulose 5-phosphate
reductoisomerase (IspC), an attractive target for antibiotic development.
To facilitate drug development against two of the ESKAPE pathogens, Acinetobacter baumannii and Klebsiella
pneumoniae, we cloned, expressed, purified, and characterized
IspC from these two Gram-negative bacteria. Enzyme inhibition assays
using IspC from these two pathogens, and compounds fosmidomycin and
FR900098, indicate IC50 values ranging from 19.5–45.5
nM. Antimicrobial susceptibility tests with these inhibitors reveal
that A. baumannii is susceptible to
FR900098, whereas K. pneumoniae is
susceptible to both compounds. Finally, to facilitate structure-based
drug design of inhibitors targeting A. baumannii IspC, we determined the 2.5 Å crystal structure of IspC from A. baumannii in complex with inhibitor FR900098,
and cofactors NADPH and magnesium.
Collapse
Affiliation(s)
- Haley S. Ball
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20109, United States of America
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Springs, Maryland 20910, United States of America
| | - Misgina B. Girma
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20109, United States of America
| | - Mosufa Zainab
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20109, United States of America
| | - Iswarduth Soojhawon
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Springs, Maryland 20910, United States of America
| | - Robin D. Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20109, United States of America
| | - Schroeder M. Noble
- Wound Infections Department, Bacterial Diseases Branch, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Springs, Maryland 20910, United States of America
| |
Collapse
|
17
|
Leite GC, Perdigão-Neto LV, Ruedas Martins RC, Rizek C, Levin AS, Costa SF. Genetic factors involved in fosfomycin resistance of multidrug-resistant Acinetobacter baumannii. INFECTION GENETICS AND EVOLUTION 2021; 93:104943. [PMID: 34051359 DOI: 10.1016/j.meegid.2021.104943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022]
Abstract
The treatment of infections caused by A. baumannii is a challenge and fosfomycin has been used as a combination therapy. Moreover, data regarding the fosfomycin resistance mechanism is scarce. The goals of this study were to evaluate fosfomycin susceptibility in polyclonal multi-resistant A. baumannii isolates and characterize the fosfomycin resistance. We analyzed 32 A. baumannii isolates from a Brazilian bacterial collection, followed by their minimum inhibitory concentration (MIC), and whole-genome sequence to detect fosfomycin resistance genes. The isolates showed a fosfomycin MIC ranging from 32 to ≥256 mg/L. All isolates were negative for fosA and fosB genes, and four isolates carried the fosX gene. Two different metabolic pathways that form peptidoglycan precursors were identified. Mutations were observed in the adenylate cyclase gene. All A. baumannii isolates studied showed Val132Ala substitutions in MurA. The analysis showed different ways that may lead to the intrinsic fosfomycin-resistance of A. baumannii, such as alterations on the glycerol-3-phosphate transporter system caused by adenylate cyclase mutations; and a possible connection of cell wall recycling by different metabolic pathways.
Collapse
Affiliation(s)
- Gleice C Leite
- Department of Infectious Diseases, University of São Paulo, São Paulo, SP 05403-000, Brazil; Laboratory of Medical Investigation 49 (LIM-49), University of São Paulo, São Paulo, SP 05403-000, Brazil
| | - Lauro V Perdigão-Neto
- Department of Infectious Diseases, University of São Paulo, São Paulo, SP 05403-000, Brazil; Laboratory of Medical Investigation 49 (LIM-49), University of São Paulo, São Paulo, SP 05403-000, Brazil
| | - Roberta C Ruedas Martins
- Department of Infectious Diseases, University of São Paulo, São Paulo, SP 05403-000, Brazil; Laboratory of Medical Investigation 49 (LIM-49), University of São Paulo, São Paulo, SP 05403-000, Brazil
| | - Camila Rizek
- Laboratory of Medical Investigation 49 (LIM-49), University of São Paulo, São Paulo, SP 05403-000, Brazil
| | - Anna Sara Levin
- Department of Infectious Diseases, University of São Paulo, São Paulo, SP 05403-000, Brazil
| | - Silvia F Costa
- Department of Infectious Diseases, University of São Paulo, São Paulo, SP 05403-000, Brazil; Laboratory of Medical Investigation 49 (LIM-49), University of São Paulo, São Paulo, SP 05403-000, Brazil; Institute of Tropical Medicine, University of São Paulo, São Paulo, SP, 05403-000, Brazil.
| |
Collapse
|
18
|
Degradation of the Escherichia coli Essential Proteins DapB and Dxr Results in Oxidative Stress, which Contributes to Lethality through Incomplete Base Excision Repair. mBio 2021; 13:e0375621. [PMID: 35130721 PMCID: PMC8822343 DOI: 10.1128/mbio.03756-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Various lethal stresses, including bactericidal antibiotics, can trigger the production of reactive oxygen species (ROS) that contribute to killing. Incomplete base excision repair (BER) of oxidized nucleotides, especially 8-oxo-dG, has been identified as a major component of ROS-induced lethality. However, the relative contributions of this pathway to death vary widely between stresses, due in part to poorly understood complex differences in the physiological changes caused by these stresses. To identify new lethal stresses that kill cells through this pathway, we screened an essential protein degradation library and found that depletion of either DapB or Dxr leads to cell death through incomplete BER; the contribution of this pathway to overall cell death is greater for DapB than for Dxr. Depletion of either protein generates oxidative stress, which increases incorporation of 8-oxo-dG into the genome. This oxidative stress is causally related to cell death, as plating on an antioxidant provided a protective effect. Moreover, incomplete BER was central to this cell death, as mutants lacking the key BER DNA glycosylases MutM and MutY were less susceptible, while overexpression of the nucleotide sanitizer MutT, which degrades 8-oxo-dGTP to prevent its incorporation, was protective. RNA sequencing of cells depleted of these proteins revealed widely different transcriptional responses to these stresses. Our discovery that oxidative stress-induced incomplete BER is highly dependent on the exact physiological changes that the cell experiences helps explain the past confusion that arose concerning the role of ROS in antibiotic lethality. IMPORTANCE Bacterial cell death is a poorly understood process. The generation of reactive oxygen species (ROS) is an apparently common response to challenges by a wide variety of lethal stresses, including bactericidal antibiotics. Incomplete BER of nucleotides damaged by these ROS, especially 8-oxo-dG, is a significant contributing factor to this lethality, but the levels of its contribution vary widely between different lethal stresses. A better understanding of the conditions that cause cells to die because of incomplete BER may lead to improved strategies for targeting this mode of death as an adjunct to antimicrobial therapy.
Collapse
|
19
|
Piselli C, Benz R. Fosmidomycin transport through the phosphate-specific porins OprO and OprP of Pseudomonas aeruginosa. Mol Microbiol 2021; 116:97-108. [PMID: 33561903 DOI: 10.1111/mmi.14693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023]
Abstract
The Gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen, responsible for many hospital-acquired infections. The bacterium is quite resistant toward many antibiotics, in particular because of the fine-tuned permeability of its outer membrane (OM). General diffusion outer membrane pores are quite rare in this organism. Instead, its OM contains many substrate-specific porins. Their expression is varying according to growth conditions and virulence. Phosphate limitations, as well as pathogenicity factors, result in the induction of the two mono- and polyphosphate-specific porins, OprP and OprO, respectively, together with an inner membrane uptake mechanism and a periplasmic binding protein. These outer membrane channels could serve as outer membrane pathways for the uptake of phosphonates. Among them are not only herbicides, but also potent antibiotics, such as fosfomycin and fosmidomycin. In this study, we investigated the interaction between OprP and OprO and fosmidomycin in detail. We could demonstrate that fosmidomycin is able to bind to the phosphate-specific binding site inside the two porins. The inhibition of chloride conductance of OprP and OprO by fosmidomycin is considerably less than that of phosphate or diphosphate, but it can be measured in titration experiments of chloride conductance and also in single-channel experiments. The results suggest that fosmidomycin transport across the OM of P. aeruginosa occurs through OprP and OprO. Our data with the ones already known in the literature show that phosphonic acid-containing antibiotics are in general good candidates to treat the infections of P. aeruginosa at the very beginning through a favorable OM transport system.
Collapse
Affiliation(s)
- Claudio Piselli
- Department of Life Sciences and Chemistry, Focus Health, Jacobs University Bremen, Bremen, Germany
| | - Roland Benz
- Rudolf-Virchow-Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
20
|
Non-hydroxamate inhibitors of 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR): A critical review and future perspective. Eur J Med Chem 2020; 213:113055. [PMID: 33303239 DOI: 10.1016/j.ejmech.2020.113055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) catalyzes the second step of the non-mevalonate (or MEP) pathway that functions in several organisms and plants for the synthesis of isoprenoids. DXR is essential for the survival of multiple pathogenic bacteria/parasites, including those that cause tuberculosis and malaria in humans. DXR function is inhibited by fosmidomycin (1), a natural product, which forms a chelate with the active site divalent metal (Mg2+/Mn2+) through its hydroxamate metal-binding group (MBG). Most of the potent DXR inhibitors are structurally similar to 1 and retain hydroxamate despite the unfavourable pharmacokinetic and toxicity profile of the latter. We provide our perspective on the lack of non-hydroxamate DXR inhibitors. We also highlight the fundamental flaws in the design of MBG in these molecules, primarily responsible for their failure to inhibit DXR. We also suggest that for designing next-generation non-hydroxamate DXR inhibitors, approaches followed for other metalloenzymes targets may be exploited.
Collapse
|
21
|
Mikati MO, Miller JJ, Osbourn DM, Barekatain Y, Ghebremichael N, Shah IT, Burnham CAD, Heidel KM, Yan VC, Muller FL, Dowd CS, Edwards RL, Odom John AR. Antimicrobial Prodrug Activation by the Staphylococcal Glyoxalase GloB. ACS Infect Dis 2020; 6:3064-3075. [PMID: 33118347 PMCID: PMC8543975 DOI: 10.1021/acsinfecdis.0c00582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
With the rising prevalence of multidrug resistance, there is an urgent need to develop novel antibiotics. Many putative antibiotics demonstrate promising in vitro potency but fail in vivo due to poor drug-like qualities (e.g., serum half-life, oral absorption, solubility, and toxicity). These drug-like properties can be modified through the addition of chemical protecting groups, creating "prodrugs" that are activated prior to target inhibition. Lipophilic prodrugging techniques, including the attachment of a pivaloyloxymethyl group, have garnered attention for their ability to increase cellular permeability by masking charged residues and the relative ease of the chemical prodrugging process. Unfortunately, pivaloyloxymethyl prodrugs are rapidly activated by human sera, rendering any membrane permeability qualities absent during clinical treatment. Identification of the bacterial prodrug activation pathway(s) will allow for the development of host-stable and microbe-targeted prodrug therapies. Here, we use two zoonotic staphylococcal species, Staphylococcus schleiferi and S. pseudintermedius, to establish the mechanism of carboxy ester prodrug activation. Using a forward genetic screen, we identify a conserved locus in both species encoding the enzyme hydroxyacylglutathione hydrolase (GloB), whose loss-of-function confers resistance to carboxy ester prodrugs. We enzymatically characterize GloB and demonstrate that it is a functional glyoxalase II enzyme, which has the capacity to activate carboxy ester prodrugs. As GloB homologues are both widespread and diverse in sequence, our findings suggest that GloB may be a useful mechanism for developing species- or genus-level prodrug targeting strategies.
Collapse
Affiliation(s)
- Marwa O Mikati
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Justin J Miller
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Damon M Osbourn
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Yasaman Barekatain
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Naomi Ghebremichael
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Ishaan T Shah
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Carey-Ann D Burnham
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Kenneth M Heidel
- Department of Chemistry, The George Washington University, Washington, DC 20052, United States
| | - Victoria C Yan
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Florian L Muller
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Cynthia S Dowd
- Department of Chemistry, The George Washington University, Washington, DC 20052, United States
| | - Rachel L Edwards
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Audrey R Odom John
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
22
|
Edwards RL, Heueck I, Lee SG, Shah IT, Miller JJ, Jezewski AJ, Mikati MO, Wang X, Brothers RC, Heidel KM, Osbourn DM, Burnham CAD, Alvarez S, Fritz SA, Dowd CS, Jez JM, Odom John AR. Potent, specific MEPicides for treatment of zoonotic staphylococci. PLoS Pathog 2020; 16:e1007806. [PMID: 32497104 PMCID: PMC7297381 DOI: 10.1371/journal.ppat.1007806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/16/2020] [Accepted: 04/11/2020] [Indexed: 12/20/2022] Open
Abstract
Coagulase-positive staphylococci, which frequently colonize the mucosal surfaces of animals, also cause a spectrum of opportunistic infections including skin and soft tissue infections, urinary tract infections, pneumonia, and bacteremia. However, recent advances in bacterial identification have revealed that these common veterinary pathogens are in fact zoonoses that cause serious infections in human patients. The global spread of multidrug-resistant zoonotic staphylococci, in particular the emergence of methicillin-resistant organisms, is now a serious threat to both animal and human welfare. Accordingly, new therapeutic targets that can be exploited to combat staphylococcal infections are urgently needed. Enzymes of the methylerythritol phosphate pathway (MEP) of isoprenoid biosynthesis represent potential targets for treating zoonotic staphylococci. Here we demonstrate that fosmidomycin (FSM) inhibits the first step of the isoprenoid biosynthetic pathway catalyzed by deoxyxylulose phosphate reductoisomerase (DXR) in staphylococci. In addition, we have both enzymatically and structurally determined the mechanism by which FSM elicits its effect. Using a forward genetic screen, the glycerol-3-phosphate transporter GlpT that facilitates FSM uptake was identified in two zoonotic staphylococci, Staphylococcus schleiferi and Staphylococcus pseudintermedius. A series of lipophilic ester prodrugs (termed MEPicides) structurally related to FSM were synthesized, and data indicate that the presence of the prodrug moiety not only substantially increased potency of the inhibitors against staphylococci but also bypassed the need for GlpT-mediated cellular transport. Collectively, our data indicate that the prodrug MEPicides selectively and robustly inhibit DXR in zoonotic staphylococci, and further, that DXR represents a promising, druggable target for future development.
Collapse
Affiliation(s)
- Rachel L. Edwards
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Isabel Heueck
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Soon Goo Lee
- University of North Carolina-Wilmington, Wilmington, North Carolina, United States of America
| | - Ishaan T. Shah
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Justin J. Miller
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew J. Jezewski
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Marwa O. Mikati
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xu Wang
- Department of Chemistry, George Washington University, Washington, DC, United States of America
| | - Robert C. Brothers
- Department of Chemistry, George Washington University, Washington, DC, United States of America
| | - Kenneth M. Heidel
- Department of Chemistry, George Washington University, Washington, DC, United States of America
| | - Damon M. Osbourn
- Department of Chemistry, Saint Louis University, St. Louis, Missouri, United States of America
| | - Carey-Ann D. Burnham
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sophie Alvarez
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Stephanie A. Fritz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cynthia S. Dowd
- Department of Chemistry, George Washington University, Washington, DC, United States of America
| | - Joseph M. Jez
- Department of Biology, Washington University, St. Louis, Missouri, United States of America
| | - Audrey R. Odom John
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
23
|
Pines G, Fankhauser RG, Eckert CA. Predicting Drug Resistance Using Deep Mutational Scanning. Molecules 2020; 25:E2265. [PMID: 32403408 PMCID: PMC7248951 DOI: 10.3390/molecules25092265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Drug resistance is a major healthcare challenge, resulting in a continuous need to develop new inhibitors. The development of these inhibitors requires an understanding of the mechanisms of resistance for a critical mass of occurrences. Recent genome editing technologies based on high-throughput DNA synthesis and sequencing may help to predict mutations resulting in resistance by testing large mutagenesis libraries. Here we describe the rationale of this approach, with examples and relevance to drug development and resistance in malaria.
Collapse
Affiliation(s)
- Gur Pines
- Department of Entomology, Agricultural Research Organization, Volcani Center, P.O.B 15159, Rishon LeZion 7505101, Israel
| | - Reilly G. Fankhauser
- Department of Dermatology, Oregon Health & Science University, Baird Hall 3225 SW Pavilion Loop, Portland, OR 97239, USA;
| | - Carrie A. Eckert
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, CO 80309, USA
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, CO 80401, USA
| |
Collapse
|
24
|
Transporters of glucose and other carbohydrates in bacteria. Pflugers Arch 2020; 472:1129-1153. [PMID: 32372286 DOI: 10.1007/s00424-020-02379-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/18/2022]
Abstract
Glucose arguably is the most important energy carrier, carbon source for metabolites and building block for biopolymers in all kingdoms of life. The proper function of animal organs and tissues depends on the continuous supply of glucose from the bloodstream. Most animals can resorb only a small number of monosaccharides, mostly glucose, galactose and fructose, while all other sugars oligosaccharides and dietary fibers are degraded and metabolized by the microbiota of the lower intestine. Bacteria, in contrast, are omnivorous. They can import and metabolize structurally different sugars and, as a consortium of different species, utilize almost any sugar, sugar derivative and oligosaccharide occurring in nature. Bacteria have membrane transport systems for the uptake of sugars against steep concentration gradients energized by ATP, the proton motive force and the high energy glycolytic intermediate phosphoenolpyruvate (PEP). Different uptake mechanisms and the broad range of overlapping substrate specificities allow bacteria to quickly adapt to and colonize changing environments. Here, we review the structures and mechanisms of bacterial representatives of (i) ATP-dependent cassette (ABC) transporters, (ii) major facilitator (MFS) superfamily proton symporters, (iii) sodium solute symporters (SSS) and (iv) enzyme II integral membrane subunits of the bacterial PEP-dependent phosphotransferase system (PTS). We give a short overview on the distribution of transporter genes and their phylogenetic relationship in different bacterial species. Some sugar transporters are hijacked for import of bacteriophage DNA and antibacterial toxins (bacteriocins) and they facilitate the penetration of polar antibiotics. Finally, we describe how the expression and activity of certain sugar transporters are controlled in response to the availability of sugars and how the presence and uptake of sugars may affect pathogenicity and host-microbiota interactions.
Collapse
|
25
|
Ball HS, Girma M, Zainab M, Riley H, Behrendt CT, Lienau C, Konzuch S, Avelar LAA, Lungerich B, Soojhawon I, Noble SM, Kurz T, Couch RD. Inhibition of the Yersinia pestis Methylerythritol Phosphate Pathway of Isoprenoid Biosynthesis by α-Phenyl-Substituted Reverse Fosmidomycin Analogues. ACS OMEGA 2020; 5:5170-5175. [PMID: 32201804 PMCID: PMC7081406 DOI: 10.1021/acsomega.9b04171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Fosmidomycin inhibits IspC (1-deoxy-d-xylulose 5-phosphate reductoisomerase), the first committed enzyme in the methylerythritol phosphate (MEP) pathway of isoprenoid biosynthesis. The MEP pathway of isoprenoid biosynthesis is essential to the causative agent of the plague, Yersinia pestis, and is entirely distinct from the corresponding mammalian pathway. To further drug development, we established structure-activity relationships of fosmidomycin analogues by assessing a suite of 17 α-phenyl-substituted reverse derivatives of fosmidomycin against Y. pestis IspC. Several of these compounds showed increased potency over fosmidomycin with IC50 values in the nanomolar range. Additionally, we performed antimicrobial susceptibility testing with Y. pestis A1122 (YpA1122). The bacteria were susceptible to several compounds with minimal inhibitory concentration (MIC) values ranging from 128 to 512 μg/mL; a correlation between the IC50 and MIC values was observed.
Collapse
Affiliation(s)
- Haley S. Ball
- Department
of Chemistry and Biochemistry, George Mason
University, Manassas, Virginia 20110, United
States
- Wound
Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Misgina Girma
- Department
of Chemistry and Biochemistry, George Mason
University, Manassas, Virginia 20110, United
States
| | - Mosufa Zainab
- Department
of Chemistry and Biochemistry, George Mason
University, Manassas, Virginia 20110, United
States
| | - Honoria Riley
- Department
of Chemistry and Biochemistry, George Mason
University, Manassas, Virginia 20110, United
States
| | - Christoph T. Behrendt
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Claudia Lienau
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sarah Konzuch
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Leandro A. A. Avelar
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Beate Lungerich
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Iswarduth Soojhawon
- Wound
Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Schroeder M. Noble
- Wound
Infections Department, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Thomas Kurz
- Institute
of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Robin D. Couch
- Department
of Chemistry and Biochemistry, George Mason
University, Manassas, Virginia 20110, United
States
| |
Collapse
|
26
|
Cao Y, Peng Q, Li S, Deng Z, Gao J. The intriguing biology and chemistry of fosfomycin: the only marketed phosphonate antibiotic. RSC Adv 2019; 9:42204-42218. [PMID: 35548698 PMCID: PMC9088020 DOI: 10.1039/c9ra08299a] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/27/2019] [Indexed: 11/29/2022] Open
Abstract
Recently infectious diseases caused by the increased emergence and rapid spread of drug-resistant bacterial isolates have been one of the main threats to global public health because of a marked surge in both morbidity and mortality. The only phosphonate antibiotic in the clinic, fosfomycin, is a small broad-spectrum molecule that effectively inhibits the initial step in peptidoglycan biosynthesis by blocking the enzyme, MurA in both Gram-positive and Gram-negative bacteria. As fosfomycin has a novel mechanism of action, low toxicity, a broad spectrum of antibacterial activity, excellent pharmacodynamic/pharmacokinetic properties, and good bioavailability, it has been approved for clinical use in the treatment of urinary tract bacterial infections in many countries for several decades. Furthermore, its potential use for difficult-to-treat bacterial infections has become promising, and fosfomycin has become an ideal candidate for the effective treatment of bacterial infections caused by multidrug-resistant isolates, especially in combination with other therapeutic drugs. Here we aim to present an overview of the biology and chemistry of fosfomycin including isolation and characterization, pharmacology, biosynthesis and chemical synthesis since its discovery in order to not only help scientists reassess the role of this exciting drug in fighting antibiotic resistance but also build the stage for discovering more novel phosphonate antibiotics in the future.
Collapse
Affiliation(s)
- Yingying Cao
- College of Life Sciences, Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Qingyao Peng
- College of Life Sciences, Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Shanni Li
- College of Life Sciences, Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Laboratory on Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Jiangtao Gao
- College of Life Sciences, Fujian Agriculture and Forestry University Fuzhou 350002 China
| |
Collapse
|
27
|
Mancini G, Bouda M, Gamrat JM, Tomsho JW. Synthesis and Antimicrobial Evaluation of γ-Borono Phosphonate Compounds in Escherichia coli and Mycobacterium smegmatis. ACS OMEGA 2019; 4:14551-14559. [PMID: 31528809 PMCID: PMC6740193 DOI: 10.1021/acsomega.9b01774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
Drug resistance in bacteria is a serious threat, and drugs with novel modes of action are constantly needed. Fosmidomycin is a naturally occurring antibiotic that inhibits the nonmevalonate pathway via inhibition of the enzyme 1-deoxylulose-5-phosphate reductoisomerase (DXR). This work is the first report in which a boronic acid is evaluated as an isostere of the retrohydroxamate moiety of fosmidomycin. We report the novel synthesis of a γ-borono phosphonate analog of fosmidomycin and its corresponding prodrugs. We evaluate the inhibition of DXR and the antimicrobial activity of γ-borono phosphonate compounds against Escherichia coli wild type, E. coli Δglycerol-3-phosphate transporter, and Mycobacterium smegmatis. Despite its structural similarities, the γ-borono phosphonate compound shows antimicrobial activity against E. coli with a mechanism of action that is different from fosmidomycin. This was proven with an underutilized method for studying in vitro inhibition of the MEP pathway in E. coli via isopentenyl pyrophosphate chemical rescue. These results indicate that these compounds may serve as a promising scaffold for developing a new class of antimicrobial agents.
Collapse
|
28
|
Courtens C, Risseeuw M, Caljon G, Maes L, Cos P, Martin A, Van Calenbergh S. Double prodrugs of a fosmidomycin surrogate as antimalarial and antitubercular agents. Bioorg Med Chem Lett 2019; 29:1232-1235. [PMID: 30879839 DOI: 10.1016/j.bmcl.2019.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/10/2019] [Accepted: 03/05/2019] [Indexed: 12/31/2022]
Abstract
A series of eleven double prodrug derivatives of a fosmidomycin surrogate were synthesized and investigated for their ability to inhibit in vitro growth of P. falciparum and M. tuberculosis. A pivaloyloxymethyl (POM) phosphonate prodrug modification was combined with various prodrug derivatisations of the hydroxamate moiety. The majority of compounds showed activity comparable with or inferior to fosmidomycin against P. falciparum. N-benzyl substituted carbamate prodrug 6f was the most active antimalarial analog with an IC50 value of 0.64 µM. Contrary to fosmidomycin and parent POM-prodrug 5, 2-nitrofuran and 2-nitrothiophene prodrugs 6i and 6j displayed promising antitubercular activities.
Collapse
Affiliation(s)
- Charlotte Courtens
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Martijn Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Guy Caljon
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Anandi Martin
- Medical Microbiology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Avenue Hippocrate 55, B-1200 Woluwe-Saint-Lambert, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium.
| |
Collapse
|
29
|
Phosphonodiamidate prodrugs of N-alkoxy analogs of a fosmidomycin surrogate as antimalarial and antitubercular agents. Bioorg Med Chem Lett 2019; 29:1051-1053. [PMID: 30857749 DOI: 10.1016/j.bmcl.2019.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 11/23/2022]
Abstract
A series of N-alkoxy analogs of a l-leucine ethyl ester phosphonodiamidate prodrug of a fosmidomycin surrogate were synthesized and investigated for their ability to inhibit in vitro growth of P. falciparum and M. tuberculosis. These compounds originate by merging a previously reported successful phosphonate derivatisation with favorable modifications of the hydroxamate moiety. None of the synthesized compounds showed enhanced activity against either P. falciparum or M. tuberculosis in comparison with the parent free hydroxamate analog.
Collapse
|
30
|
Pines G, Oh EJ, Bassalo MC, Choudhury A, Garst AD, Fankhauser RG, Eckert CA, Gill RT. Genomic Deoxyxylulose Phosphate Reductoisomerase (DXR) Mutations Conferring Resistance to the Antimalarial Drug Fosmidomycin in E. coli. ACS Synth Biol 2018; 7:2824-2832. [PMID: 30462485 DOI: 10.1021/acssynbio.8b00219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sequence to activity mapping technologies are rapidly developing, enabling the generation and isolation of mutations conferring novel phenotypes. Here we used the CRISPR enabled trackable genome engineering (CREATE) technology to investigate the inhibition of the essential ispC gene in its native genomic context in Escherichia coli. We created a full saturation library of 33 sites proximal to the ligand binding pocket and challenged this library with the antimalarial drug fosmidomycin, which targets the ispC gene product, DXR. This selection is especially challenging since it is relatively weak in E. coli, with multiple naturally occurring pathways for resistance. We identified several previously unreported mutations that confer fosmidomycin resistance, in highly conserved sites that also exist in pathogens including the malaria-inducing Plasmodium falciparum. This approach may have implications for the isolation of resistance-conferring mutations and may affect the design of future generations of fosmidomycin-based drugs.
Collapse
Affiliation(s)
- Gur Pines
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, Boulder, Colorado 80309, United States
| | - Eun Joong Oh
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, Boulder, Colorado 80309, United States
| | - Marcelo C. Bassalo
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, 347 UCB, Boulder, Colorado 80309, United States
| | - Alaksh Choudhury
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, Boulder, Colorado 80309, United States
| | - Andrew D. Garst
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
| | - Reilly G. Fankhauser
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
| | - Carrie A. Eckert
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
- Biosciences Center, National Renewable Energy Laboratory, 15013 Denver West Parkway, Golden, Colorado 80401, United States
| | - Ryan T. Gill
- Renewable and Sustainable Energy Institute, University of Colorado Boulder, 027 UCB, Boulder, Colorado 80309, United States
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 596 UCB, Boulder, Colorado 80309, United States
| |
Collapse
|
31
|
Courtens C, Risseeuw M, Caljon G, Cos P, Van Calenbergh S. Acyloxybenzyl and Alkoxyalkyl Prodrugs of a Fosmidomycin Surrogate as Antimalarial and Antitubercular Agents. ACS Med Chem Lett 2018; 9:986-989. [PMID: 30344904 DOI: 10.1021/acsmedchemlett.8b00223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Two classes of prodrugs of a fosmidomycin surrogate were synthesized and investigated for their ability to inhibit in vitro growth of P. falciparum and M. tuberculosis. To this end, a novel efficient synthesis route was developed involving a cross metathesis reaction as a key step. Alkoxyalkyl prodrugs show decent antimalarial activities, but acyloxybenzyl prodrugs proved to be the most interesting and show enhanced antimalarial and antitubercular activity. The most active antimalarial analogues show low nanomolar IC50 values.
Collapse
Affiliation(s)
- Charlotte Courtens
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Martijn Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Guy Caljon
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| |
Collapse
|
32
|
Oany AR, Mia M, Pervin T, Hasan MN, Hirashima A. Identification of potential drug targets and inhibitor of the pathogenic bacteria Shigella flexneri 2a through the subtractive genomic approach. In Silico Pharmacol 2018; 6:11. [PMID: 30607324 DOI: 10.1007/s40203-018-0048-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/28/2018] [Indexed: 01/09/2023] Open
Abstract
Shigella flexneri 2a is one of the most pathogenic bacteria among the Shigella spp., which is responsible for dysentery and causes masses of deaths throughout the world per year. A proper identification of the potential drug targets and inhibitors is crucial for the treatment of the shigellosis due to their emerging multidrug resistance (MDR) patterns. In this study, a systematic subtractive approach was implemented for the identification of novel therapeutic targets of S. flexneri 2a (301) through genome-wide metabolic pathway analysis of the essential genes and proteins. Ligand-based virtual screening and ADMET analyses were also made for the identification of potential inhibitors as well. Initially, we found 70 essential unique proteins as novel targets. After subsequent prioritization, finally we got six unique targets as the potential therapeutic targets and their three-dimensional models were built thereafter. Aspartate-β-semialdehyde dehydrogenase (ASD), was the most potent target among them and used for docking analysis through ligand-based virtual screening. The compound 3 (PubChem CID: 11319750) suited well as the best inhibitor of the ASD through ADMET and enzyme inhibition capacity analysis. To end, we hope that our proposed therapeutic targets and its inhibitors might give some breakthrough to treat shigellosis efficiently in in vitro.
Collapse
Affiliation(s)
- Arafat Rahman Oany
- 1Department of Biotechnology and Genetic Engineering, Life Science Faculty, Mawlana Bhashani Science and Technology University, Tangail, 1902 Bangladesh
| | - Mamun Mia
- 1Department of Biotechnology and Genetic Engineering, Life Science Faculty, Mawlana Bhashani Science and Technology University, Tangail, 1902 Bangladesh
| | - Tahmina Pervin
- 2Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Md Nazmul Hasan
- 3Department of Genetic Engineering and Biotechnology, Jessore University of Science and Technology, Jessore, Bangladesh
| | - Akinori Hirashima
- 4Laboratory of Pesticide Chemistry, Division of Molecular Biosciences, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 812-8581 Japan
| |
Collapse
|
33
|
Sanders S, Bartee D, Harrison MJ, Phillips PD, Koppisch AT, Freel Meyers CL. Growth medium-dependent antimicrobial activity of early stage MEP pathway inhibitors. PLoS One 2018; 13:e0197638. [PMID: 29771999 PMCID: PMC5957436 DOI: 10.1371/journal.pone.0197638] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/04/2018] [Indexed: 01/01/2023] Open
Abstract
The in vivo microenvironment of bacterial pathogens is often characterized by nutrient limitation. Consequently, conventional rich in vitro culture conditions used widely to evaluate antibacterial agents are often poorly predictive of in vivo activity, especially for agents targeting metabolic pathways. In one such pathway, the methylerythritol phosphate (MEP) pathway, which is essential for production of isoprenoids in bacterial pathogens, relatively little is known about the influence of growth environment on antibacterial properties of inhibitors targeting enzymes in this pathway. The early steps of the MEP pathway are catalyzed by 1-deoxy-d-xylulose 5-phosphate (DXP) synthase and reductoisomerase (IspC). The in vitro antibacterial efficacy of the DXP synthase inhibitor butylacetylphosphonate (BAP) was recently reported to be strongly dependent upon growth medium, with high potency observed under nutrient limitation and exceedingly weak activity in nutrient-rich conditions. In contrast, the well-known IspC inhibitor fosmidomycin has potent antibacterial activity in nutrient-rich conditions, but to date, its efficacy had not been explored under more relevant nutrient-limited conditions. The goal of this work was to thoroughly characterize the effects of BAP and fosmidomycin on bacterial cells under varied growth conditions. In this work, we show that activities of both inhibitors, alone and in combination, are strongly dependent upon growth medium, with differences in cellular uptake contributing to variance in potency of both agents. Fosmidomycin is dissimilar to BAP in that it displays relatively weaker activity in nutrient-limited compared to nutrient-rich conditions. Interestingly, while it has been generally accepted that fosmidomycin activity depends upon expression of the GlpT transporter, our results indicate for the first time that fosmidomycin can enter cells by an alternative mechanism under nutrient limitation. Finally, we show that the potency and relationship of the BAP-fosmidomycin combination also depends upon the growth medium, revealing a striking loss of BAP-fosmidomycin synergy under nutrient limitation. This change in BAP-fosmidomycin relationship suggests a shift in the metabolic and/or regulatory networks surrounding DXP accompanying the change in growth medium, the understanding of which could significantly impact targeting strategies against this pathway. More generally, our findings emphasize the importance of considering physiologically relevant growth conditions for predicting the antibacterial potential MEP pathway inhibitors and for studies of their intracellular targets.
Collapse
Affiliation(s)
- Sara Sanders
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - David Bartee
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Mackenzie J. Harrison
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Paul D. Phillips
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Andrew T. Koppisch
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Caren L. Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
34
|
Wang X, Dowd CS. The Methylerythritol Phosphate Pathway: Promising Drug Targets in the Fight against Tuberculosis. ACS Infect Dis 2018; 4:278-290. [PMID: 29390176 DOI: 10.1021/acsinfecdis.7b00176] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a severe infectious disease in need of new chemotherapies especially for drug-resistant cases. To meet the urgent requirement of new TB drugs with novel modes of action, the TB research community has been validating numerous targets from several biosynthetic pathways. The methylerythritol phosphate (MEP) pathway is utilized by Mtb for the biosynthesis of isopentenyl pyrophosphate (IPP) and its isomer dimethylallyl pyrophosphate (DMAPP), the universal five-carbon building blocks of isoprenoids. While being a common biosynthetic pathway in pathogens, the MEP pathway is completely absent in humans. Due to its unique presence in pathogens as well as the essentiality of the MEP pathway in Mtb, the enzymes in this pathway are promising targets for the development of new drugs against tuberculosis. In this Review, we discuss three enzymes in the MEP pathway: 1-deoxy-d-xylulose-5-phosphate synthase (DXS), 1-deoxy-d-xylulose-5-phosphate reductoisomerase (IspC/DXR), and 2 C-methyl-d-erythritol 2,4-cyclodiphosphate synthase (IspF), which appear to be the most promising antitubercular drug targets. Structural and mechanistic features of these enzymes are reviewed, as well as selected inhibitors that show promise as antitubercular agents.
Collapse
Affiliation(s)
- Xu Wang
- Department of Chemistry, George Washington University, 800 22nd Street NW, Washington, D.C. 20052, United States
| | - Cynthia S. Dowd
- Department of Chemistry, George Washington University, 800 22nd Street NW, Washington, D.C. 20052, United States
| |
Collapse
|
35
|
Gayán E, Cambré A, Michiels CW, Aertsen A. RpoS-independent evolution reveals the importance of attenuated cAMP/CRP regulation in high hydrostatic pressure resistance acquisition in E. coli. Sci Rep 2017; 7:8600. [PMID: 28819154 PMCID: PMC5561100 DOI: 10.1038/s41598-017-08958-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/20/2017] [Indexed: 11/09/2022] Open
Abstract
High hydrostatic pressure (HHP) processing is an attractive non-thermal alternative to food pasteurization. Nevertheless, the large inter- and intra-species variations in HHP resistance among foodborne pathogens and the ease by which they can acquire extreme resistance are an issue of increasing concern. Since RpoS activity has been considered as a central determinant in the HHP resistance of E. coli and its pathovars, this study probed for the potential of an E. coli MG1655 ΔrpoS mutant to acquire HHP resistance by directed evolution. Despite the higher initial HHP sensitivity of the ΔrpoS mutant compared to the wild-type strain, evolved lineages of the former readily managed to restore or even succeed wild-type levels of resistance. A number of these ΔrpoS derivatives were affected in cAMP/CRP regulation, and this could be causally related to their HHP resistance. Subsequent inspection revealed that some of previously isolated HHP-resistant mutants derived from the wild-type strain also incurred a causal decrease in cAMP/CRP regulation. cAMP/CRP attenuated HHP-resistant mutants also exhibited higher resistance to fosfomycin, a preferred treatment for STEC infections. As such, this study reveals attenuation of cAMP/CRP regulation as a relevant and RpoS-independent evolutionary route towards HHP resistance in E. coli that coincides with fosfomycin resistance.
Collapse
Affiliation(s)
- Elisa Gayán
- KU Leuven, Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S), Faculty of Bioscience Engineering, Leuven, Belgium
| | - Alexander Cambré
- KU Leuven, Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S), Faculty of Bioscience Engineering, Leuven, Belgium
| | - Chris W Michiels
- KU Leuven, Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S), Faculty of Bioscience Engineering, Leuven, Belgium
| | - Abram Aertsen
- KU Leuven, Laboratory of Food Microbiology, Department of Microbial and Molecular Systems (M2S), Faculty of Bioscience Engineering, Leuven, Belgium.
| |
Collapse
|
36
|
|
37
|
Sanders S, Vierling RJ, Bartee D, DeColli AA, Harrison MJ, Aklinski JL, Koppisch AT, Freel Meyers CL. Challenges and Hallmarks of Establishing Alkylacetylphosphonates as Probes of Bacterial 1-Deoxy-d-xylulose 5-Phosphate Synthase. ACS Infect Dis 2017. [PMID: 28636325 DOI: 10.1021/acsinfecdis.6b00168] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
1-Deoxy-d-xylulose 5-phosphate (DXP) synthase catalyzes the thiamin diphosphate (ThDP)-dependent formation of DXP from pyruvate and d-glyceraldehyde 3-phosphate. DXP is at a metabolic branch point in bacteria, feeding into the methylerythritol phosphate pathway to indispensable isoprenoids and acting as a precursor for biosynthesis of essential cofactors in central metabolism, pyridoxal phosphate and ThDP, the latter of which is also required for DXP synthase catalysis. DXP synthase follows a unique random sequential mechanism and possesses an unusually large active site. These features have guided the design of sterically demanding alkylacetylphosphonates (alkylAPs) toward the development of selective DXP synthase inhibitors. alkylAPs studied here display selective, low μM inhibitory activity against DXP synthase. They are weak inhibitors of bacterial growth in standard nutrient rich conditions. However, bacteria are significantly sensitized to most alkylAPs in defined minimal growth medium, with minimal inhibitory concentrations (MICs) ranging from low μM to low mM and influenced by alkyl-chain length. The longest analog (C8) displays the weakest antimicrobial activity and is a substrate for efflux via AcrAB-TolC. The dependence of inhibitor potency on growth environment emphasizes the need for antimicrobial screening conditions that are relevant to the in vivo microbial microenvironment during infection. DXP synthase expression and thiamin supplementation studies offer support for DXP synthase as an intracellular target for some alkylAPs and reveal both the challenges and intriguing aspects of these approaches to study target engagement.
Collapse
Affiliation(s)
- Sara Sanders
- Department of Pharmacology
and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Ryan J. Vierling
- Department of Pharmacology
and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - David Bartee
- Department of Pharmacology
and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Alicia A. DeColli
- Department of Pharmacology
and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Mackenzie J. Harrison
- Department
of Chemistry, Northern Arizona University, Flagstaff, Arizona 86011, United States
| | - Joseph L. Aklinski
- Department
of Chemistry, Northern Arizona University, Flagstaff, Arizona 86011, United States
| | - Andrew T. Koppisch
- Department
of Chemistry, Northern Arizona University, Flagstaff, Arizona 86011, United States
| | - Caren L. Freel Meyers
- Department of Pharmacology
and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
38
|
Ohkoshi Y, Sato T, Suzuki Y, Yamamoto S, Shiraishi T, Ogasawara N, Yokota SI. Mechanism of Reduced Susceptibility to Fosfomycin in Escherichia coli Clinical Isolates. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5470241. [PMID: 28197413 PMCID: PMC5288514 DOI: 10.1155/2017/5470241] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/22/2016] [Accepted: 12/27/2016] [Indexed: 11/29/2022]
Abstract
In recent years, multidrug resistance of Escherichia coli has become a serious problem. However, resistance to fosfomycin (FOM) has been low. We screened E. coli clinical isolates with reduced susceptibility to FOM and characterized molecular mechanisms of resistance and reduced susceptibility of these strains. Ten strains showing reduced FOM susceptibility (MIC ≥ 8 μg/mL) in 211 clinical isolates were found and examined. Acquisition of genes encoding FOM-modifying enzyme genes (fos genes) and mutations in murA that underlie high resistance to FOM were not observed. We examined ability of FOM incorporation via glucose-6-phosphate (G6P) transporter and sn-glycerol-3-phosphate transporter. In ten strains, nine showed lack of growth on M9 minimum salt agar supplemented with G6P. Eight of the ten strains showed fluctuated induction by G6P of uhpT that encodes G6P transporter expression. Nucleotide sequences of the uhpT, uhpA, glpT, ptsI, and cyaA shared several deletions and amino acid mutations in the nine strains with lack of growth on G6P-supplemented M9 agar. In conclusion, reduction of uhpT function is largely responsible for the reduced sensitivity to FOM in clinical isolates that have not acquired FOM-modifying genes or mutations in murA. However, there are a few strains whose mechanisms of reduced susceptibility to FOM are still unclear.
Collapse
Affiliation(s)
- Yasuo Ohkoshi
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
- Department of Clinical Laboratory, NTT East Sapporo Hospital, Chuo-ku, Sapporo 060-0061, Japan
| | - Toyotaka Sato
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Yuuki Suzuki
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Soh Yamamoto
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Tsukasa Shiraishi
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Noriko Ogasawara
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Shin-ichi Yokota
- Department of Microbiology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
39
|
Munier M, Tritsch D, Krebs F, Esque J, Hemmerlin A, Rohmer M, Stote RH, Grosdemange-Billiard C. Synthesis and biological evaluation of phosphate isosters of fosmidomycin and analogs as inhibitors of Escherichia coli and Mycobacterium smegmatis 1-deoxyxylulose 5-phosphate reductoisomerases. Bioorg Med Chem 2017; 25:684-689. [DOI: 10.1016/j.bmc.2016.11.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 11/26/2022]
|
40
|
Chekan JR, Cogan DP, Nair SK. Molecular Basis for Resistance Against Phosphonate Antibiotics and Herbicides. MEDCHEMCOMM 2016; 7:28-36. [PMID: 26811741 PMCID: PMC4723106 DOI: 10.1039/c5md00351b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Research in recent years have illuminated data on the mechanisms and targets of phosphonic acid antibiotics and herbicides, including fosfomycin, glyphosate, fosmidomycin and FR900098. Here we review the current state of knowledge of the structural and biochemical characterization of resistance mechanisms against these bioactive natural products. Advances in the understanding of these resistance determinants have spurred knowledge-based campaigns aimed towards the design of derivatives that retain biological activity but are less prone to tolerance.
Collapse
Affiliation(s)
- Jonathan R. Chekan
- Department of Biochemistry, University of Illinois at Urbana Champaign, USA
- Center for Biophysics and Computational Biology, University of Illinois at Urbana Champaign, USA
| | - Dillon P. Cogan
- Department of Biochemistry, University of Illinois at Urbana Champaign, USA
- Center for Biophysics and Computational Biology, University of Illinois at Urbana Champaign, USA
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois at Urbana Champaign, USA
- Center for Biophysics and Computational Biology, University of Illinois at Urbana Champaign, USA
- Institute for Genomic Biology, University of Illinois at Urbana Champaign, USA
| |
Collapse
|
41
|
Armstrong CM, Meyers DJ, Imlay LS, Freel Meyers C, Odom AR. Resistance to the antimicrobial agent fosmidomycin and an FR900098 prodrug through mutations in the deoxyxylulose phosphate reductoisomerase gene (dxr). Antimicrob Agents Chemother 2015; 59:5511-9. [PMID: 26124156 PMCID: PMC4538460 DOI: 10.1128/aac.00602-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/20/2015] [Indexed: 11/20/2022] Open
Abstract
There is a pressing need for new antimicrobial therapies to combat globally important drug-resistant human pathogens, including Plasmodium falciparum malarial parasites, Mycobacterium tuberculosis, and Gram-negative bacteria, including Escherichia coli. These organisms all possess the essential methylerythritol phosphate (MEP) pathway of isoprenoid biosynthesis, which is not found in humans. The first dedicated enzyme of the MEP pathway, 1-deoxy-d-xylulose 5-phosphate reductoisomerase (Dxr), is inhibited by the phosphonic acid antibiotic fosmidomycin and its analogs, including the N-acetyl analog FR900098 and the phosphoryl analog fosfoxacin. In order to identify mutations in dxr that confer resistance to these drugs, a library of E. coli dxr mutants was screened at lethal fosmidomycin doses. The most resistant allele (with the S222T mutation) alters the fosmidomycin-binding site of Dxr. The expression of this resistant allele increases bacterial resistance to fosmidomycin and other fosmidomycin analogs by 10-fold. These observations confirm that the primary cellular target of fosmidomycin is Dxr. Furthermore, cell lines expressing Dxr-S222T will be a powerful tool to confirm the mechanisms of action of future fosmidomycin analogs.
Collapse
Affiliation(s)
- Christopher M Armstrong
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David J Meyers
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Leah S Imlay
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Audrey R Odom
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
42
|
Chofor R, Sooriyaarachchi S, Risseeuw MDP, Bergfors T, Pouyez J, Johny C, Haymond A, Everaert A, Dowd CS, Maes L, Coenye T, Alex A, Couch RD, Jones TA, Wouters J, Mowbray SL, Van Calenbergh S. Synthesis and Bioactivity of β-Substituted Fosmidomycin Analogues Targeting 1-Deoxy-d-xylulose-5-phosphate Reductoisomerase. J Med Chem 2015; 58:2988-3001. [DOI: 10.1021/jm5014264] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- René Chofor
- Laboratory
for Medicinal Chemistry (FFW), Universiteit Gent, Ottergemsesteenweg
460, B-9000 Gent, Belgium
| | - Sanjeewani Sooriyaarachchi
- Department
of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Biomedical
Center, Box 596, SE-751 24 Uppsala, Sweden
| | - Martijn D. P. Risseeuw
- Laboratory
for Medicinal Chemistry (FFW), Universiteit Gent, Ottergemsesteenweg
460, B-9000 Gent, Belgium
| | - Terese Bergfors
- Department
of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Biomedical
Center, Box 596, SE-751 24 Uppsala, Sweden
| | - Jenny Pouyez
- Department
of Chemistry, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Chinchu Johny
- Department
of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20110, United States
| | - Amanda Haymond
- Department
of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20110, United States
| | - Annelien Everaert
- Laboratory
of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Cynthia S. Dowd
- Department
of Chemistry, George Washington University, Washington, D.C. 20052, United States
| | - Louis Maes
- Laboratory
for Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical,
Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein
1, B-2610 Antwerp, Belgium
| | - Tom Coenye
- Laboratory
of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Alexander Alex
- Evenor Consulting Ltd., The
New Barn, Mill Lane, Eastry, Kent CT13 0JW, United Kingdom
| | - Robin D. Couch
- Department
of Chemistry and Biochemistry, George Mason University, Manassas, Virginia 20110, United States
| | - T. Alwyn Jones
- Department
of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Biomedical
Center, Box 596, SE-751 24 Uppsala, Sweden
| | - Johan Wouters
- Department
of Chemistry, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Sherry L. Mowbray
- Department
of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Biomedical
Center, Box 596, SE-751 24 Uppsala, Sweden
| | - Serge Van Calenbergh
- Laboratory
for Medicinal Chemistry (FFW), Universiteit Gent, Ottergemsesteenweg
460, B-9000 Gent, Belgium
| |
Collapse
|
43
|
Haymond A, Johny C, Dowdy T, Schweibenz B, Villarroel K, Young R, Mantooth CJ, Patel T, Bases J, Jose GS, Jackson ER, Dowd CS, Couch RD. Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase). PLoS One 2014; 9:e106243. [PMID: 25171339 PMCID: PMC4149570 DOI: 10.1371/journal.pone.0106243] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022] Open
Abstract
The methylerythritol phosphate (MEP) pathway found in many bacteria governs the synthesis of isoprenoids, which are crucial lipid precursors for vital cell components such as ubiquinone. Because mammals synthesize isoprenoids via an alternate pathway, the bacterial MEP pathway is an attractive target for novel antibiotic development, necessitated by emerging antibiotic resistance as well as biodefense concerns. The first committed step in the MEP pathway is the reduction and isomerization of 1-deoxy-D-xylulose-5-phosphate (DXP) to methylerythritol phosphate (MEP), catalyzed by MEP synthase. To facilitate drug development, we cloned, expressed, purified, and characterized MEP synthase from Yersinia pestis. Enzyme assays indicate apparent kinetic constants of KMDXP = 252 µM and KMNADPH = 13 µM, IC50 values for fosmidomycin and FR900098 of 710 nM and 231 nM respectively, and Ki values for fosmidomycin and FR900098 of 251 nM and 101 nM respectively. To ascertain if the Y. pestis MEP synthase was amenable to a high-throughput screening campaign, the Z-factor was determined (0.9) then the purified enzyme was screened against a pilot scale library containing rationally designed fosmidomycin analogs and natural product extracts. Several hit molecules were obtained, most notably a natural product allosteric affector of MEP synthase and a rationally designed bisubstrate derivative of FR900098 (able to associate with both the NADPH and DXP binding sites in MEP synthase). It is particularly noteworthy that allosteric regulation of MEP synthase has not been described previously. Thus, our discovery implicates an alternative site (and new chemical space) for rational drug development.
Collapse
Affiliation(s)
- Amanda Haymond
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Chinchu Johny
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Tyrone Dowdy
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Brandon Schweibenz
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Karen Villarroel
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Richard Young
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Clark J. Mantooth
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Trishal Patel
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Jessica Bases
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Geraldine San Jose
- Department of Chemistry, George Washington University, Washington DC, United States of America
| | - Emily R. Jackson
- Department of Chemistry, George Washington University, Washington DC, United States of America
| | - Cynthia S. Dowd
- Department of Chemistry, George Washington University, Washington DC, United States of America
| | - Robin D. Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| |
Collapse
|
44
|
Sub-inhibitory fosmidomycin exposures elicits oxidative stress in Salmonella enterica serovar Typhimurium LT2. PLoS One 2014; 9:e95271. [PMID: 24751777 PMCID: PMC3994034 DOI: 10.1371/journal.pone.0095271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 03/26/2014] [Indexed: 01/16/2023] Open
Abstract
Fosmidomycin is a time-dependent nanomolar inhibitor of methylerythritol phosphate (MEP) synthase, which is the enzyme that catalyzes the first committed step in the MEP pathway to isoprenoids. Importantly, fosmidomycin is one of only a few MEP pathway-specific inhibitors that exhibits antimicrobial activity. Most inhibitors identified to date only exhibit activity against isolated pathway enzymes. The MEP pathway is the sole route to isoprenoids in many bacteria, yet has no human homologs. The development of inhibitors of this pathway holds promise as novel antimicrobial agents. Similarly, analyses of the bacterial response toward MEP pathway inhibitors provides valuable information toward the understanding of how emergent resistance may ultimately develop to this class of antibiotics. We have examined the transcriptional response of Salmonella enterica serovar typhimurium LT2 to sub-inhibitory concentrations of fosmidomycin via cDNA microarray and RT-PCR. Within the regulated genes identified by microarray were a number of genes encoding enzymes associated with the mediation of reactive oxygen species (ROS). Regulation of a panel of genes implicated in the response of cells to oxidative stress (including genes for catalases, superoxide dismutases, and alkylhydrogen peroxide reductases) was investigated and mild upregulation in some members was observed as a function of fosmidomycin exposure over time. The extent of regulation of these genes was similar to that observed for comparable exposures to kanamycin, but differed significantly from tetracycline. Furthermore, S. typhimurium exposed to sub-inhibitory concentrations of fosmidomycin displayed an increased sensitivity to exogenous H2O2 relative to either untreated controls or kanamycin-treated cells. Our results suggest that endogenous oxidative stress is one consequence of exposures to fosmidomycin, likely through the temporal depletion of intracellular isoprenoids themselves, rather than other mechanisms that have been proposed to facilitate ROS accumulation in bacteria (e.g. cell death processes or the ability of the antibiotic to redox cycle).
Collapse
|
45
|
Chofor R, Risseeuw MDP, Pouyez J, Johny C, Wouters J, Dowd CS, Couch RD, Van Calenbergh S. Synthetic Fosmidomycin analogues with altered chelating moieties do not inhibit 1-deoxy-d-xylulose 5-phosphate Reductoisomerase or Plasmodium falciparum growth in vitro. Molecules 2014; 19:2571-87. [PMID: 24566322 PMCID: PMC6271069 DOI: 10.3390/molecules19022571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 11/17/2022] Open
Abstract
Fourteen new fosmidomycin analogues with altered metal chelating groups were prepared and evaluated for inhibition of E. coli Dxr, M. tuberculosis Dxr and the growth of P. falciparum K1 in human erythrocytes. None of the synthesized compounds showed activity against either enzyme or the Plasmodia. This study further underlines the importance of the hydroxamate functionality and illustrates that identifying effective alternative bidentate ligands for this target enzyme is challenging.
Collapse
Affiliation(s)
- René Chofor
- Laboratory for Medicinal Chemistry, Ghent University, Harelbekestraat 72, Ghent B-9000, Belgium.
| | - Martijn D P Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Harelbekestraat 72, Ghent B-9000, Belgium.
| | - Jenny Pouyez
- Department of Chemistry, University of Namur, UNamur, Rue de Bruxelles 61, Namur B-5000, Belgium.
| | - Chinchu Johny
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA.
| | - Johan Wouters
- Department of Chemistry, University of Namur, UNamur, Rue de Bruxelles 61, Namur B-5000, Belgium.
| | - Cynthia S Dowd
- Department of Chemistry, George Washington University, Washington, DC 20052, USA.
| | - Robin D Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA.
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Harelbekestraat 72, Ghent B-9000, Belgium.
| |
Collapse
|
46
|
Nikolaidis I, Favini-Stabile S, Dessen A. Resistance to antibiotics targeted to the bacterial cell wall. Protein Sci 2014; 23:243-59. [PMID: 24375653 DOI: 10.1002/pro.2414] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/21/2013] [Accepted: 12/23/2013] [Indexed: 11/10/2022]
Abstract
Peptidoglycan is the main component of the bacterial cell wall. It is a complex, three-dimensional mesh that surrounds the entire cell and is composed of strands of alternating glycan units crosslinked by short peptides. Its biosynthetic machinery has been, for the past five decades, a preferred target for the discovery of antibacterials. Synthesis of the peptidoglycan occurs sequentially within three cellular compartments (cytoplasm, membrane, and periplasm), and inhibitors of proteins that catalyze each stage have been identified, although not all are applicable for clinical use. A number of these antimicrobials, however, have been rendered inactive by resistance mechanisms. The employment of structural biology techniques has been instrumental in the understanding of such processes, as well as the development of strategies to overcome them. This review provides an overview of resistance mechanisms developed toward antibiotics that target bacterial cell wall precursors and its biosynthetic machinery. Strategies toward the development of novel inhibitors that could overcome resistance are also discussed.
Collapse
Affiliation(s)
- I Nikolaidis
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, 6 rue Jules Horowitz, 38027, Grenoble, France; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Grenoble, France; Centre National de la Recherche Scientifique (CNRS), UMR 5075, Grenoble, France; Bijvoet Center for Biomolecular Research, Department of Biochemistry of Membranes, Utrecht University, Utrecht, The Netherlands
| | | | | |
Collapse
|
47
|
Smith JM, Warrington NV, Vierling RJ, Kuhn ML, Anderson WF, Koppisch AT, Freel Meyers CL. Targeting DXP synthase in human pathogens: enzyme inhibition and antimicrobial activity of butylacetylphosphonate. J Antibiot (Tokyo) 2014; 67:77-83. [PMID: 24169798 PMCID: PMC3946878 DOI: 10.1038/ja.2013.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 11/24/2022]
Abstract
The unique methylerythritol phosphate pathway for isoprenoid biosynthesis is essential in most bacterial pathogens. The first enzyme in this pathway, 1-deoxy-D-xylulose 5-phosphate (DXP) synthase, catalyzes a distinct thiamin diphosphate (ThDP)-dependent reaction to form DXP from D-glyceraldehyde 3-phosphate (D-GAP) and pyruvate and represents a potential anti-infective drug target. We have previously demonstrated that the unnatural bisubstrate analog, butylacetylphosphonate (BAP), exhibits selective inhibition of Escherichia coli DXP synthase over mammalian ThDP-dependent enzymes. Here, we report the selective inhibition by BAP against recombinant DXP synthase homologs from Mycobacterium tuberculosis, Yersinia pestis and Salmonella enterica. We also demonstrate antimicrobial activity of BAP against both Gram-negative and Gram-positive strains (including E. coli, S. enterica and Bacillus anthracis), and several clinically isolated pathogens. Our results suggest a mechanism of action involving inhibition of DXP synthase and show that BAP acts synergistically with established antimicrobial agents, highlighting a potential strategy to combat emerging resistance in bacterial pathogens.
Collapse
Affiliation(s)
- Jessica M Smith
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole V Warrington
- Deptartment of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, USA
| | - Ryan J Vierling
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Misty L Kuhn
- Center for Structural Genomics of Infectious Diseases, Northwestern Feinberg School of Medicine, Department of Molecular Pharmacology and Biological Chemistry, Chicago, IL, USA
| | - Wayne F Anderson
- Center for Structural Genomics of Infectious Diseases, Northwestern Feinberg School of Medicine, Department of Molecular Pharmacology and Biological Chemistry, Chicago, IL, USA
| | - Andrew T Koppisch
- Deptartment of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, USA
| | - Caren L Freel Meyers
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
48
|
Boronat A, Rodríguez-Concepción M. Terpenoid biosynthesis in prokaryotes. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 148:3-18. [PMID: 25523226 DOI: 10.1007/10_2014_285] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prokaryotic organisms (archaea and eubacteria) are found in all habitats where life exists on our planet. This would not be possible without the astounding biochemical plasticity developed by such organisms. Part of the metabolic diversity of prokaryotes was transferred to eukaryotic cells when endosymbiotic prokaryotes became mitochondria and plastids but also in a large number of horizontal gene transfer episodes. A group of metabolites produced by all free-living organisms is terpenoids (also known as isoprenoids). In prokaryotes, terpenoids play an indispensable role in cell-wall and membrane biosynthesis (bactoprenol, hopanoids), electron transport (ubiquinone, menaquinone), or conversion of light into chemical energy (chlorophylls, bacteriochlorophylls, rhodopsins, carotenoids), among other processes. But despite their remarkable structural and functional diversity, they all derive from the same metabolic precursors. Here we describe the metabolic pathways producing these universal terpenoid units and provide a complete picture of the main terpenoid compounds found in prokaryotic organisms.
Collapse
Affiliation(s)
- Albert Boronat
- Centre for Research in Agricultural Genomics (CRAG) CSIC-IRTA-UAB-UB, Campus UAB Bellaterra, 08193, Barcelona, Spain
| | | |
Collapse
|
49
|
Hemmerlin A, Tritsch D, Hammann P, Rohmer M, Bach TJ. Profiling of defense responses in Escherichia coli treated with fosmidomycin. Biochimie 2013; 99:54-62. [PMID: 24262605 DOI: 10.1016/j.biochi.2013.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022]
Abstract
The mevalonate-independent isoprenoid biosynthesis pathway has been recognized as a promising target for designing new antibiotics. But pathogens treated with compounds such as fosmidomycin, a slow binding inhibitor of 1-deoxy-D-xylulose 5-phosphate reducto-isomerase, the second enzyme in this pathway, develop rapid drug resistance. In Escherichia coli, acquired resistance results mostly from inactivating the cAMP-dependent glpT transporter, thereby preventing import of the inhibitor. Such mutant strains are characterized by cross-resistance to fosfomycin, by susceptibility to efflux pump inhibitors, by disability to use glycerol 3-phosphate as a carbon source or by increased activity of the promoter controlling the expression of the glpABC regulon when grown in presence of fosmidomycin. The quite challenging task consists in conceiving new and efficient inhibitors avoiding resistance acquisition. They should be efficient in blocking the target enzyme, but should also be durably taken up by the organism. To address this issue, it is essential to characterize the mechanisms the pathogen exploits to defeat the antibiotic before resistance is acquired. Having this in mind, a 2-D Fluorescence Difference Gel Electrophoresis proteomic approach has been applied to identify defense responses in E. coli cells being shortly exposed to fosmidomycin (3 h). It seems that combined strategies are promptly induced. The major one consists in preventing toxic effects of the compound either by adapting metabolism and/or by getting rid of the molecule. The strategy adopted by the bacteria is to eliminate the drug from the cell or to increase the tolerance to oxidative stress. The design of new, but still efficient drugs, needs consideration of such rapid modulations required to adapt cell growth in contact of the inhibitor.
Collapse
Affiliation(s)
- Andréa Hemmerlin
- CNRS UPR 2357, Institut de Biologie Moléculaire des Plantes, Conventionné avec l'Université de Strasbourg, 28 rue Goethe, F-67083 Strasbourg Cedex, France.
| | - Denis Tritsch
- Université de Strasbourg/CNRS, Institut de Chimie UMR 7177, 4 rue Blaise Pascal, F-67070 Strasbourg Cedex, France
| | - Philippe Hammann
- CNRS, FRC 1589, Plateforme Protéomique Esplanade, 15 Rue René Descartes, F-67084 Strasbourg Cedex, France
| | - Michel Rohmer
- Université de Strasbourg/CNRS, Institut de Chimie UMR 7177, 4 rue Blaise Pascal, F-67070 Strasbourg Cedex, France
| | - Thomas J Bach
- CNRS UPR 2357, Institut de Biologie Moléculaire des Plantes, Conventionné avec l'Université de Strasbourg, 28 rue Goethe, F-67083 Strasbourg Cedex, France
| |
Collapse
|
50
|
Hartmann M, Hemmerlin A, Gas-Pascual E, Gerber E, Tritsch D, Rohmer M, Bach TJ. The effect of MEP pathway and other inhibitors on the intracellular localization of a plasma membrane-targeted, isoprenylable GFP reporter protein in tobacco BY-2 cells. F1000Res 2013; 2:170. [PMID: 24555083 DOI: 10.12688/f1000research.2-170.v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2013] [Indexed: 11/20/2022] Open
Abstract
We have established anin vivovisualization system for the geranylgeranylation of proteins in a stably transformed tobacco BY-2 cell line, based on the expression of a dexamethasone-inducible GFP fused to the carboxy-terminal basic domain of the rice calmodulin CaM61, which naturally bears a CaaL geranylgeranylation motif (GFP-BD-CVIL). By using pathway-specific inhibitors it was demonstrated that inhibition of the methylerythritol phosphate (MEP) pathway with known inhibitors like oxoclomazone and fosmidomycin, as well as inhibition of the protein geranylgeranyltransferase type 1 (PGGT-1), shifted the localization of the GFP-BD-CVIL protein from the membrane to the nucleus. In contrast, the inhibition of the mevalonate (MVA) pathway with mevinolin did not affect the localization. During the present work, this test system has been used to examine the effect of newly designed inhibitors of the MEP pathway and inhibitors of sterol biosynthesis such as squalestatin, terbinafine and Ro48-8071. In addition, we also studied the impact of different post-prenylation inhibitors or those suspected to affect the transport of proteins to the plasma membrane on the localization of the geranylgeranylable fusion protein GFP-BD-CVIL.
Collapse
Affiliation(s)
- Michael Hartmann
- Département Réseaux Métaboliques, Institut de Biologie Moléculaire des Plantes, CNRS UPR 2357, Université de Strasbourg, F-67083 Strasbourg, France ; Current address: Institute of Biological Chemistry, Washington State University, Pullman WA, 99164-6340, USA
| | - Andrea Hemmerlin
- Département Réseaux Métaboliques, Institut de Biologie Moléculaire des Plantes, CNRS UPR 2357, Université de Strasbourg, F-67083 Strasbourg, France
| | - Elisabet Gas-Pascual
- Département Réseaux Métaboliques, Institut de Biologie Moléculaire des Plantes, CNRS UPR 2357, Université de Strasbourg, F-67083 Strasbourg, France ; Current address: Department of Horticulture and Crop Science, Ohio State University, Wooster OH, 44691, USA
| | - Esther Gerber
- Département Réseaux Métaboliques, Institut de Biologie Moléculaire des Plantes, CNRS UPR 2357, Université de Strasbourg, F-67083 Strasbourg, France ; Current address: Deinove SA, F-34830 Clapiers, France
| | - Denis Tritsch
- UMR 7177 CNRS, Laboratoire de Chimie et Biochimie des Microorganismes, Institut de Chimie de Strasbourg, Université de Strasbourg, F-67008 Strasbourg, France
| | - Michel Rohmer
- UMR 7177 CNRS, Laboratoire de Chimie et Biochimie des Microorganismes, Institut de Chimie de Strasbourg, Université de Strasbourg, F-67008 Strasbourg, France
| | - Thomas J Bach
- Département Réseaux Métaboliques, Institut de Biologie Moléculaire des Plantes, CNRS UPR 2357, Université de Strasbourg, F-67083 Strasbourg, France
| |
Collapse
|